Interacting chemokine signals regulate dendritic cells in acute brain injury

Charlotte Israelsson, Annika Kylberg, Henrik Bengtsson, Lars Hillered, Ted Ebendal, Charlotte Israelsson, Annika Kylberg, Henrik Bengtsson, Lars Hillered, Ted Ebendal

Abstract

Brain trauma is known to activate inflammatory cells via various chemokine signals although their interactions remain to be characterized. Mice deficient in Ccl3, Ccr2 or Cxcl10 were compared with wildtype mice after controlled cortical impact injury. Expression of Ccl3 in wildtypes was rapidly upregulated in resident, regularly spaced reactive microglia. Ccl3-deficiency enhanced endothelial expression of platelet selectin and invasion of peripheral inflammatory cells. Appearance of Ccr2 transcripts, encoding the Ccl2 receptor, reflected invasion of lysozyme 2-expressing phagocytes and classical antigen-presenting dendritic cells expressing major histocompatibility complex class II. Ccr2 also directed clustered plasmacytoid dendritic cells positive for the T-cell attracting chemokine Cxcl10. A reduction in Ccr2 and dendritic cells was found in injured wildtype cortex after cyclophosphamide treatment resembling effects of Ccr2-deficiency. The findings demonstrate the feasibility to control inflammation in the injured brain by regulating chemokine-dependent pathways.

Conflict of interest statement

Competing Interests: The authors have declared that no competing interests exist.

Figures

Figure 1. Traumatic brain injury (TBI) in…
Figure 1. Traumatic brain injury (TBI) in wildtype (wt) and chemokine-deficient (Ccl3−/− and Ccr2−/−) mice.
(a) In situ hybridization revealed intense Ccl3 expression in the perilesional zone four hours after trauma, in a pattern resembling distribution of activated microglial cells. (b) Temporal patterns of Ccl3 and Ccr2 transcript levels in the injured wt neocortex revealed by qRT-PCR. (c) The Ccr2 transcript was further upregulated in Ccl3−/− mice as compared to wt mice three days postinjury. Gfap expression was also strongly upregulated but with no differences between the strains. (d) The Ccl2 transcript, encoding a Ccr2 ligand, was upregulated after injury with no differences between wt and Ccl3−/− mice as shown by microarray analysis. In contrast, Selp transcript expressed by endothelial cells was more upregulated in Ccl3−/− compared to wt brains. (e) Lyz2 expression and the microglial surface marker isolectin B4 (IB4) showed only partial overlap three days post-injury in wt brains (in situ hybridization and histochemistry). The Bst2, characterizing pDCs, exhibited a different expression pattern (insert). (f) At three days postinjury, the Lyz2 increase shifted in opposite directions in the Ccl3−/− and Ccr2−/− compared to wt brains. (g) Cavity volume was reduced in the Ccr2−/− compared to wt mice seven days after injury in accordance with downregulation of inflammatory Lyz2-response.
Figure 2. Interactions among chemokine transcripts analyzed…
Figure 2. Interactions among chemokine transcripts analyzed with qRT-PCR and in situ hybridization in neocortex three days after TBI.
(a) The Ccl3 transcript expression did not differ between Ccr2−/− and wt brains. (b) Compared to wt mice, Cxcl10 transcript showed opposite changes in expression in Ccl3−/− and Ccr2−/− brains. (c) A clustered cell pattern was seen for the Cxcl10 transcript by in situ hybridization in all injured brains, although with different intensities depending on genotype. Expression was stronger in the injured Ccl3−/− mice and weaker in the Ccr2−/− mice compared to wt brains. (d) Ccl3 and Ccr2 upregulation showed no differences in wt compared to Cxcl10−/− injured brains by qRT-PCR. (e) Suggested model of interactions among chemokine ligands Ccl3 and Cxcl10 and the chemokine receptor Ccr2 three days after brain injury.
Figure 3. Inflammatory cells in the injured…
Figure 3. Inflammatory cells in the injured wt and Ccr2−/− cortex examined by flow cytometry and qRT-PCR.
(a) Cells detected with FITC-CD45 in combination with either APC-PDCA-1 (Bst2) or APC-Cd11c (Itgax) in neocortex of wt or Ccr2−/− mice three days after injury. Upper panel left shows gating for Cd11c. The middle and right panels show wt and Ccr2−/− results, respectively, demonstrating reduced number of Cd11c-positive cells in the Ccr2−/− mice. Lower panel shows that PDCA-1 positive cells were reduced in Ccr2−/− brains. (b) Quantitative flow cytometry data from wt and Ccr2−/− mice three days postinjury. Isotype controls showed only trace signals. (c) Temporal expression patterns of six inflammatory-related transcripts (Itgax, H2-Aa, Bst2, Cxcl10, Lyz2 and Hmox1) after injury in wt and Ccr2−/− brains.
Figure 4. Inflammatory-related transcripts measured in Ccl3…
Figure 4. Inflammatory-related transcripts measured in Ccl3−/− and Ccr2−/− mice with F2 hybrid background three days postinjury.
Lyz2 expression was upregulated in homozygous Ccl3 knockouts but downregulated in Ccr2−/− as in the parental strains, as was Bst2. Both Itgax and Cxcl10 levels were less upregulated in the injured F2 Ccr2−/− brains. Gfap expression increased to the same extent in injured wt and F2 hybrid brains.
Figure 5. Cell sorting and immunodepletion of…
Figure 5. Cell sorting and immunodepletion of dendritic cells in neocortex three days after TBI in wt mice analyzed by qRT-PCR.
(a) Cd11c-positive cells sorted on magnetic microbeads expressed enhanced H2-Aa and Itgax levels. In contrast, Bst2 was enriched in cells sorted on anti-PDCA-1 microbeads. (b) Cortical levels of inflammatory transcripts in mice injected with control immunoglobulin or with antibodies directed to PDCA-1. Depletion of pDCs was not accompanied by shifts in Itgax or H2-Aa transcript levels whereas a distinct reduction of Bst2 and Cxcl10 occurred.
Figure 6. Cyclophosphamide treatment in wt mice…
Figure 6. Cyclophosphamide treatment in wt mice analyzed three days after TBI.
(a) Microarray showed markedly weakened injury-activated Ccr2 and H2-Aa levels while upregulation of the fractalkine receptor Cx3cr1 was not affected. (b) Itgax was also dampened in mice subjected to cyclophosphamide treatment as shown by qRT-PCR whereas Ccl3 upregulation was not impaired. (c) Flow cytometry demonstrated reduced number of brain cells gated for Cd11c in mice treated with cyclophosphamide. Isotype controls (upper middle panel) showed only trace labeling of inflammatory cells. Quantitative data from flow cytometry assays (right), show a substantial reduction of Cd11c-positive cDCs in the injured brains treated with cyclophosphamide compared to mice injected with saline.

References

    1. Israelsson C, Bengtsson H, Kylberg A, Kullander K, Lewén A, et al. (2008) Distinct cellular patterns of upregulated chemokine expression supporting a prominent inflammatory role in traumatic brain injury. J Neurotrauma 25: 959–974.
    1. Israelsson C, Bengtsson H, Lobell A, Nilsson LNG, Kylberg A, et al. (2010) Appearance of Cxcl10-expressing cell clusters is common for traumatic brain injury and neurodegenerative disorders. Eur J Neurosci 31: 852–863.
    1. Nimmerjahn A, Kirchhoff F, Helmchen F (2005) Resting microglial cells are highly dynamic surveillants of brain parenchyma in vivo. Science 308: 1314–1318.
    1. Steinman RM, Banchereau J (2007) Taking dendritic cells into medicine. Nature 449: 419–426).
    1. Charo IF, Ransohoff RM (2006) The many roles of chemokines and chemokine receptors in inflammation. N Engl J Med 354: 610–621.
    1. Finnie JW (2013) Neuroinflammation: beneficial and detrimental effects after traumatic brain injury. Inflammopharmacology 21: 309–320.
    1. Smith DH, Soares HD, Pierce JS, Perlman KG, Saatman KE, et al. (1995) A model of parasagittal controlled cortical impact in the mouse: cognitive and histopathologic effects. J Neurotrauma 12: 169–178.
    1. Laurer HL, McIntosh TK (1999) Experimental models of brain trauma. Curr Opin Neurol 12: 715–721.
    1. Cook DN, Beck MA, Coffman TM, Kirby SL, Sheridan JF, et al. (1995) Requirement of MIP-1 alpha for an inflammatory response to viral infection. Science 269: 1583–1585.
    1. Kuziel WA, Morgan SJ, Dawson TC, Griffin S, Smithies O, et al. (1997) Severe reduction in leukocyte adhesion and monocyte extravasation in mice deficient in CC chemokine receptor 2. Proc Natl Acad Sci U S A 94: 12053–12058.
    1. Blasius AL, Giurisato E, Cella M, Schreiber RD, Shaw AS, et al. (2006) Bone marrow stromal cell antigen 2 is a specific marker of type I IFN-producing cells in the naive mouse, but a promiscuous cell surface antigen following IFN stimulation. J Immunol 177: 3260–3265.
    1. Dufour JH, Dziejman M, Liu MT, Leung JH, Lane TE, et al. (2002) IFN-gamma-inducible protein 10 (IP-10; CXCL10)-deficient mice reveal a role for IP-10 in effector T cell generation and trafficking. J Immunol 168: 3195–3204.
    1. Geissmann F, Manz MG, Jung S, Sieweke MH, Merad M, et al. (2010) Development of monocytes, macrophages, and dendritic cells. Science 327: 656–661.
    1. Auffray C, Sieweke MH, Geissmann F (2009) Blood monocytes: development, heterogeneity, and relationship with dendritic cells. Annu Rev Immunol 27: 669–692.
    1. Ajami B, Bennett JL, Krieger C, McNagny KM, Rossi FM (2011) Infiltrating monocytes trigger EAE progression, but do not contribute to the resident microglia pool. Nat Neurosci 14: 1142–1149.
    1. Reizis B, Bunin A, Ghosh HS, Lewis KL, Sisirak V (2011) Plasmacytoid dendritic cells: recent progress and open questions. Annu Rev Immunol 29: 163–183.
    1. Schwartz M, Shechter R (2010) Systemic inflammatory cells fight off neurodegenerative disease. Nat Rev Neurol 6: 405–410.
    1. Aguzzi A, Barres BA, Bennett ML (2013) Microglia: scapegoat, saboteur, or something else? Science 339: 156–161.
    1. Prinz M, Priller J, Sisodia SS, Ransohoff RM (2011) Heterogeneity of CNS myeloid cells and their roles in neurodegeneration. Nat Neurosci 14: 1227–1235.
    1. Goldmann T, Wieghofer P, Müller PF, Wolf Y, Varol D, et al. (2013) A new type of microglia gene targeting shows TAK1 to be pivotal in CNS autoimmune inflammation. Nat Neurosci 16: 1618–1626.
    1. Miller JC, Brown BD, Shay T, Gautier EL, Jojic V, et al. (2012) Deciphering the transcriptional network of the dendritic cell lineage. Nat Immunol 13: 888–899.
    1. Steinman RM (2012) Decisions about dendritic cells: past, present, and future. Annu Rev Immunol 30: 1–22.
    1. Colton CA (2013) Immune heterogeneity in neuroinflammation: dendritic cells in the brain. J Neuroimmune Pharmacol 8: 145–162.
    1. Greter M, Heppner FL, Lemos MP, Odermatt BM, Goebels N, et al. (2005) Dendritic cells permit immune invasion of the CNS in an animal model of multiple sclerosis. Nat Med 11: 328–334.
    1. Colonna M, Trinchieri G, Liu YJ (2004) Plasmacytoid dendritic cells in immunity. Nat Immunol 5: 1219–1226.
    1. Megjugorac NJ, Young HA, Amrute SB, Olshalsky SL, Fitzgerald-Bocarsly P (2004) Virally stimulated plasmacytoid dendritic cells produce chemokines and induce migration of T and NK cells. J Leukoc Biol 75: 504–514.
    1. Bendriss-Vermare N, Burg S, Kanzler H, Chaperot L, Duhen T, et al. (2005) Virus overrides the propensity of human CD40L-activated plasmacytoid dendritic cells to produce Th2 mediators through synergistic induction of IFN-{gamma} and Th1 chemokine production. J Leukoc Biol 78: 954–966.
    1. Saederup N, Cardona AE, Croft K, Mizutani M, Cotleur AC, et al. (2010) Selective chemokine receptor usage by central nervous system myeloid cells in CCR2-red fluorescent protein knock-in mice. PLoS One 5: e13693.
    1. Mildner A, Mack M, Schmidt H, Brück W, Djukic M, et al. (2009) CCR2+Ly-6Chi monocytes are crucial for the effector phase of autoimmunity in the central nervous system. Brain 132: 2487–2500.
    1. Liu S, Zhang L, Wu Q, Wu Q, Wang T (2013) Chemokine CCL2 Induces Apoptosis in Cortex Following Traumatic Brain Injury. J Mol Neurosci 51: 1021–1029.
    1. Clausen BE, Burkhardt C, Reith W, Renkawitz R, Förster I (1999) Conditional gene targeting in macrophages and granulocytes using LysMcre mice. Transgenic Res 8: 265–277.
    1. Dimitrijevic OB, Stamatovic SM, Keep RF, Andjelkovic AV (2007) Absence of the chemokine receptor CCR2 protects against cerebral ischemia/reperfusion injury in mice. Stroke 38: 1345–1353.
    1. Huyan XH, Lin YP, Gao T, Chen RY, Fan YM (2011) Immunosuppressive effect of cyclophosphamide on white blood cells and lymphocyte subpopulations from peripheral blood of Balb/c mice. Int Immunopharmacol 11: 1293–1297.
    1. Mestas J, Hughes CCW (2004) Of mice and not men: differences between mouse and human immunology. J Immunol 172: 2731–2738.
    1. Jenkins SJ, Ruckerl D, Cook PC, Jones LH, Finkelman FD, et al. (2011) Local macrophage proliferation, rather than recruitment from the blood, is a signature of TH2 inflammation. Science 332: 1284–1288.
    1. Murray PJ, Wynn TA (2011) Protective and pathogenic functions of macrophage subsets. Nat Rev Immunol 11: 723–737.
    1. Seok J, Warren HS, Cuenca AG, Mindrinos MN, Baker HV, et al. (2013) Genomic responses in mouse models poorly mimic human inflammatory diseases. Proc Natl Acad Sci U S A 110: 3507–3512.
    1. Pan J, Xia L, McEver RP (1998) Comparison of promoters for the murine and human P-selectin genes suggests species-specific and conserved mechanisms for transcriptional regulation in endothelial cells. J Biol Chem 273: 10058–10067.
    1. Pashenkov M, Huang Y-M, Kostulas V, Haglund M, Söderström M, et al. (2001) Two subsets of dendritic cells are present in human cerebrospinal fluid. Brain 124: 480–492.
    1. Stefini R, Catenacci E, Piva S, Sozzani S, Valerio A, et al. (2008) Chemokine detection in the cerebral tissue of patients with posttraumatic brain contusions. J Neurosurg 108: 958–962.
    1. Helmy A, Carpenter KL, Menon DK, Pickard JD, Hutchinson PJA (2011) The cytokine response to human traumatic brain injury: temporal profiles and evidence for cerebral parenchymal production. J Cereb Blood Flow Metab 31: 658–670.
    1. Helmy A, Antoniades CA, Guilfoyle MR, Carpenter KL, Hutchinson PJ (2012) Principal component analysis of the cytokine and chemokine response to human traumatic brain injury. PLoS One 7: e39677.
    1. Semple BD, Bye N, Rancan M, Ziebell JM, Morganti-Kossmann MC (2010) Role of CCL2 (MCP-1) in traumatic brain injury (TBI): evidence from severe TBI patients and CCL2−/− mice. J Cereb Blood Flow Metab 30: 769–782.
    1. Ho L, Zhao W, Dams-O'Connor K, Tang CY, Gordon W, et al. (2012) Elevated plasma MCP-1 concentration following traumatic brain injury as a potential “predisposition” factor associated with an increased risk for subsequent development of Alzheimer's disease. J Alzheimers Dis 31: 301–313.
    1. Lasky-Su J, Himes BE, Raby BA, Klanderman BJ, Senter Sylvia J, et al. (2012) HLA-DQ strikes again: genome-wide association study further confirms HLA-DQ in the diagnosis of asthma among adults. Clin Exp Allergy 42: 1724–1733.
    1. Zhu WH, Lu J-H, Lin J, Xi J-Y, Lu J, et al. (2012) HLA-DQA1*03:02/DQB1*03:03:02 is strongly associated with susceptibility to childhood-onset ocular myasthenia gravis in Southern Han Chinese. J Neuroimmunol 247: 81–85.
    1. Megiorni F, Pizzuti A (2012) HLA-DQA1 and HLA-DQB1 in Celiac disease predisposition: practical implications of the HLA molecular typing. J Biomed Sci 19: 88.

Source: PubMed

3
Abonnieren