Articles

  • KSBNS 2024

Article

Original Article

Exp Neurobiol 2012; 21(4): 151-157

Published online December 30, 2012

https://doi.org/10.5607/en.2012.21.4.151

© The Korean Society for Brain and Neural Sciences

Role of TGF-β in Survival of Phagocytizing Microglia: Autocrine Suppression of TNF-α Production and Oxidative Stress

Keun-Young Ryu1, Geum-Sil Cho2, Hua Zi Piao3 and Won-Ki Kim2*

1College of Medicine, Korea University, Seoul 136-705, 2Department of Neuroscience, College of Medicine, Korea University, Seoul 136-705, Korea, 3Department of Pharmacology, College of Medicine, Yanbian University, Yanji 133002, China

Correspondence to: *To whom correspondence should be addressed.
TEL: 82-2-920-6094, FAX: 82-2-953-6095
e-mail: wonki@korea.ac.kr

Microglia are recognized as residential macrophageal cells in the brain. Activated microglia play a critical role in removal of dead or damaged cells through phagocytosis activity. During phagocytosis, however, microglia should survive under the harmful condition of self-producing ROS and pro-inflammatory mediators. TGF-β has been known as a classic anti-inflammatory cytokine and controls both initiation and resolution of inflammation by counter-acting inflammatory cytokines. In the present study, to understand the self-protective mechanism, we studied time-dependent change of TNF-α and TGF-β production in microglia phagocytizing opsonized-beads (i.e., polystyrene microspheres). We found that microglia phagocytized opsonized-bead in a time-dependent manner and simultaneously produced both TNF-α and TGF-β. However, while TNF-α production gradually decreased after 6 h, TGF-β production remained at increased level. Microglial cells pre-treated with lipopolysaccharides (a strong immunostimulant, LPS) synergistically increased the production of TNF-α and TGF-β both. However, LPS-pretreated microglia produced TNF-α in a more sustained manner and became more vulnerable, probably due to the marked and sustained production of TNF-α and reduced TGF-β. Intracellular oxidative stress appears to change in parallel with the microglial production of TNF-α. These results indicate TGF-β contributes for the survival of phagocytizing microglia through autocrine suppression of TNF-α production and oxidative stress.

Keywords: microglia, LPS, phagocytosis, TNF-α, TGF-β, ROS

Macrophages play an important role in the innate and adaptive immune responses, and produce inflammatory mediators during infection. Inflammatory reaction is necessary to pathogen removal, but sustained inflammation can damage the tissue [1]. Microglial cells are recognized as residential macrophages in the CNS and involved in pathogen removal and inflammatory responses depending on pathophysiological conditions [2]. When microglial cells are activated, phagocytosis occurs together with the release of cytokines including tumor necrosis factor (TNF), interleukin-1β (IL-1β) and other inflammatory mediators such as reactive oxygen species and nitric oxide (NO) [2-4]. In contrast, when removing apoptotic cells or myelin debris, microglia release anti-inflammatory factors such as IL-10, TGF-β, prostaglandin E2, and platelet-activating factor (PAF) [5, 6]. However, uptake of apoptotic cells by microglia down-regulate pro-inflammatory cytokines such as TNF-α, IL-12, IL-1β and other inflammatory mediators [6, 7].

TGF-β is a regulatory molecule with diverse effects on cell proliferation, differentiation, migration, and survival that affect multiple biological processes, including development, carcinogenesis, fibrosis, wound healing, and immune responses [8, 9]. Especially, the critical role of TGF-β in the immune system is to maintain tolerance via the regulation of lymphocyte proliferation, differentiation, and survival. However, TGF-β controls both initiation and resolution of inflammation by regulating cell differentiation and inflammatory cytokines and costimulatory molecules [10, 11]. It seems that TGF-β acts initially as pro-inflammatory cytokines, but when inflammation is ongoing with other proinflammatory mediators, TGF-β acts as anti-inflammatory cytokines [9, 12]. It has been reported that phagocytosis of apoptotic cells leads to TGF-β secretion in macrophage, which inhibits the production of inflammatory cytokines and chemokines including IL-1β, TNF-α, GM-CSF, and IL-8 [6, 13]. In vitro, TGF-β inhibits the expression of several LPS-induced inflammatory mediators such as TNF-α and MMP-12 as well as chemokines including MIP-1α and MIP-2 [14, 15]. Reactive oxygen and nitrogen species produced by activated macrophages are important mediators against invading microbes. TGF-β downregulates the production of nitric oxide (NO) and superoxide ion and inhibits the expression of inducible NO synthase (iNOS) in activated macrophages [16]. Interestingly, however, phagocytizing microglia survive despite of the marked production of pro-inflammatory cytokine and reactive oxygen or nitrogen species. In the present study, we investigated the autocrine regulation those harmful mediators in phagocytizing microglia.

Materials

Minimum Essential Medium (MEM), trypsin/EDTA, penicillin/streptomycin and fetal bovine serum (FBS) were purchased from Gibco BRL (Grand Island, NY, USA). Polystyrene microspheres (PS) (1 µm,diam) (Polysciences Inc.), Bovine serum albumin (BSA.), Rabbit Anti-bovine serum albumin antibody (ICN) and LPS were purchased from Sigma. TNF-α and TGF-β ELISA kits were purchased from R&D Systems (Minneapolis, USA). ED-1 and GFAP were purchased from BD Pharmingen.

Cell cultures

Primary culture of mixed glial and microglia were prepared as previously described [17]. In brief, cells were cultured from the prefrontal cortices of 1- to 2.day-old Sprague-Dawley rat pups. Mixed glial cells were dissociated by mild trypsinization (for 8 min at 37℃, with 0.1% trypsin-containing HBSS) and passed through sterile nylon sieves (130 µm, pore size) into MEM (Minimum Essential Medium) containing 10% heat-inactivated FBS, penicillin and streptomycin 10 ml. They were then plated onto poly-D-lysine (2 µg/ml)-coated 75-cm2 culture bottles and maintained in MEM supplemented with 10% FBS (7-8 days in culture). The mixed glial cells were then trypsinized, washed, and plated in growth medium onto poly-L-lysine-coated 48-well plates. Microglia-enriched cultures were prepared from primary mixed glial cell cultures as described [18]. To obtain microglial cells, the flasks were shaken at 200 r.p.m. for 1 min at 37℃ on day 8 of the mixed glia cultures. The medium containing detached microglial cells was collected and centrifuged at 2000 r.p.m. for 200 s. The medium after centrifuge was taken as microglial conditioned medium. Cells were res-uspended, counted and plated at a density of 1.05×106 cells/mm2 in 48- or 6-well plates in conditioned medium. After 12 h, microglia were used for experiments.

Phagocytosis assay

To opsonize particles with anti-BSA antibody, particles were initially associated with BSA [19]. To allow nonspecific adsorption of BSA onto particles, polystyrene microspheres (PS) (diameter=1 µm) were incubated with 10 mg/ml BSA for 1 h at 37℃ and washed by centrifugation. To conjugate BSA to PS, PS were incubation with 0.5% glutaraldehyde overnight, then washed by centrifugation, incubated with BSA 10 mg/ml for 2 h at room temperature. Particles with associated albumin were opsonized for 30 min with rabbit anti-BSA antibodies in PBS with a dilution ratio of 1:25. The PS washed with PBS and used in phagocytosis assay.

To study phagocytotic activity, cells were incubated with opsonized-bead at 37℃ for 3 , 6 , 9 , 12 and 24 h and then washed with PBS thrice to remove serum and unbound beads [20]. Then cells were lysed by DNA fragment lysis buffer containing 10 mM Tris-HCl, 10 mM EDTA, 0.5% Triton X .100, and 0.2% SDS [21]. The beads were washed twice with PBS, and the absorbance at 550 nm was determined using a microtiter plate reader (Molecular Devices, USA).

Determination of TNF-α and TGF-β

TNF-α and TGF-β levels were determined by ELISA kit (BioSource Camarillo, CA) [22]. To analyze TNF-α and TGF-β levels, 3×105/ml microglia were incubated overnight. After a washing step with 1% FBS medium, beads were added to each well for various times and then supernatants were collected for determination of TNF-α and TGF-β levels. The sensitivity of the assay was 15.6 pg/ml.

Immunocytochemical staining

Cells grown on 48 well plates were fixed for 10 min at room temperature with 100% ethanol [23]. To inactivate endogenous peroxidase, cells were incubated with PBS containing 3% H2O2 for 5 min at room temperature. Nonspecific staining was blocked by incubating cells with PBS containing 8% bovine serum albumin (BSA) for 30 min at room temperature. Cells were then incubated with primary antibody for 2 h at room temperature. The following primary antibodies were used: anti-GFAP Ab at a dilution of 1:500; anti-ED-1 Ab at a dilution of 1:250. After washing extensively with PBS, cells were probed for 1 h at room temperature with anti-mouse horseradish-peroxidase-conjugated secondary antibody at a dilution of 1:200. The immunoreactivity was visualized with diaminobenzidine in the presence of 0.1% H2O2. For negative controls the first antibodies were omitted.

Determination of cell viability: lactate dehydrogenase (LDH) release

Cell injury or death was assessed by morphological examination using a phase-contrast microscope (DM IL, Leica, Germany) or by measuring the amount of lactate dehydrogenase (LDH) released into the culture medium employing a diagnostic kit (Sigma-Aldrich, St. Louis, MO), as previously described [24]. Absorbance readings were measured at 450 nm using a SpectraMax microplate reader (Molecular Devices, Sunnyvale, CA) and are expressed as the % of total LDH release, which was derived from sister cultures subjected to repeated freeze/thaw cycles.

Measurement of intracellular oxidative stress: DCF-DA assay

Cells were loaded with 30 µM CM-H2DCFDA. CM-H2DCFDA diffuses through cell membranes and is hydrolyzed by intracellular esterases to the nonfluorescent analog dichlorofluorescein (DCFH). DCFH then reacts with intracellular free radicals such as peroxyl radical, peroxynitrite, or hydrogen peroxide to form dichlorofluorescin (DCF), a green fluorescent dye. Two hours after loading, cells were washed with EBSS buffer containing 0.1% BSA and 2.5 mM probenecid. Fluorescent intensities were then measured at 488 nm of excitation wavelength and 525 nm of emission wavelength employing a fluorescence microscope (DM IL HC Fluo, Leica, Germany) equipped with a digital camera (DFC420C, Leica, Germany). The intensity of fluorescence was quantified by an image analyzer (TOMORO ScopeEye 3.5, Seoul, Korea).

Statistical analysis

Data are expressed as the mean±standard deviation (s.d) and analyzed for statistical significance by using one-way ANOVA followed by Scheffe's test for multiple comparisons. A p value <0.05 was considered significant.

First, we observed the time-dependent phagocytosis of opsonized beads in cultured mixed glial cells (Fig. 1). Opsonized beads were rapidly phagocytized by glial cells, especially microglial cells, which are immunopositive to ED-1 (Fig. 1A, b), but not by GFAP-immunopositive astroglial cells (Fig. 1A, c). Initially, bead uptake by microglial cells occurred rapidly, and gradually increased later (Fig. 1B).

We further determined the production of TNF-α and TGF-β in microglia phagocytizing beads (diameter=1 µm). Regardless of opsonization, phagocytosis of beads stimulated microglial production of TNF-α and TGF-β (Fig. 2). While the level of TNF-α production decreased gradually after 6 h, TGF-β release remained high (Fig. 2A, B). However, opsonized bead increased much more production of TNF-α and TGF-β in microglia (Fig. 2C, D). However, there was no synergistic increase of TNF-α and TGF-β production between oponized bead and LPS treatment (Fig. 2C, D). While the level of TNF-α production (Fig. 2A) remained high, TGF-β (Fig. 2B) release was gradually decreased (Fig. 2C, D).

Since bead-activated microglia release TGF-β and down-regulate production of TNF-α (Fig. 2), we neutralized TGF-β by using anti-TGF-β Ab to study whether down-regulation of TNF-α was caused by co-produced TGF-β (Fig. 3). Expectedly, neutralization of TGF-β using anti-TGF-β Ab augmented the LDH release, TNF-α production and intracellular oxidative stress in bead-activated microglia.

In the central nervous system, resident microglial cells are major inflammatory cells responding to various kinds of brain injury. Activated microglia potentiates damage to BBB integrity and exert neurotoxic functions through the production of reactive oxygen species (ROS), cytokines (IL-1β, IL-6, TNF-α), and MMP-9. Activated microglia are also known to remove dead cells or other debris in brain lesions. In turn, however, microglial cells can be damaged by those biologically active molecules produced by themselves. To understand the self-protective mechanism, in the present study we studied the temporal changes of production of pro- and anti-inflammatory cytokines as well as ROS in phagocytizing microglia.

TNF-α is a pro-inflammatory cytokine mediating an array of inflammatory response, including activation of leukocytes, production of cytokines, chemokines and adhesion molecules, and stimulation of proteases in neutrophils and endothelial cells [25]. TGF-β is generally thought to have anti-inflammatory effects, but it may have both pro- and anti-inflammatory effects depending on the context in which it is acting [9]. Previous reports also showed that TGF-β might play an important role in early steps of inflammation by inducing monocytes to improve their survival, migration and stimulatory capacity and by attracting both CD4+ and CD8+ T cells to sites of inflammation [26, 27].

When phagocytosis of opsonized-bead induced microglial activation in our experiments (Fig. 2A, B), TGF-β as well as TNF-α are simultaneously released, demonstrating that TGF-β participates in early steps of inflammation. TNF-α production increased steadily at first, but it decreased 6 h later, whereas TGF-β level lasted high for longer than 1 d (Fig. 2A, B). From these results, we can speculate that autocrine secretion of TGF-β from activated microglia results in a gradual decrease of inflammatory TNF-α production. It also consistent with the result that direct addition of TGF-β to LPS-stimulated macrophages partially inhibited both LDH release (our unpublished results) and production of pro-inflammatory cytokines such as TNF-α, IL-1β, IL-8 and GM-CSF, thereby proving paracrine and autocrine inhibitory effect of TGF-β [6]. We also observed that when LPS-stimulated microglia phagocytized opsonized-bead, microglial production of TNF-α was up-regulated, but production of TGF-β was down-regulated (Fig. 2, C and D). From this result, we speculate that phagocytosis of opsonized-bead influences microglial state in a favor of pro-inflammatory state.

TGF-β plays a role in resolution of inflammation not only by acting as an anti-inflammatory cytokine as afore-mentioned, but also by promoting healing and repairing of damaged tissue/cell during inflammation [28]. The abilities of TGF-β for promotion of healing [29] make TGF-β ligands as potential therapeutic agents in several disease such as cutaneous wounds [30], mucositis [31, 32], bone defect [25] and stroke [33, 34]. Especially in stroke, administration of TGF-β into the brain reduced infarct size in experimental animal models of ischemia, while antagonizing the endogenous actions of TGF-β with an injection of a soluble TGF-β type II receptor resulted in a drastic increase in infarct area [35]. Considering anti-inflammatory/cytoprotective activities against TNF-α and anti-oxidant activities, TGF-β would be of great therapeutic benefit to inflammation-related diseases such as cerebral stroke, Parkinson's disease and others.

  1. Porcheray F, Viaud S, Rimaniol AC, Léone C, Samah B, Dereuddre-Bosquet N, Dormont D, Gras G. Macrophage activation switching: an asset for the resolution of inflammation. Clin Exp Immunol 2005;142:481-489.
    Pubmed
  2. Hanisch UK, Kettenmann H. Microglia: active sensor and versatile effector cells in the normal and pathologic brain. Nat Neurosci 2007;10:1387-1394.
    Pubmed
  3. Nathan C. Points of control in inflammation. Nature 2002;420:846-852.
    Pubmed
  4. Sell S. Inflammation and wound healing. Immunology, immunopathology and immunity. 6th ed. Herndon, VA: American Society for Microbiology Press, 2001; 2001. p. 33-96.
  5. Liu Y, Hao W, Letiembre M, Walter S, Kulanga M, Neumann H, Fassbender K. Suppression of microglial inflammatory activity by myelin phagocytosis: role of p47-PHOX-mediated generation of reactive oxygen species. J Neurosci 2006;26:12904-12913.
    Pubmed
  6. Fadok VA, Bratton DL, Konowal A, Freed PW, Westcott JY, Henson PM. Macrophages that have ingested apoptotic cells in vitro inhibit proinflammatory cytokine production through autocrine/paracrine mechanisms involving TGF-beta, PGE2, and PAF. J Clin Invest 1998;101:890-898.
    Pubmed
  7. Magnus T, Chan A, Grauer O, Toyka KV, Gold R. Microglial phagocytosis of apoptotic inflammatory T cells leads to down-regulation of microglial immune activation. J Immunol 2001;167:5004-5010.
    Pubmed
  8. Blobe GC, Schiemann WP, Lodish HF. Role of transforming growth factor β in human disease. N Engl J Med 2000;342:1350-1358.
    Pubmed
  9. Worthington JJ, Fenton TM, Czajkowska BI, Klementowicz JE, Travis MA. Regulation of TGFβ in the immune system: An emerging role for integrins and dendritic cells. Immunobiology 2012;217:1259-1265.
    Pubmed
  10. Letterio JJ, Roberts AB. Regulation of immune responses by TGF-β. Annu Rev Immunol 1998;16:137-161.
    Pubmed
  11. Wahl SM. Transforming growth factor β: the good, the bad, and the ugly. J Exp Med 1994;180:1587-1590.
    Pubmed
  12. Li MO, Wan YY, Sanjabi S, Robertson AK, Flavell RA. Transforming growth factor-β regulation of immune responses. Annu Rev Immunol 2006;24:99-146.
    Pubmed
  13. McDonald PP, Fadok VA, Bratton D, Henson PM. Transcriptional and translational regulation of inflammatory mediator production by endogenous TGF-β in macrophages that have ingested apoptotic cells. J Immunol 1999;163:6164-6172.
    Pubmed
  14. Bogdan C, Paik J, Vodovotz Y, Nathan C. Contrasting mechanisms for suppression of macrophage cytokine release by transforming growth factor-β and interleukin-10. J Biol Chem 1992;267:23301-23308.
    Pubmed
  15. Werner F, Jain MK, Feinberg MW, Sibinga NE, Pellacani A, Wiesel P, Chin MT, Topper JN, Perrella MA, Lee ME. Transforming growth factor-β1 inhibition of macrophage activation is mediated via Smad3. J Biol Chem 2000;275:36653-36658.
    Pubmed
  16. Vodovotz Y, Bogdan C, Paik J, Xie QW, Nathan C. Mechanisms of suppression of macrophage nitric oxide release by transforming growth factorβ. J Exp Med 1993;178:605-613.
    Pubmed
  17. Kim WK, Ko KH. Potentiation of N-methyl-D-aspartate-mediated neurotoxicity by immunostimulated murine microglia. J Neurosci Res 1998;54:17-26.
    Pubmed
  18. Kong LY, Lai C, Wilson BC, Simpson JN, Hong JS. Protein tyrosine kinase inhibitors decrease lipopolysaccharide-induced proinflammatory cytokine production in mixed glia, microglia-enriched or astrocyte-enriched cultures. Neurochem Int 1997;30:491-497.
    Pubmed
  19. Oh YK, Swanson JA. Different fates of phagocytosed particles after delivery into macrophage lysosomes. J Cell Biol 1996;132:585-593.
    Pubmed
  20. Lowry MB, Duchemin AM, Robinson JM, Anderson CL. Functional separation of pseudopod extension and particle internalization during Fc gamma receptor-mediated phagocytosis. J Exp Med 1998;187:161-176.
    Pubmed
  21. Bruce-Keller AJ, Keeling JL, Keller JN, Huang FF, Camondola S, Mattson MP. Antiinflammatory effects of estrogen on microglial activation. Endocrinology 2000;141:3646-3656.
    Pubmed
  22. Kim WG, Mohney RP, Wilson B, Jeohn GH, Liu B, Hong JS. Regional difference in susceptibility to lipopolysaccharide-induced neurotoxicity in the rat brain: role of microglia. J Neurosci 2000;20:6309-6316.
    Pubmed
  23. Choi JJ, Oh YK, Kim HS, Kim HC, Ko KH, Kim WK. Mimosine prevents the death of glucose-deprived immunostimulated astrocytes by scavenging peroxynitrite. Glia 2002;39:37-46.
    Pubmed
  24. Choi IY, Lee JC, Ju C, Hwang S, Cho GS, Lee HW, Choi WJ, Jeong LS, Kim WK. A3 adenosine receptor agonist reduces brain ischemic injury and inhibits inflammatory cell migration in rats. Am J Pathol 2011;179:2042-2052.
    Pubmed
  25. Sumner DR, Turner TM, Urban RM, Leven RM, Hawkins M, Nichols EH, McPherson JM, Galante JO. Locally delivered rhTGF-β2 enhances bone ingrowth and bone regeneration at local and remote sites of skeletal injury. J Orthop Res 2001;19:85-94.
    Pubmed
  26. Randolph GJ, Sanchez-Schmitz G, Liebman RM, Schäkel K. The CD16(+) (FcgammaRIII(+)) subset of human monocytes preferentially becomes migratory dendritic cells in a model tissue setting. J Exp Med 2002;196:517-527.
    Pubmed
  27. Adams DH, Hathaway M, Shaw J, Burnett D, Elias E, Strain AJ. Transforming growth factor-beta induces human T lymphocyte migration in vitro. J Immunol 1991;147:609-612.
    Pubmed
  28. Flanders KC, Burmester JK. Medical applications of transforming growth factor-β. Clin Med Res 2003;1:13-20.
    Pubmed
  29. Wang XJ, Han G, Owens P, Siddiqui Y, Li AG. Role of TGF beta-mediated inflammation in cutaneous wound healing. J Investig Dermatol Symp Proc 2006;11:112-117.
  30. Roberts AB, Sporn MB. Transforming growth factor-β. The molecular and cellular biology of wound repair. New York, NY: Plenum Press, 1996; 1996. p. 275-308.
  31. Robson MC, Phillip LG, Cooper DM, Lyle WG, Robson LE, Odom L, Hill DP, Hanham AF, Ksander GA. Safety and effect of transforming growth factor-β2 for treatment of venous stasis ulcers. Wound Repair Regen 1995;3:157-167.
    Pubmed
  32. Hirshberg J, Coleman J, Marchant B, Rees RS. TGF-β3 in the treatment of pressure ulcers: a preliminary report. Adv Skin Wound Care 2001;14:91-95.
    Pubmed
  33. Flanders KC, Ren RF, Lippa CF. Transforming growth factor-βs in neurodegenerative disease. Prog Neurobiol 1998;54:71-85.
    Pubmed
  34. Jin R, Yang G, Li G. Inflammatory mechanisms in ischemic stroke: role of inflammatory cells. J Leukoc Biol 2010;87:779-789.
    Pubmed
  35. Pál G, Vincze C, Renner E, Wappler EA, Nagy Z, Lovas G, Dobolyi A. Time course, distribution and cell types of induction of transforming growth factor betas following middle cerebral artery occlusion in the rat brain. PLoS One 2012;7:e46731.
    Pubmed