Crosstalk between nonclassical monocytes and alveolar macrophages mediates transplant ischemia-reperfusion injury through classical monocyte recruitment

Chitaru Kurihara, Emilia Lecuona, Qiang Wu, Wenbin Yang, Félix L Núñez-Santana, Mahzad Akbarpour, Xianpeng Liu, Ziyou Ren, Wenjun Li, Melissa Querrey, Sowmya Ravi, Megan L Anderson, Emily Cerier, Haiying Sun, Megan E Kelly, Hiam Abdala-Valencia, Ali Shilatifard, Thalachallour Mohanakumar, G R Scott Budinger, Daniel Kreisel, Ankit Bharat, Chitaru Kurihara, Emilia Lecuona, Qiang Wu, Wenbin Yang, Félix L Núñez-Santana, Mahzad Akbarpour, Xianpeng Liu, Ziyou Ren, Wenjun Li, Melissa Querrey, Sowmya Ravi, Megan L Anderson, Emily Cerier, Haiying Sun, Megan E Kelly, Hiam Abdala-Valencia, Ali Shilatifard, Thalachallour Mohanakumar, G R Scott Budinger, Daniel Kreisel, Ankit Bharat

Abstract

Primary graft dysfunction (PGD) is the predominant cause of early graft loss following lung transplantation. We recently demonstrated that donor pulmonary intravascular nonclassical monocytes (NCM) initiate neutrophil recruitment. Simultaneously, host-origin classical monocytes (CM) permeabilize the vascular endothelium to allow neutrophil extravasation necessary for PGD. Here, we show that a CCL2-CCR2 axis is necessary for CM recruitment. Surprisingly, although intravital imaging and multichannel flow cytometry revealed that depletion of donor NCM abrogated CM recruitment, single cell RNA sequencing identified donor alveolar macrophages (AM) as predominant CCL2 secretors. Unbiased transcriptomic analysis of murine tissues combined with murine KOs and chimeras indicated that IL-1β production by donor NCM was responsible for the early activation of AM and CCL2 release. IL-1β production by NCM was NLRP3 inflammasome dependent and inhibited by treatment with a clinically approved sulphonylurea. Production of CCL2 in the donor AM occurred through IL-1R-dependent activation of the PKC and NF-κB pathway. Accordingly, we show that IL-1β-dependent paracrine interaction between donor NCM and AM leads to recruitment of recipient CM necessary for PGD. Since depletion of donor NCM, IL-1β, or IL-1R antagonism and inflammasome inhibition abrogated recruitment of CM and PGD and are feasible using FDA-approved compounds, our findings may have potential for clinical translation.

Keywords: Organ transplantation; Transplantation.

Conflict of interest statement

Conflict of interest: DK has a pending US patent entitled “Compositions and methods for detecting CCR2 receptors” (application no. 15/611,577).

Figures

Figure 1. The CCL2-CCR2 axis is necessary…
Figure 1. The CCL2-CCR2 axis is necessary for recipient classical monocytes (CM) recruitment to the allograft.
(A) Flow cytometry quantification of CM (live CD45+Ly6G–NK1.1–CD11b+SiglecF–CD24–Ly6Chi) recruited into the allograft after treatment of recipients with i.v. IgG isotype or anti-CCR2 antibodies (n = 5). (B) Flow cytometry quantification of CM as described in A, recruited into the allograft using WT or Ccr2–/– recipient mice (n = 5). (C) Flow cytometry quantification of CM (live CD45+Ly6G–NK1.1–CD19–CD11b+CF4/80–CD11c–Ly6Chi) in spleens of WT or Ccr2–/– recipients after lung transplant (n = 5–6). (D) Flow cytometry quantification of CM as described in A, recruited into the allograft after treatment of recipients with i.v. IgG isotype or anti-CCL2 antibodies in recipient mice (n = 3). Graphs show means ± SD. Graphs were analyzed by unpaired Student’s t test. ***P < 0.001; ****P < 0.0001.
Figure 2. Depletion of donor nonclassical monocytes…
Figure 2. Depletion of donor nonclassical monocytes (NCM) suppresses the recruitment of recipient splenic classical monocytes (CM) to the allograft.
(AC) Intravital 2-photon imaging between 2 and 2.5 hours after reperfusion. (A) Representative still images of WT and Nr4a1–/– donor grafts. Green, CCR2-GFP; red, Qdot655 blood vessels. (B and C) CCR2-GFP cell density (B) and percent of extravasated CCR2-GFP (C) calculated using NIH ImageJ software in WT and Nr4a1–/– mice grafts after transplantation. (D) Flow cytometry quantification of CM (live CD45+Ly6G–NK1.1–CD11b+SiglecF–CD24–Ly6Chi) recruited into the allograft after i.v. injection of PBS liposomes (PBS-lip) or clodronate liposomes (Clo-lip) in donor mice or using WT or Nr4a1–/– as donor lungs (n = 5). (E) Normalized counts per minute (CPM) of Ccl2 in sorted donor NCM isolated from allografts 2 and 24 hours after transplant. (F) Relative Ccl2 mRNA levels of human and mouse NCM isolated before and after reperfusion (n = 3). (G) CCL2 levels in blood from mice in D (n = 5). Graphs show means ± SD. The graph in B was analyzed by 2-way ANOVA followed by Sidak’s post hoc test. *WT versus Nr4a1–/–; #WT 2 hours versus WT 2.5 hours. The graph in E was analyzed by 1-way ANOVA followed by Tukey’s post hoc test. Graphs in C, D, F, and G were analyzed by unpaired Student’s t test. #P < 0.05, **P < 0.01; ***P < 0.001; ****P < 0.0001. Scale bar: 50 μm.
Figure 3. Depletion of donor alveolar macrophages…
Figure 3. Depletion of donor alveolar macrophages (AM) suppresses recruitment of recipient classical monocytes (CM) to the transplanted lung.
(A) Flow cytometry showing percentage of cells of donor (CD45.1) and recipient (CD45.2) origin in the allograft 24 hours after transplantation. Neutrophils were gated as live CD45+Ly6G+CD11b+CD24+SSChi; AM were gated as live CD45+Ly6G–NK1.1–SiglecF+CD64+CD11c+; CM were gated as live CD45+Ly6G–NK1.1–CD11b+SiglecF–CD24–Ly6Chi; NCM were gated as live CD45+Ly6G–NK1.1–CD11b+SiglecF–CD24–Ly6Clo (n = 3); and interstitial macrophages (IM) were gated as live CD45+Ly6G–NK1.1–CD11b+MHC II+CD11c+CD64+CD24− (n = 3–6). (B) UMAP plot (left) and feature plots (middle and right) showing specific cell populations and expression of Ccl2 in naive lungs and in allografts (Allo) 24 hours after transplant. (C) Flow cytometry showing percentage of RFP+ cells in lung allografts after transplantation of donor Ccl2-rfp grafts into WT recipient. AM and NCM were gated as in A. (D) Normalized counts per minute (CPM) of Ccl2 in sorted mouse AM isolated from allografts 2 hours after transplant (n = 4). (E) Relative Ccl2 mRNA levels of human and mouse AM isolated before and after reperfusion (n = 3–5). (F) Flow cytometry quantification of CM gated as in A, recruited into the allograft after intratracheal administration of PBS liposomes (PBS-lip) or clodronate liposomes (Clo-lip) in the donor mice (n = 4). (G) Flow cytometry quantification of extravasated neutrophils in the allograft gated as in A, after intratracheal administration of PBS-lip or Clo-lip in donor mice (n = 3). (H) Relative Ccl2 mRNA levels in AM isolated from WT, Cd169Cre, Ccl-rfpfl/fl, and Cd169CreCcl2-rfpfl/fl allografts and compared with naive AM (n = 5). (I) Blood CCL2 levels after transplant combinations described in H (n = 5). (J) Flow cytometry quantification of CM gated as in A, after transplant combinations described in H (n = 5). Graphs show means ± SD. Graphs in CG were analyzed by unpaired Student’s t test. Graphs in HJ were analyzed by 1-way ANOVA followed by Tukey’s post hoc test. *P < 0.05; **P < 0.01; ***P < 0.001; ****P < 0.0001.
Figure 4. IL-1β is necessary for donor…
Figure 4. IL-1β is necessary for donor alveolar macrophages (AM) to produce CCL2.
(A) AM were isolated from WT or Il1r–/– mice and incubated in vitro with PBS or 1 ng/μL of mouse recombinant IL-1β. Ccl2 mRNA expression was measured by qPCR 24 hours after incubation (n = 6–8). (B) AM isolated from WT mice were incubated for 30 minutes in vitro with DMSO (control), 50 μM LY294002 (LY), 10 μM Bisindolylmaleimide-I (Bis), or 1 μM BAY11-7082 before adding 1 ng/μL of mouse recombinant IL-1β. Ccl2 mRNA expression was measured by qPCR 24 hours after incubation (n = 3–7). (C) Donor AM were isolated from WT or Il1r–/– allografts 24 hours after transplant, and Ccl2 mRNA expression was measured by qPCR (n = 4–5). (D) Flow cytometry quantification of CM (live CD45+Ly6G–NK1.1–CD11b+SiglecF–CD24–Ly6Chi) from allografts harvested at the indicated times from experiments in which donor lungs were treated with PBS or anakinra, or in which recipients were treated with isotype control and anti–IL-1β antibody (n = 3 per group). Graphs show means ± SD. Graphs in AC were analyzed by unpaired Student’s t test. Graph in D was analyzed by 2-way ANOVA, followed by Sidak’s post hoc test. *PBS versus anakinra; #isotype versus anti–IL-1β antibody. *P < 0.05; **P < 0.01; ***P < 0.001; ****P < 0.0001; ###P < 0.001; ####P < 0.0001.
Figure 5. Nonclassical monocyte–derived (NCM-derived) IL-1β promotes…
Figure 5. Nonclassical monocyte–derived (NCM-derived) IL-1β promotes CCL2 production by donor alveolar macrophages (AM).
(A) UMAP plot (left) and feature plots (middle and right) showing specific cell populations and expression of Il1β in naive lungs and in allografts (Allo) 24 hours after transplantation. (B) Ccl2 mRNA levels determined by qPCR in AM from the allograft after i.v. administration of IgG isotype or anti-Ly6G antibodies in recipient mice (n = 2–3). (C) Relative Il1β mRNA levels of mouse donor NCM isolated before and after reperfusion (n = 3–4). (D) Ccl2 mRNA levels determined by qPCR in AM isolated WT or Nr4a1–/– lung allografts 24 hours after transplantation (n = 4–6). (E) Flow cytometry quantification of CM (live CD45+Ly6G–NK1.1–CD11b+SiglecF–CD24–Ly6Chi) recruited into the allograft after treatment of recipients with i.v. IgG isotype or anti-CXCL2 antibodies (n = 5). Graphs show means ± SD. Graphs were analyzed by unpaired Student’s t test.*P < 0.05.
Figure 6. Nonclassical monocytes (NCM) produce IL-1β…
Figure 6. Nonclassical monocytes (NCM) produce IL-1β via NLRP3 inflammasome activation.
(A) Ccl2 mRNA levels determined by qPCR in AM isolated from WT or Nlrp3–/– lung allografts 24 hours after transplantation (n = 3). (B) Flow cytometry quantification of CM (live CD45+Ly6G–NK1.1–CD11b+SiglecF–CD24–Ly6Chi) recruited to allografts from WT, Nlrp3–/–, or Nlrp3–/– reconstituted with WT NCM donor mice (n = 3–4). (C) Flow cytometry quantification of neutrophils (live CD45+Ly6G+CD11b+CD24+SSChi) recruited into WT of Nlrp3–/– allografts 24 hours after transplantation (n = 5–7). (D) Flow cytometry quantification of CM gated as in C, recruited into the allograft after donor treatment with 50 μg/g body weight glyburide (n = 4). (E) Flow cytometry quantification of neutrophils gated as in B, recruited into the allograft after i.p. administration glyburide in the donor mice (n = 4–7). (F) Flow cytometry quantification of extravasated neutrophils in the allograft gated as in B, after donor treatment with glyburide (n = 4). Graphs show means ± SD. Graph in C was analyzed by 1-way ANOVA, followed by Tukey’s post hoc test. All other graphs were analyzed by unpaired Student’s t test. *P < 0.05; ***P < 0.001; ****P < 0.0001.

References

    1. Bharat A, et al. Immunological link between primary graft dysfunction and chronic lung allograft rejection. Ann Thorac Surg. 2008;86(1):189–195. doi: 10.1016/j.athoracsur.2008.03.073.
    1. Bharat A, Kreisel D. Immunopathogenesis of primary graft dysfunction after lung transplantation. Ann Thorac Surg. 2018;105(3):671–674. doi: 10.1016/j.athoracsur.2017.11.007.
    1. Kreisel D, et al. Bcl3 prevents acute inflammatory lung injury in mice by restraining emergency granulopoiesis. J Clin Invest. 2011;121(1):265–276. doi: 10.1172/JCI42596.
    1. Sayah DM, et al. Neutrophil extracellular traps are pathogenic in primary graft dysfunction after lung transplantation. Am J Respir Crit Care Med. 2015;191(4):455–463. doi: 10.1164/rccm.201406-1086OC.
    1. Chiu S, Bharat A. Role of monocytes and macrophages in regulating immune response following lung transplantation. Curr Opin Organ Transplant. 2016;21(3):239–245. doi: 10.1097/MOT.0000000000000313.
    1. Zheng Z, et al. Donor pulmonary intravascular nonclassical monocytes recruit recipient neutrophils and mediate primary lung allograft dysfunction. Sci Transl Med. 2017;9(394):eaal4508. doi: 10.1126/scitranslmed.aal4508.
    1. Bharat A, et al. Non-classical monocytes in tissue injury and cancer. Oncotarget. 2017;8(63):106171–106172. doi: 10.18632/oncotarget.22584.
    1. Lamb CA, Kirby JA. Donor intravascular monocyte trafficking: a potential therapeutic target for primary graft dysfunction following lung transplantation? Thorax. 2018;73(4):303–304. doi: 10.1136/thoraxjnl-2017-210274.
    1. Hsiao HM, et al. Spleen-derived classical monocytes mediate lung ischemia-reperfusion injury through IL-1β. J Clin Invest. 2018;128(7):2833–2847. doi: 10.1172/JCI98436.
    1. Swirski FK, et al. Identification of splenic reservoir monocytes and their deployment to inflammatory sites. Science. 2009;325(5940):612–616. doi: 10.1126/science.1175202.
    1. Peters W, et al. Chemokine receptor 2 serves an early and essential role in resistance to Mycobacterium tuberculosis. Proc Natl Acad Sci U S A. 2001;98(14):7958–7963. doi: 10.1073/pnas.131207398.
    1. Tsou CL, et al. Critical roles for CCR2 and MCP-3 in monocyte mobilization from bone marrow and recruitment to inflammatory sites. J Clin Invest. 2007;117(4):902–909. doi: 10.1172/JCI29919.
    1. Gelman AE, et al. CCR2 regulates monocyte recruitment as well as CD4 T1 allorecognition after lung transplantation. Am J Transplant. 2010;10(5):1189–1199. doi: 10.1111/j.1600-6143.2010.03101.x.
    1. Misharin AV, et al. Monocyte-derived alveolar macrophages drive lung fibrosis and persist in the lung over the life span. J Exp Med. 2017;214(8):2387–2404. doi: 10.1084/jem.20162152.
    1. Bissonnette EY, et al. Cross-talk between alveolar macrophages and lung epithelial cells is essential to maintain lung homeostasis. Front Immunol. 2020;11:583042.
    1. Akbarpour M, et al. Residual endotoxin induces primary graft dysfunction through ischemia/reperfusion-primed alveolar macrophages. J Clin Invest. 2020;130(8):4456–4469. doi: 10.1172/JCI135838.
    1. Tsui P, et al. Generation, characterization and biological activity of CCL2 (MCP-1/JE) and CCL12 (MCP-5) specific antibodies. Hum Antibodies. 2007;16(3–4):117–125.
    1. Shi C, et al. Bone marrow mesenchymal stem and progenitor cells induce monocyte emigration in response to circulating toll-like receptor ligands. Immunity. 2011;34(4):590–601. doi: 10.1016/j.immuni.2011.02.016.
    1. Koch CM, et al. A beginner’s guide to analysis of RNA sequencing data. Am J Respir Cell Mol Biol. 2018;59(2):145–157. doi: 10.1165/rcmb.2017-0430TR.
    1. Bharat A, et al. Flow cytometry reveals similarities between lung macrophages in humans and mice. Am J Respir Cell Mol Biol. 2016;54(1):147–149. doi: 10.1165/rcmb.2015-0147LE.
    1. Burke SJ, et al. IL-1β reciprocally regulates chemokine and insulin secretion in pancreatic β-cells via NF-κB. Am J Physiol Endocrinol Metab. 2015;309(8):E715–E726. doi: 10.1152/ajpendo.00153.2015.
    1. Kaushik DK, et al. Interleukin-1β orchestrates underlying inflammatory responses in microglia via Krüppel-like factor 4. J Neurochem. 2013;127(2):233–244. doi: 10.1111/jnc.12382.
    1. Mendoza-Rodriguez MG, et al. IL-1β inflammatory cytokine-induced TP63 isoform ΔNP63α signaling cascade contributes to cisplatin resistance in human breast cancer cells. Int J Mol Sci. 2019;20(2):E270.
    1. Lin CY, et al. IL-1β-induced mesenchymal stem cell migration involves MLCK activation via PKC signaling. Cell Transplant. 2015;24(10):2011–2028. doi: 10.3727/096368914X685258.
    1. Boivin G, et al. Durable and controlled depletion of neutrophils in mice. Nat Commun. 2020;11(1):2762. doi: 10.1038/s41467-020-16596-9.
    1. Swanson KV, et al. The NLRP3 inflammasome: molecular activation and regulation to therapeutics. Nat Rev Immunol. 2019;19(8):477–489. doi: 10.1038/s41577-019-0165-0.
    1. Lamkanfi M, et al. Glyburide inhibits the Cryopyrin/Nalp3 inflammasome. J Cell Biol. 2009;187(1):61–70. doi: 10.1083/jcb.200903124.
    1. Serbina NV, Pamer EG. Monocyte emigration from bone marrow during bacterial infection requires signals mediated by chemokine receptor CCR2. Nat Immunol. 2006;7(3):311–317. doi: 10.1038/ni1309.
    1. Gschwandtner M, et al. More than just attractive: how CCL2 influences myeloid cell behavior beyond chemotaxis. Front Immunol. 2019;10:2759.
    1. Wang B, et al. In vivo imaging implicates CCR2(+) monocytes as regulators of neutrophil recruitment during arthritis. Cell Immunol. 2012;278(1–2):103–112.
    1. Mantovani A, et al. Interleukin-1 and related cytokines in the regulation of inflammation and immunity. Immunity. 2019;50(4):778–795. doi: 10.1016/j.immuni.2019.03.012.
    1. Dinarello CA. Biologic basis for interleukin-1 in disease. Blood. 1996;87(6):2095–2147. doi: 10.1182/blood.V87.6.2095.bloodjournal8762095.
    1. Liu QS, et al. Erythropoietin pretreatment exerts anti-inflammatory effects in hepatic ischemia/reperfusion-injured rats via suppression of the TLR2/NF-κB pathway. Transplant Proc. 2015;47(2):283–289. doi: 10.1016/j.transproceed.2014.10.045.
    1. Miura K, et al. Protective effect of neutralization of the extracellular high-mobility group box 1 on renal ischemia-reperfusion injury in miniature swine. Transplantation. 2014;98(9):937–943. doi: 10.1097/TP.0000000000000358.
    1. Sloand EM, et al. Inhibition of interleukin-1beta-converting enzyme in human hematopoietic progenitor cells results in blockade of cytokine-mediated apoptosis and expansion of their proliferative potential. Exp Hematol. 1998;26(11):1093–1099.
    1. Rega FR, et al. IL-1beta in bronchial lavage fluid is a non-invasive marker that predicts the viability of the pulmonary graft from the non-heart-beating donor. J Heart Lung Transplant. 2005;24(1):20–28. doi: 10.1016/j.healun.2003.10.004.
    1. Andreasson AS, et al. Profiling inflammation and tissue injury markers in perfusate and bronchoalveolar lavage fluid during human ex vivo lung perfusion. Eur J Cardiothorac Surg. 2017;51(3):577–586.
    1. Geudens N, et al. Azithromycin reduces airway inflammation in a murine model of lung ischaemia reperfusion injury. Transpl Int. 2008;21(7):688–695. doi: 10.1111/j.1432-2277.2008.00670.x.
    1. Rollins BJ, et al. Cytokine-activated human endothelial cells synthesize and secrete a monocyte chemoattractant, MCP-1/JE. Am J Pathol. 1990;136(6):1229–1233.
    1. Nelken NA, et al. Monocyte chemoattractant protein-1 in human atheromatous plaques. J Clin Invest. 1991;88(4):1121–1127. doi: 10.1172/JCI115411.
    1. Brough D, Rothwell NJ. Caspase-1-dependent processing of pro-interleukin-1beta is cytosolic and precedes cell death. J Cell Sci. 2007;120(Pt 5):772–781.
    1. Lopez-Castejon G, Brough D. Understanding the mechanism of IL-1β secretion. Cytokine Growth Factor Rev. 2011;22(4):189–195. doi: 10.1016/j.cytogfr.2011.10.001.
    1. Schroder K, et al. The NLRP3 inflammasome: a sensor for metabolic danger? Science. 2010;327(5963):296–300. doi: 10.1126/science.1184003.
    1. Coeshott C, et al. Converting enzyme-independent release of tumor necrosis factor alpha and IL-1beta from a stimulated human monocytic cell line in the presence of activated neutrophils or purified proteinase 3. Proc Natl Acad Sci U S A. 1999;96(11):6261–6266. doi: 10.1073/pnas.96.11.6261.
    1. Sugawara S, et al. Neutrophil proteinase 3-mediated induction of bioactive IL-18 secretion by human oral epithelial cells. J Immunol. 2001;167(11):6568–6575. doi: 10.4049/jimmunol.167.11.6568.
    1. Netea MG, et al. Inflammasome-independent regulation of IL-1-family cytokines. Annu Rev Immunol. 2015;33:49–77. doi: 10.1146/annurev-immunol-032414-112306.
    1. Bryan NB, et al. Activation of inflammasomes requires intracellular redistribution of the apoptotic speck-like protein containing a caspase recruitment domain. J Immunol. 2009;182(5):3173–3182. doi: 10.4049/jimmunol.0802367.
    1. de Almeida L, et al. The PYRIN domain-only protein POP1 inhibits inflammasome assembly and ameliorates inflammatory disease. Immunity. 2015;43(2):264–276. doi: 10.1016/j.immuni.2015.07.018.
    1. Dorfleutner A, Stehlik C. A dRAStic RHOAdblock of Pyrin inflammasome activation. Nat Immunol. 2016;17(8):900–902. doi: 10.1038/ni.3511.
    1. Khare S, et al. An NLRP7-containing inflammasome mediates recognition of microbial lipopeptides in human macrophages. Immunity. 2012;36(3):464–476. doi: 10.1016/j.immuni.2012.02.001.
    1. Khare S, et al. The PYRIN domain-only protein POP3 inhibits ALR inflammasomes and regulates responses to infection with DNA viruses. Nat Immunol. 2014;15(4):343–353. doi: 10.1038/ni.2829.
    1. Radian AD, et al. ATP binding by NLRP7 is required for inflammasome activation in response to bacterial lipopeptides. Mol Immunol. 2015;67(2 Pt B):294–302.
    1. Ratsimandresy RA, et al. The PYRIN domain-only protein POP2 inhibits inflammasome priming and activation. Nat Commun. 2017;8:15556.
    1. Ratsimandresy RA, et al. The AIM2 inflammasome is a central regulator of intestinal homeostasis through the IL-18/IL-22/STAT3 pathway. Cell Mol Immunol. 2017;14(1):127–142. doi: 10.1038/cmi.2016.35.
    1. Stehlik C, Dorfleutner A. COPs and POPs: modulators of inflammasome activity. J Immunol. 2007;179(12):7993–7998. doi: 10.4049/jimmunol.179.12.7993.
    1. Ruohonen ST, et al. Potentiation of glibenclamide hypoglycaemia in mice by MK-467, a peripherally acting Alpha2-adrenoceptor antagonist. Basic Clin Pharmacol Toxicol. 2015;117(6):392–398. doi: 10.1111/bcpt.12440.
    1. Koh GC, et al. Glyburide reduces bacterial dissemination in a mouse model of melioidosis. PLoS Negl Trop Dis. 2013;7(10):2500.
    1. Liu L, et al. The pathogenic role of NLRP3 inflammasome activation in inflammatory bowel diseases of both mice and humans. J Crohns Colitis. 2017;11(6):737–750.
    1. Van Rooijen N, Sanders A. Liposome mediated depletion of macrophages: mechanism of action, preparation of liposomes and applications. J Immunol Methods. 1994;174(1–2):83–93.
    1. Halle S, et al. Induced bronchus-associated lymphoid tissue serves as a general priming site for T cells and is maintained by dendritic cells. J Exp Med. 2009;206(12):2593–2601. doi: 10.1084/jem.20091472.
    1. Gelman AE, et al. Cutting edge: acute lung allograft rejection is independent of secondary lymphoid organs. J Immunol. 2009;182(7):3969–3973. doi: 10.4049/jimmunol.0803514.
    1. Kreisel D, et al. In vivo two-photon imaging reveals monocyte-dependent neutrophil extravasation during pulmonary inflammation. Proc Natl Acad Sci U S A. 2010;107(42):18073–18078. doi: 10.1073/pnas.1008737107.
    1. Satija R, et al. Spatial reconstruction of single-cell gene expression data. Nat Biotechnol. 2015;33(5):495–502. doi: 10.1038/nbt.3192.
    1. Aran D, et al. Reference-based analysis of lung single-cell sequencing reveals a transitional profibrotic macrophage. Nat Immunol. 2019;20(2):163–172. doi: 10.1038/s41590-018-0276-y.

Source: PubMed

3
Suscribir