Alveolar macrophages develop from fetal monocytes that differentiate into long-lived cells in the first week of life via GM-CSF

Martin Guilliams, Ismé De Kleer, Sandrine Henri, Sijranke Post, Leen Vanhoutte, Sofie De Prijck, Kim Deswarte, Bernard Malissen, Hamida Hammad, Bart N Lambrecht, Martin Guilliams, Ismé De Kleer, Sandrine Henri, Sijranke Post, Leen Vanhoutte, Sofie De Prijck, Kim Deswarte, Bernard Malissen, Hamida Hammad, Bart N Lambrecht

Abstract

Tissue-resident macrophages can develop from circulating adult monocytes or from primitive yolk sac-derived macrophages. The precise ontogeny of alveolar macrophages (AMFs) is unknown. By performing BrdU labeling and parabiosis experiments in adult mice, we found that circulating monocytes contributed minimally to the steady-state AMF pool. Mature AMFs were undetectable before birth and only fully colonized the alveolar space by 3 d after birth. Before birth, F4/80(hi)CD11b(lo) primitive macrophages and Ly6C(hi)CD11b(hi) fetal monocytes sequentially colonized the developing lung around E12.5 and E16.5, respectively. The first signs of AMF differentiation appeared around the saccular stage of lung development (E18.5). Adoptive transfer identified fetal monocytes, and not primitive macrophages, as the main precursors of AMFs. Fetal monocytes transferred to the lung of neonatal mice acquired an AMF phenotype via defined developmental stages over the course of one week, and persisted for at least three months. Early AMF commitment from fetal monocytes was absent in GM-CSF-deficient mice, whereas short-term perinatal intrapulmonary GM-CSF therapy rescued AMF development for weeks, although the resulting AMFs displayed an immature phenotype. This demonstrates that tissue-resident macrophages can also develop from fetal monocytes that adopt a stable phenotype shortly after birth in response to instructive cytokines, and then self-maintain throughout life.

Figures

Figure 1.
Figure 1.
AMFs self-maintain locally with minimal contribution from circulating hematopoietic precursors. BAL (A) and total lung (B) were harvested from adult mice and stained for CD11b, F4/80, SiglecF, CD64, Ly-6C, and MHCII and evaluated for autofluorescence, FSC, and SSC profile (C). (D) CD45.1+CD45.2+ mice were lethally irradiated and reconstituted with equal amounts of CD45.1+ WT and CD45.2+CCR2−/− BM cells. Radio-resistant cells, distinguished as CD45.1+CD45.2+ cells, were gated out, and the remaining BM-derived cells of the indicated populations were analyzed for their ratio of CD45.1+ WT and CD45.2+CCR2−/− cells. (E) Summary data from five independent chimeric mice. (F) Parabiotic mice were generated by suturing together CD45.1+ WT and CD45.2+CCR2−/− mice. The percentage of cells of CD45.1+ WT donor origin was determined in B cells, neutrophils, and monocytes in the blood and AMFs in the lungs of the CD45.2+CCR2−/− parabionts. (G) Adult C57BL/6 mice received BrdU continuously for 28 d. Mice were sacrificed at regular intervals, and incorporation of BrdU in lung AMFs and monocytes was evaluated. (H) Adult C57BL/6 mice were sacrificed and Ki-67 expression (left) was determined in AMFs. The isotype staining is shown (right). Data represent at least three (A– E) or two (F– H) independent experiments involving at least six (E) and four (F and G) independent mice.
Figure 2.
Figure 2.
Alveolar MFs appear in the alveolar space during the first week of life. Lungs were harvested at various time points before and after (PND) the DOB. (A and C) Flow cytometry staining for CD45+ cells. (B) CD11b+F4/80+ myeloid cells were gated and analyzed for CD11c and SiglecF expression. (D) Paraffin lung sections stained with hematoxylin. (E) Cryosection of lungs stained with DAPI (blue) and SiglecF (red). Data in A–E represent at least two independent experiments involving at least three independent mice per time point.
Figure 3.
Figure 3.
Fetal MFs, fetal monocytes, preAMFs, and mature AMFs appear in consecutive waves during lung development. Lungs were harvested at various time points before and after (PND3, PND14) the date of birth (DOB). CD11b+F4/80+ myeloid cells were subdivided into CD11cloSigle-Flo, CD11cintSigle-Flo, and CD11chiSiglecFhi cells (A) and analyzed for Ly-6C, F4/80 (B), and CD64 expression (C). (D) Fetal monocytes (isolated at E17), fetal MFs (isolated at E17), preAMFs (isolated at the DOB), and mature AMFs (isolated from adult mice) were sorted, put in culture in vitro overnight in complete medium, and subjected to electromagnetic microscopy to assess their capacity to adhere to cell culture plastic and general morphology. (E) Percentage of fetal MFs, fetal monocytes, preAMFs, and mature AMFs among CD45+ cells in the lungs at the indicated time points. Data in A–E represent at least two independent experiments involving at least three independent mice per time point.
Figure 4.
Figure 4.
Fetal monocytes give rise to self-maintaining AMFs through a preAMF intermediate step. (A and B) CD45.1+ or CD45.2+ E17 embryos were sacrificed and fetal monocytes and fetal MFs were FACS sorted and mixed together at a 50:50 ratio (A; left, CD45.1+ fetal monocytes and CD45.2+ fetal MFs; right, CD45.1+ fetal MFs and CD45.2+ fetal monocytes) and transferred into CD45.1+CD45.2+ mice on their DOB. 7 d after transfer, CD45.1+CD45.2+ recipient mice were sacrificed and the presence of CD45.1+ or CD45.2+ donor-derived AMFs was evaluated. (B) Summary of data from four independent recipient mice. (C) CD45.1+ E17 embryos were sacrificed, and fetal monocytes were FACS sorted and transferred into CD45.2+ mice on their DOB. At the indicated time points after transfer, the expression of CD11c, CD11b, Ly-6C, SiglecF, and F4/80 was evaluated in CD45.1+ fetal monocyte–derived cells. (D) The percentage of CD45.1+ fetal monocyte–derived cells among mature AMFs at the indicated time points after transfer. Data represent at least two (A– D) independent experiments, with at least three recipient mice per time point.
Figure 5.
Figure 5.
Csf2−/− mice lack preAMFs and mature AMFs throughout life. (A) Lung epithelial cells isolated at the indicated time points before and after birth were FACS sorted, and expression of GM-CSF mRNA was measured by RT-PCR. The expression levels were normalized by comparison with expression of the HPRT housekeeping gene. (B) GM-CSF levels in lung homogenates was measured by ELISA at the indicated time points before and after birth. (C) Csf2−/− mice were sacrificed on their DOB. Lungs were homogenized and CD11b+F4/80+ myeloid cells (gated as indicated) were assessed for Ly-6C, CD64, CD11c, F4/80, and SiglecF expression. (D) GM-CSF-R (CD116) expression was evaluated on fetal MFs, fetal monocytes, preAMFs, and mature AMFs on the days mentioned using the gating strategy depicted in Fig. 3. (E) The presence of CD11c+SiglecF+ AMFs was evaluated in the BAL of WT and Csf2−/− mice at the indicated time points after birth. (F and G) The presence of F4/80hiCD11blo splenic MFs and liver MFs (Kupffer Cells) was evaluated in adult WT and Csf2−/− mice. (H and I) BAL cells from adult WT and Csf2−/− mice were subjected to a Percoll gradient to remove dead cells and protein debris. The resulting CD64+F4/80+ MFs were assessed for Ly-6C, CD11b, CD11c, and SiglecF expression (I). Data represent two (H and I) and three (A–G) independent experiments.
Figure 6.
Figure 6.
Perinatal GM-CSF treatment of Csf2−/− mice restores the generation of self-maintaining preAMFs. (A and B) Csf2−/− mice were treated 1 time (1x) on the first day after birth, 3 times (3x) on the first 3 d after birth, or 5 times (5x) on the first 5 d of birth with rGM-CSF or PBS i.n. (1 rGM-CSF or PBS treatment per day). rGM-CSF–treated or PBS-treated Csf2−/− mice were sacrificed on PND 7. (A) Lungs were homogenized and CD11b+F4/80+ myeloid cells were assessed for CD11c and SiglecF expression. (B) Expression of FSC, SSC, Ly-6C, CD64, CD11c, F4/80, and SiglecF on SiglecFloCD11cloCD11bhiLy-6Chi monocytes and SiglecFintCD11chiCD11bhiLy-6Chi immature AMFs harvested from Csf2−/− mice treated for 5 consecutive days with rGM-CSF. (C) 5 wk after 5 consecutive rGM-CSF treatments, Csf2−/− mice were sacrificed and the presence of CD11c+SiglecF+ cells in the BAL was evaluated. (D) WT or Csf2−/− mice treated with 5 consecutive treatments of rGM-CSF or PBS were sacrificed at 7 wk of age, and the development of alveolar proteinosis was evaluated by measuring the protein concentration in the BAL. Data represent two (D) and three (A– C) independent experiments, with at least three recipient mice per time point.
Figure 7.
Figure 7.
Terminal differentiation of GM-CSF–rescued immature AMFs requires a GM-CSF–replete host.Csf2−/− mice treated with 5 consecutive treatments of rGM-CSF were sacrificed at 7 d of age. The lungs were homogenized and CD45.2+ CD11cintSiglecFint preAMFs were FACS sorted (profile of the sorted cells before transfer is shown in A) and transferred into CD45.1+ WT mice on their DOB. 2 d, 9 d, and 6 wk after transfer, CD45.1+ recipient mice were sacrificed, and the presence of CD45.2+ donor-derived cells was evaluated in the lungs (B–D) and BAL (E). Their CD11b, F4/80, CD11c, and SiglecF expression profile was also assessed. Data represent two independent experiments with at least three recipient mice per time point.

References

    1. Ajami B., Bennett J.L., Krieger C., Tetzlaff W., Rossi F.M. 2007. Local self-renewal can sustain CNS microglia maintenance and function throughout adult life. Nat. Neurosci. 10:1538–1543 10.1038/nn2014
    1. Ajami B., Bennett J.L., Krieger C., McNagny K.M., Rossi F.M. 2011. Infiltrating monocytes trigger EAE progression, but do not contribute to the resident microglia pool. Nat. Neurosci. 14:1142–1149 10.1038/nn.2887
    1. Bain C.C., Scott C.L., Uronen-Hansson H., Gudjonsson S., Jansson O., Grip O., Guilliams M., Malissen B., Agace W.W., Mowat A.M. 2013. Resident and pro-inflammatory macrophages in the colon represent alternative context-dependent fates of the same Ly6Chi monocyte precursors. Mucosal Immunol. 6:498–510 10.1038/mi.2012.89
    1. Baker A.D., Malur A., Barna B.P., Ghosh S., Kavuru M.S., Malur A.G., Thomassen M.J. 2010. Targeted PPARgamma deficiency in alveolar macrophages disrupts surfactant catabolism. J. Lipid Res. 51:1325–1331 10.1194/jlr.M001651
    1. Bogunovic M., Ginhoux F., Helft J., Shang L., Hashimoto D., Greter M., Liu K., Jakubzick C., Ingersoll M.A., Leboeuf M., et al. 2009. Origin of the lamina propria dendritic cell network. Immunity. 31:513–525 10.1016/j.immuni.2009.08.010
    1. Bonfield T.L., Farver C.F., Barna B.P., Malur A., Abraham S., Raychaudhuri B., Kavuru M.S., Thomassen M.J. 2003. Peroxisome proliferator-activated receptor-gamma is deficient in alveolar macrophages from patients with alveolar proteinosis. Am. J. Respir. Cell Mol. Biol. 29:677–682 10.1165/rcmb.2003-0148OC
    1. Bouwens L., Baekeland M., De Zanger R., Wisse E. 1986a. Quantitation, tissue distribution and proliferation kinetics of Kupffer cells in normal rat liver. Hepatology. 6:718–722 10.1002/hep.1840060430
    1. Bouwens L., Knook D.L., Wisse E. 1986b. Local proliferation and extrahepatic recruitment of liver macrophages (Kupffer cells) in partial-body irradiated rats. J. Leukoc. Biol. 39:687–697
    1. Chorro L., Sarde A., Li M., Woollard K.J., Chambon P., Malissen B., Kissenpfennig A., Barbaroux J.B., Groves R., Geissmann F. 2009. Langerhans cell (LC) proliferation mediates neonatal development, homeostasis, and inflammation-associated expansion of the epidermal LC network. J. Exp. Med. 206:3089–3100 10.1084/jem.20091586
    1. Costa G., Kouskoff V., Lacaud G. 2012. Origin of blood cells and HSC production in the embryo. Trends Immunol. 33:215–223 10.1016/j.it.2012.01.012
    1. Gautier E.L., Chow A., Spanbroek R., Marcelin G., Greter M., Jakubzick C., Bogunovic M., Leboeuf M., van Rooijen N., Habenicht A.J., et al. 2012a. Systemic analysis of PPARγ in mouse macrophage populations reveals marked diversity in expression with critical roles in resolution of inflammation and airway immunity. J. Immunol. 189:2614–2624 10.4049/jimmunol.1200495
    1. Gautier E.L., Shay T., Miller J., Greter M., Jakubzick C., Ivanov S., Helft J., Chow A., Elpek K.G., Gordonov S., et al. ; Immunological Genome Consortium 2012b. Gene-expression profiles and transcriptional regulatory pathways that underlie the identity and diversity of mouse tissue macrophages. Nat. Immunol. 13:1118–1128 10.1038/ni.2419
    1. Geissmann F., Jung S., Littman D.R. 2003. Blood monocytes consist of two principal subsets with distinct migratory properties. Immunity. 19:71–82 10.1016/S1074-7613(03)00174-2
    1. Geissmann F., Manz M.G., Jung S., Sieweke M.H., Merad M., Ley K. 2010. Development of monocytes, macrophages, and dendritic cells. Science. 327:656–661 10.1126/science.1178331
    1. Ginhoux F., Greter M., Leboeuf M., Nandi S., See P., Gokhan S., Mehler M.F., Conway S.J., Ng L.G., Stanley E.R., et al. 2010. Fate mapping analysis reveals that adult microglia derive from primitive macrophages. Science. 330:841–845 10.1126/science.1194637
    1. Godleski J.J., Brain J.D. 1972. The origin of alveolar macrophages in mouse radiation chimeras. J. Exp. Med. 136:630–643 10.1084/jem.136.3.630
    1. Golde D.W. 1979. Alveolar proteinosis and the overfed macrophage. Chest. 76:119–120 10.1378/chest.76.2.119
    1. Golde D.W., Territo M., Finley T.N., Cline M.J. 1976. Defective lung macrophages in pulmonary alveolar proteinosis. Ann. Intern. Med. 85:304–309 10.7326/0003-4819-85-3-304
    1. Gomez Perdiguero E., Schulz C., Geissmann F. 2013. Development and homeostasis of “resident” myeloid cells: The case of the microglia. Glia. 61:112–120 10.1002/glia.22393
    1. Gonzalez-Rothi R.J., Harris J.O. 1986. Pulmonary alveolar proteinosis. Further evaluation of abnormal alveolar macrophages. Chest. 90:656–661 10.1378/chest.90.5.656
    1. Greter M., Helft J., Chow A., Hashimoto D., Mortha A., Agudo-Cantero J., Bogunovic M., Gautier E.L., Miller J., Leboeuf M., et al. 2012a. GM-CSF controls nonlymphoid tissue dendritic cell homeostasis but is dispensable for the differentiation of inflammatory dendritic cells. Immunity. 36:1031–1046 10.1016/j.immuni.2012.03.027
    1. Greter M., Lelios I., Pelczar P., Hoeffel G., Price J., Leboeuf M., Kündig T.M., Frei K., Ginhoux F., Merad M., Becher B. 2012b. Stroma-derived interleukin-34 controls the development and maintenance of langerhans cells and the maintenance of microglia. Immunity. 37:1050–1060 10.1016/j.immuni.2012.11.001
    1. Harris J.O. 1979. Pulmonary alveolar proteinosis: abnormal in vitro function of alveolar macrophages. Chest. 76:156–159 10.1378/chest.76.2.156
    1. Hashimoto D., Chow A., Noizat C., Teo P., Beasley M.B., Leboeuf M., Becker C.D., See P., Price J., Lucas D., et al. 2013. Tissue-resident macrophages self-maintain locally throughout adult life with minimal contribution from circulating monocytes. Immunity. 38:792–804 10.1016/j.immuni.2013.04.004
    1. Hoeffel G., Wang Y., Greter M., See P., Teo P., Malleret B., Leboeuf M., Low D., Oller G., Almeida F., et al. 2012. Adult Langerhans cells derive predominantly from embryonic fetal liver monocytes with a minor contribution of yolk sac-derived macrophages. J. Exp. Med. 209:1167–1181 10.1084/jem.20120340
    1. Huffman J.A., Hull W.M., Dranoff G., Mulligan R.C., Whitsett J.A. 1996. Pulmonary epithelial cell expression of GM-CSF corrects the alveolar proteinosis in GM-CSF-deficient mice. J. Clin. Invest. 97:649–655 10.1172/JCI118461
    1. Kamath A.T., Henri S., Battye F., Tough D.F., Shortman K. 2002. Developmental kinetics and lifespan of dendritic cells in mouse lymphoid organs. Blood. 100:1734–1741
    1. Kennedy D.W., Abkowitz J.L. 1997. Kinetics of central nervous system microglial and macrophage engraftment: analysis using a transgenic bone marrow transplantation model. Blood. 90:986–993
    1. Kennedy D.W., Abkowitz J.L. 1998. Mature monocytic cells enter tissues and engraft. Proc. Natl. Acad. Sci. USA. 95:14944–14949 10.1073/pnas.95.25.14944
    1. Lambrecht B.N. 2006. Alveolar macrophage in the driver’s seat. Immunity. 24:366–368 10.1016/j.immuni.2006.03.008
    1. Landsman L., Jung S. 2007. Lung macrophages serve as obligatory intermediate between blood monocytes and alveolar macrophages. J. Immunol. 179:3488–3494
    1. LeVine A.M., Reed J.A., Kurak K.E., Cianciolo E., Whitsett J.A. 1999. GM-CSF-deficient mice are susceptible to pulmonary group B streptococcal infection. J. Clin. Invest. 103:563–569 10.1172/JCI5212
    1. Liu K., Waskow C., Liu X., Yao K., Hoh J., Nussenzweig M. 2007. Origin of dendritic cells in peripheral lymphoid organs of mice. Nat. Immunol. 8:578–583 10.1038/ni1462
    1. Misharin A.V., Morales-Nebreda L., Mutlu G.M., Budinger G.R., Perlman H. 2013. Flow Cytometric Analysis of the Macrophages and Dendritic Cell Subsets in the Mouse Lung. Am. J. Respir. Cell Mol. Biol. (Epub ahead of print)
    1. Morrisey E.E., Hogan B.L. 2010. Preparing for the first breath: genetic and cellular mechanisms in lung development. Dev. Cell. 18:8–23 10.1016/j.devcel.2009.12.010
    1. Murphy J., Summer R., Wilson A.A., Kotton D.N., Fine A. 2008. The prolonged life-span of alveolar macrophages. Am. J. Respir. Cell Mol. Biol. 38:380–385 10.1165/rcmb.2007-0224RC
    1. Nugent K.M., Pesanti E.L. 1983. Macrophage function in pulmonary alveolar proteinosis. Am. Rev. Respir. Dis. 127:780–781
    1. Paine R., III, Morris S.B., Jin H., Wilcoxen S.E., Phare S.M., Moore B.B., Coffey M.J., Toews G.B. 2001. Impaired functional activity of alveolar macrophages from GM-CSF-deficient mice. Am. J. Physiol. Lung Cell. Mol. Physiol. 281:L1210–L1218
    1. Plantinga M., Guilliams M., Vanheerswynghels M., Deswarte K., Branco-Madeira F., Toussaint W., Vanhoutte L., Neyt K., Killeen N., Malissen B., et al. 2013. Conventional and monocyte-derived CD11b(+) dendritic cells initiate and maintain T helper 2 cell-mediated immunity to house dust mite allergen. Immunity. 38:322–335 10.1016/j.immuni.2012.10.016
    1. Reed J.A., Ikegami M., Cianciolo E.R., Lu W., Cho P.S., Hull W., Jobe A.H., Whitsett J.A. 1999. Aerosolized GM-CSF ameliorates pulmonary alveolar proteinosis in GM-CSF-deficient mice. Am. J. Physiol. 276:L556–L563
    1. Reed J.A., Ikegami M., Robb L., Begley C.G., Ross G., Whitsett J.A. 2000. Distinct changes in pulmonary surfactant homeostasis in common beta-chain- and GM-CSF-deficient mice. Am. J. Physiol. Lung Cell. Mol. Physiol. 278:L1164–L1171
    1. Schulz C., Gomez Perdiguero E., Chorro L., Szabo-Rogers H., Cagnard N., Kierdorf K., Prinz M., Wu B., Jacobsen S.E., Pollard J.W., et al. 2012. A lineage of myeloid cells independent of Myb and hematopoietic stem cells. Science. 336:86–90 10.1126/science.1219179
    1. Serbina N.V., Pamer E.G. 2006. Monocyte emigration from bone marrow during bacterial infection requires signals mediated by chemokine receptor CCR2. Nat. Immunol. 7:311–317 10.1038/ni1309
    1. Shibata Y., Berclaz P.Y., Chroneos Z.C., Yoshida M., Whitsett J.A., Trapnell B.C. 2001. GM-CSF regulates alveolar macrophage differentiation and innate immunity in the lung through PU.1. Immunity. 15:557–567 10.1016/S1074-7613(01)00218-7
    1. Suzuki T., Sakagami T., Rubin B.K., Nogee L.M., Wood R.E., Zimmerman S.L., Smolarek T., Dishop M.K., Wert S.E., Whitsett J.A., et al. 2008. Familial pulmonary alveolar proteinosis caused by mutations in CSF2RA. J. Exp. Med. 205:2703–2710 10.1084/jem.20080990
    1. Takahashi K., Yamamura F., Naito M. 1989. Differentiation, maturation, and proliferation of macrophages in the mouse yolk sac: a light-microscopic, enzyme-cytochemical, immunohistochemical, and ultrastructural study. J. Leukoc. Biol. 45:87–96
    1. Tamoutounour S., Henri S., Lelouard H., de Bovis B., de Haar C., van der Woude C.J., Woltman A.M., Reyal Y., Bonnet D., Sichien D., et al. 2012. CD64 distinguishes macrophages from dendritic cells in the gut and reveals the Th1-inducing role of mesenteric lymph node macrophages during colitis. Eur. J. Immunol. 42:3150–3166 10.1002/eji.201242847
    1. Tarling J.D., Lin H.S., Hsu S. 1987. Self-renewal of pulmonary alveolar macrophages: evidence from radiation chimera studies. J. Leukoc. Biol. 42:443–446
    1. Thomassen M.J., Barna B.P., Malur A.G., Bonfield T.L., Farver C.F., Malur A., Dalrymple H., Kavuru M.S., Febbraio M. 2007. ABCG1 is deficient in alveolar macrophages of GM-CSF knockout mice and patients with pulmonary alveolar proteinosis. J. Lipid Res. 48:2762–2768 10.1194/jlr.P700022-JLR200
    1. van Furth R., Cohn Z.A. 1968. The origin and kinetics of mononuclear phagocytes. J. Exp. Med. 128:415–435 10.1084/jem.128.3.415
    1. van oud Alblas A.B., van Furth R. 1979. Origin, Kinetics, and characteristics of pulmonary macrophages in the normal steady state. J. Exp. Med. 149:1504–1518 10.1084/jem.149.6.1504
    1. Varol C., Vallon-Eberhard A., Elinav E., Aychek T., Shapira Y., Luche H., Fehling H.J., Hardt W.D., Shakhar G., Jung S. 2009. Intestinal lamina propria dendritic cell subsets have different origin and functions. Immunity. 31:502–512 10.1016/j.immuni.2009.06.025
    1. Vermaelen K., Pauwels R. 2004. Accurate and simple discrimination of mouse pulmonary dendritic cell and macrophage populations by flow cytometry: methodology and new insights. Cytometry A. 61:170–177 10.1002/cyto.a.20064
    1. Virolainen M. 1968. Hematopoietic origin of macrophages as studied by chromosome markers in mice. J. Exp. Med. 127:943–952 10.1084/jem.127.5.943
    1. Wang Y., Szretter K.J., Vermi W., Gilfillan S., Rossini C., Cella M., Barrow A.D., Diamond M.S., Colonna M. 2012. IL-34 is a tissue-restricted ligand of CSF1R required for the development of Langerhans cells and microglia. Nat. Immunol. 13:753–760 10.1038/ni.2360
    1. Willinger T., Rongvaux A., Takizawa H., Yancopoulos G.D., Valenzuela D.M., Murphy A.J., Auerbach W., Eynon E.E., Stevens S., Manz M.G., Flavell R.A. 2011. Human IL-3/GM-CSF knock-in mice support human alveolar macrophage development and human immune responses in the lung. Proc. Natl. Acad. Sci. USA. 108:2390–2395 10.1073/pnas.1019682108
    1. Yamamoto T., Naito M., Moriyama H., Umezu H., Matsuo H., Kiwada H., Arakawa M. 1996. Repopulation of murine Kupffer cells after intravenous administration of liposome-encapsulated dichloromethylene diphosphonate. Am. J. Pathol. 149:1271–1286
    1. Yona S., Kim K.W., Wolf Y., Mildner A., Varol D., Breker M., Strauss-Ayali D., Viukov S., Guilliams M., Misharin A., et al. 2013. Fate mapping reveals origins and dynamics of monocytes and tissue macrophages under homeostasis. Immunity. 38:79–91 10.1016/j.immuni.2012.12.001
    1. Yoshida M., Ikegami M., Reed J.A., Chroneos Z.C., Whitsett J.A. 2001. GM-CSF regulates protein and lipid catabolism by alveolar macrophages. Am. J. Physiol. Lung Cell. Mol. Physiol. 280:L379–L386
    1. Zsengellér Z.K., Reed J.A., Bachurski C.J., LeVine A.M., Forry-Schaudies S., Hirsch R., Whitsett J.A. 1998. Adenovirus-mediated granulocyte-macrophage colony-stimulating factor improves lung pathology of pulmonary alveolar proteinosis in granulocyte-macrophage colony-stimulating factor-deficient mice. Hum. Gene Ther. 9:2101–2109 10.1089/hum.1998.9.14-2101

Source: PubMed

3
Abonner