Macrophage dysfunction impairs resolution of inflammation in the wounds of diabetic mice

Savita Khanna, Sabyasachi Biswas, Yingli Shang, Eric Collard, Ali Azad, Courtney Kauh, Vineet Bhasker, Gayle M Gordillo, Chandan K Sen, Sashwati Roy, Savita Khanna, Sabyasachi Biswas, Yingli Shang, Eric Collard, Ali Azad, Courtney Kauh, Vineet Bhasker, Gayle M Gordillo, Chandan K Sen, Sashwati Roy

Abstract

Background: Chronic inflammation is a characteristic feature of diabetic cutaneous wounds. We sought to delineate novel mechanisms involved in the impairment of resolution of inflammation in diabetic cutaneous wounds. At the wound-site, efficient dead cell clearance (efferocytosis) is a pre-requisite for the timely resolution of inflammation and successful healing.

Methodology/principal findings: Macrophages isolated from wounds of diabetic mice showed significant impairment in efferocytosis. Impaired efferocytosis was associated with significantly higher burden of apoptotic cells in wound tissue as well as higher expression of pro-inflammatory and lower expression of anti-inflammatory cytokines. Observations related to apoptotic cell load at the wound site in mice were validated in the wound tissue of diabetic and non-diabetic patients. Forced Fas ligand driven elevation of apoptotic cell burden at the wound site augmented pro-inflammatory and attenuated anti-inflammatory cytokine response. Furthermore, successful efferocytosis switched wound macrophages from pro-inflammatory to an anti-inflammatory mode.

Conclusions/significance: Taken together, this study presents first evidence demonstrating that diabetic wounds suffer from dysfunctional macrophage efferocytosis resulting in increased apoptotic cell burden at the wound site. This burden, in turn, prolongs the inflammatory phase and complicates wound healing.

Conflict of interest statement

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

Figures

Figure 1. Increased number of apoptotic cells…
Figure 1. Increased number of apoptotic cells in wounds of diabetic mice and humans.
A, Representative mosaic images from day 3 wounds of diabetic (db/db) or non-diabetic (db/+) mice stained with active caspase 3 (brown). Counterstaining was performed using hematoxylin (blue). The mosaic images of whole wounds were collected under 20× magnification guided by MosaiX software (Zeiss) and a motorized stage. Each mosaic image was generated by combining 12–14 images. Inset: higher magnification image of the boxed area marked in the mosaic image. scale bar (inset)  = 10 µm; B–C, quantification of active caspase 3 (B) or TUNEL positive cells (C). Data are shown as mean ± SD (n = 3); *, p<0.05 versus control non diabetic (db/+) mice; D. A representative Western blot image of active caspase-3 (casp-3) and GAPDH (housekeeping) in day3 wound tissue extracts of diabetic (db/db) or non-diabetic (db/+) mice. E. Densitometry data of blot shown in panel D. Data shown are mean ± SD (n = 3). *, p<0.05 compared to db/+ mice; F–G, wound biopsies were obtained from non-diabetic or diabetic patients presented at the wound clinic. Specimens (3 mm punch) were obtained from the edge of wounds immunostained using active caspase 3 (brown) antibody as a marker of apoptotic cells. Counterstaining was performed using hematoxylin (blue); F. microscopic images, arrows indicate positive cells. Scale bar  = 50 µm; G, quantification of active caspase 3 positive areas shown in F. Data shown are mean ± SD (n = 3); * p< 0.05 versus non diabetic leg wounds.
Figure 2. Identification of apoptotic neutrophils and…
Figure 2. Identification of apoptotic neutrophils and endothelial cells in diabetic wounds.
Representative immunostained section of: A, day 3 wound showing neutrophils (green) and active casp-3 (red) staining; and B, day 7 wound showing endothelial cells (CD31, green) and active casp-3 (red) staining. Nuclear counterstaining was performed using DAPI (blue). i, Low power (20x) images, Scale bar  = 50 µm.; ii-iv, high powered images of the boxed area in i showing active casp-3 (red) and DAPI (blue) (ii) anti-neutrophil or anti-CD31 (green) and DAPI (blue) (iii); and merged images of anti-neutrophil/anti-CD31 and active casp-3. Casp3 positive neutrophils/endothelial cells are shown with white arrows. Scale bar (Aii-iv and Bii-iv)  = 10 µm.
Figure 3. Dead cell clearance by wound-site…
Figure 3. Dead cell clearance by wound-site macrophages.
For dead cell clearance assay, wound macrophages were co-cultured with cell-tracker labeled (red) thymocytes. A, Thymocyte apopotosis detected using Annexin V (FITC conjugated). Annexin V binds to externalized phosphatidyl serine (PS), a characteristics of apoptotic cells. Such treatment resulted in over 90% cells becoming phosphatidylserine (PS) positive. Data are mean ± SD; p<0.05 (n = 4). B, F4/80-FITC (green) and DAPI (blue, nuclear) stained wound macrophage establishing link with an apoptotic thymocyte (red); C, wound macrophage (F4/80-FITC and DAPI stained) engulfed a number of red apoptotic thymocytes; D, co-cultures of control (untreated, viable) thymocytes (red) with wound macrophage (DIC image) followed by wash; E, co-cultures of apoptotic thymocytes (red) with wound macrophages (differential image contrast, DIC image) followed by wash; F–G, Representative high magnification image of a macrophage in DIC (F) or stained with F4/80 FITC (green, G) showing engulfed and adhered (white arrows) apoptotic thymocytes (red). H, scoring of thymocytes engulfed by macrophage. Data are presented as phagocytic index which is defined as total number of apoptotic cells engulfed by macrophages in a field of view divided by total number of macrophage presented in the view. This approach enables normalization of the data against macrophage number. Data presented as mean ± SD (n = 3). *, p<0.01 compared to macrophage co-cultured with control thymocytes.
Figure 4. Dead cell clearance activity is…
Figure 4. Dead cell clearance activity is impaired in wound-site macrophages harvested from diabetic mice.
A, representative images of macrophage (phase contrast) from diabetic (db/db) and their matched control non diabetic (db/+) co-cultured with apoptotic thymocytes (red); B–D, quantification of dead cell clearance activity of wound macrophages from three different genetic backgrounds; B, phagocytic index of wound macrophages harvested 3, 7 or 15 day post implantation from db/+ (non-diabetic control) or db/db (type 2 diabetes); C, phagocytic index of wound macrophages harvested day 5 post implantation from NOR (control) or NOD (type 1 diabetes); and D, phagocytic index of wound macrophages harvested day 5 post implantation from Akita (Ins2Akita, type 1 diabetes) & C57Bl6 (non-diabetic controls). Data are mean ±SD (n = 3).*, p<0.05 versus control mice.
Figure 5. Increased pro-inflammatory cytokine levels in…
Figure 5. Increased pro-inflammatory cytokine levels in diabetic wounds and in wound-site macrophages.
A, Cytokine levels in excisional wound tissue collected on days 1, 3 and 7 post-wounding were measured using ELISA. Data are presented as pg cytokine levels per mg of wet tissue. Mean ± SD (n = 5).*, p<0.05 db/db versus db/+; B, PVA sponges were harvested on days 3, 7 or 15 after implantation and macrophages were isolated. Macrophages (1×106) were seeded in 6-well plates. Cytokine levels in culture media was measured 24 h post-seeding using ELISA. Mean ± SD (n = 4). *, p<0.05 db/db versus db/+.
Figure 6. Topical application of Fas-activating anti-CD95…
Figure 6. Topical application of Fas-activating anti-CD95 JO2 to the wound-site increased apoptotic cell count while not inducing apoptosis in keratinocytes.
A, visualization of TUNEL stained apoptotic cells in day 5 wound tissue treated with anti-murine CD95 (clone:JO2, 2 µg/wound) or vehicle containing isotype control (IgG2). Positive control (wound tissue treated with proteinase K and nuclease) showing TUNEL positive apoptotic cells with green nuclei stain; B, scoring of apoptotic cells in wound tissue sections stained with TUNEL. *, p<0.05 compared to the paired vehicle-treated wounds; C, a representative Western blot image of active caspase-3 (casp-3) and GAPDH (housekeeping) in tissue extracts from IgG2 or JO2 treated d3 wounds; D, densitometric data of blot shown in panel C. Data shown are mean ± SD (n = 3). *, p<0.05 compared to IgG2 treated wounds; E, JO2 treatment did not induce keratinocyte apoptosis. Keratin-14 (green), active caspase-3 (red) and DAPI (blue) stained migrating epithelial tip in placebo (left) or JO2 treated (right) wounds. Scale bar  = 20 µm. Active caspase-3 staining was observed in the granulation tissue but not in the hyper-proliferative epithelium or epithelial tip following JO2 treatment; F, wound area as percentage of initial wound determined on the day 3 after wounding. Data are shown as mean ± SD (n = 4).*, p<0.05 versus corresponding control IgG2 treated wound.
Figure 7. Increasing dead cell burden in…
Figure 7. Increasing dead cell burden in wounds resulted in increased pro-inflammatory cytokine levels.
Wound tissue treated with anti-CD95 JO2 or control IgG2 were harvested on days 0, 1 and 3 post-wounding. Cytokine levels from paired (control and treated) wound tissue were measured using ELISA on the indicated days post-wounding. Significant increase in pro-inflammatory cytokines (TNFα, IL-6, IL1β) and decrease in levels of anti-inflammatory cytokines IL-10 and TGFβ1 was noted in wounds that had increased apoptotic cell load. Data (mean ±SD, n = 5) are presented as pg cytokine per mg wound tissue. *, p<0.05 compared to IgG treated control side.
Figure 8. Efferocytosis of apoptotic cells by…
Figure 8. Efferocytosis of apoptotic cells by wound macrophages resulted in suppression of pro-inflammatory TNFα gene and protein expression.
Following apoptotic cell clearance assay the non-phagocytosed thymocytes were removed by washing and cells were challenged with LPS (1 µg/ml) and IFNγ (10 ng/ml) for 4 h (gene expression) or 16 h (protein expression). TNFα gene (A) and protein (B) expression were measured using real-time PCR and ELISA, respectively. mRNA expression data are presented as % change compared to LPS+IFNγ non-activated control samples. Protein data is expressed as concentration of TNF-α secreted in culture media. Data are mean ±SD (n = 4); **, p<0.01 compared to macrophage cultured with viable cells.

References

    1. Ramsey SD, Newton K, Blough D, McCulloch DK, Sandhu N, et al. Incidence, outcomes, and cost of foot ulcers in patients with diabetes. Diabetes Care. 1999;22:382–387.
    1. Brem H, Tomic-Canic M. Cellular and molecular basis of wound healing in diabetes.[comment]. Journal of Clinical Investigation. 2007;117:1219–1222.
    1. Pierce GF. Inflammation in nonhealing diabetic wounds: the space-time continuum does matter.[comment]. American Journal of Pathology. 2001;159:399–403.
    1. Gillitzer R, Goebeler M. Chemokines in cutaneous wound healing. Journal of Leukocyte Biology. 2001;69:513–521.
    1. Martin P. Wound healing–aiming for perfect skin regeneration. Science. 1997;276:75–81.
    1. Maruyama K, Asai J, Ii M, Thorne T, Losordo DW, et al. Decreased macrophage number and activation lead to reduced lymphatic vessel formation and contribute to impaired diabetic wound healing. American Journal of Pathology. 2007;170:1178–1191.
    1. Abrass CK, Hori M. Alterations in Fc receptor function of macrophages from streptozotocin-induced diabetic rats. J Immunol. 1984;133:1307–1312.
    1. Weisberg SP, McCann D, Desai M, Rosenbaum M, Leibel RL, et al. Obesity is associated with macrophage accumulation in adipose tissue.[see comment]. Journal of Clinical Investigation. 2003;112:1796–1808.
    1. Goren I, Muller E, Schiefelbein D, Christen U, Pfeilschifter J, et al. Systemic anti-TNFalpha treatment restores diabetes-impaired skin repair in ob/ob mice by inactivation of macrophages. Journal of Investigative Dermatology. 2007;127:2259–2267.
    1. Singer AJ, Clark RA. Cutaneous wound healing. New England Journal of Medicine. 1999;341:738–746.
    1. Desmouliere A, Redard M, Darby I, Gabbiani G. Apoptosis mediates the decrease in cellularity during the transition between granulation tissue and scar. American Journal of Pathology. 1995;146:56–66.
    1. Meszaros AJ, Reichner JS, Albina JE. Macrophage phagocytosis of wound neutrophils. J Leukoc Biol. 1999;65:35–42.
    1. deCathelineau AM, Henson PM. The final step in programmed cell death: phagocytes carry apoptotic cells to the grave. Essays in Biochemistry. 2003;39:105–117.
    1. Gardai SJ, Bratton DL, Ogden CA, Henson PM. Recognition ligands on apoptotic cells: a perspective. Journal of Leukocyte Biology. 2006;79:896–903.
    1. Vandivier RW, Henson PM, Douglas IS. Burying the dead: the impact of failed apoptotic cell removal (efferocytosis) on chronic inflammatory lung disease. Chest. 2006;129:1673–1682.
    1. Erwig LP, Henson PM. Clearance of apoptotic cells by phagocytes. Cell Death Differ Epub ahead of print 2007
    1. Fadok VA. Clearance: the last and often forgotten stage of apoptosis. J Mammary Gland Biol Neoplasia. 1999;4:203–211.
    1. Rosen A, Casciola-Rosen L. Autoantigens as substrates for apoptotic proteases: implications for the pathogenesis of systemic autoimmune disease. Cell Death Differ. 1999;6:6–12.
    1. Kaneto H, Kajimoto Y, Miyagawa J, Matsuoka T, Fujitani Y, et al. Beneficial effects of antioxidants in diabetes: possible protection of pancreatic beta-cells against glucose toxicity. Diabetes. 1999;48:2398–2406.
    1. Sablina AA, Budanov AV, Ilyinskaya GV, Agapova LS, Kravchenko JE, et al. The antioxidant function of the p53 tumor suppressor.[see comment]. Nature Medicine. 2005;11:1306–1313.
    1. Esterbauer H, Schaur RJ, Zollner H. Chemistry and biochemistry of 4-hydroxynonenal, malonaldehyde and related aldehydes. Free Radical Biology & Medicine. 1991;11:81–128.
    1. Breyer MD, Bottinger E, Brosius FC, 3rd, Coffman TM, Harris RC, et al. Mouse models of diabetic nephropathy. J Am Soc Nephrol. 2005;16:27–45.
    1. Eming SA, Krieg T, Davidson JM. Inflammation in wound repair: molecular and cellular mechanisms. Journal of Investigative Dermatology. 2007;127:514–525.
    1. Verhoven B, Schlegel RA, Williamson P. Mechanisms of phosphatidylserine exposure, a phagocyte recognition signal, on apoptotic T lymphocytes. Journal of Experimental Medicine. 1995;182:1597–1601.
    1. Kiener PA, Davis PM, Starling GC, Mehlin C, Klebanoff SJ, et al. Differential induction of apoptosis by Fas-Fas ligand interactions in human monocytes and macrophages. Journal of Experimental Medicine. 1997;185:1511–1516.
    1. Matsue H, Kobayashi H, Hosokawa T, Akitaya T, Ohkawara A. Keratinocytes constitutively express the Fas antigen that mediates apoptosis in IFN gamma-treated cultured keratinocytes. Archives of Dermatological Research. 1995;287:315–320.
    1. Glass EJ, Stewart J, Matthews DM, Collier A, Clarke BF, et al. Impairment of monocyte “lectin-like” receptor activity in type 1 (insulin-dependent) diabetic patients. Diabetologia. 1987;30:228–231.
    1. Glass EJ, Stewart J, Weir DM. Altered immune function in alloxan-induced diabetes in mice. Clin Exp Immunol. 1986;65:614–621.
    1. Wheat LJ. Infection and diabetes mellitus. Diabetes Care. 1980;3:187–197.
    1. O'Brien BA, Fieldus WE, Field CJ, Finegood DT. Clearance of apoptotic beta-cells is reduced in neonatal autoimmune diabetes-prone rats. Cell Death & Differentiation. 2002;9:457–464.
    1. Liu BF, Miyata S, Kojima H, Uriuhara A, Kusunoki H, et al. Low phagocytic activity of resident peritoneal macrophages in diabetic mice: relevance to the formation of advanced glycation end products. Diabetes. 1999;48:2074–2082.
    1. Maree AF, Komba M, Dyck C, Labecki M, Finegood DT, et al. Quantifying macrophage defects in type 1 diabetes. Journal of Theoretical Biology. 2005;233:533–551.
    1. Darby IA, Bisucci T, Hewitson TD, MacLellan DG. Apoptosis is increased in a model of diabetes-impaired wound healing in genetically diabetic mice. International Journal of Biochemistry & Cell Biology. 1997;29:191–200.
    1. Brownlee M. Biochemistry and molecular cell biology of diabetic complications. Nature. 2001;414:813–820.
    1. Sudo C, Ogawara H, Saleh AW, Nishimoto N, Utsugi T, et al. Clinical significance of neutrophil apoptosis in peripheral blood of patients with type 2 diabetes mellitus. Lab Hematol. 2007;13:108–112.
    1. Zhang Z, Liew CW, Handy DE, Zhang Y, Leopold JA, et al. High glucose inhibits glucose-6-phosphate dehydrogenase, leading to increased oxidative stress and {beta}-cell apoptosis. FASEB J 2009
    1. Bouma G, Nikolic T, Coppens JM, van Helden-Meeuwsen CG, Leenen PJ, et al. NOD mice have a severely impaired ability to recruit leukocytes into sites of inflammation. European Journal of Immunology. 2005;35:225–235.
    1. Wang J, Takeuchi T, Tanaka S, Kubo SK, Kayo T, et al. A mutation in the insulin 2 gene induces diabetes with severe pancreatic beta-cell dysfunction in the Mody mouse. J Clin Invest. 1999;103:27–37.
    1. Sullivan SR, Underwood RA, Gibran NS, Sigle RO, Usui ML, et al. Validation of a model for the study of multiple wounds in the diabetic mouse (db/db). Plast Reconstr Surg. 2004;113:953–960.
    1. Trousdale RK, Jacobs S, Simhaee DA, Wu JK, Lustbader JW. Wound closure and metabolic parameter variability in a db/db mouse model for diabetic ulcers. J Surg Res. 2009;151:100–107.
    1. Hummel KP, Dickie MM, Coleman DL. Diabetes, a new mutation in the mouse. Science. 1966;153:1127–1128.
    1. Chen H, Charlat O, Tartaglia LA, Woolf EA, Weng X, et al. Evidence that the diabetes gene encodes the leptin receptor: identification of a mutation in the leptin receptor gene in db/db mice. Cell. 1996;84:491–495.
    1. Chow F, Ozols E, Nikolic-Paterson DJ, Atkins RC, Tesch GH. Macrophages in mouse type 2 diabetic nephropathy: correlation with diabetic state and progressive renal injury. Kidney Int. 2004;65:116–128.
    1. Werner S, Grose R. Regulation of wound healing by growth factors and cytokines. Physiological Reviews. 2003;83:835–870.
    1. Moore KW, de Waal Malefyt R, Coffman RL, O'Garra A. Interleukin-10 and the interleukin-10 receptor. Annual Review of Immunology. 2001;19:683–765.
    1. Liechty KW, Kim HB, Adzick NS, Crombleholme TM. Fetal wound repair results in scar formation in interleukin-10-deficient mice in a syngeneic murine model of scarless fetal wound repair. Journal of Pediatric Surgery. 2000;35:866–872; discussion 872-863.
    1. Lundberg JE, Roth TP, Dunn RM, Doyle JW. Comparison of IL-10 levels in chronic venous insufficiency ulcers and autologous donor tissue. Archives of Dermatological Research. 1998;290:669–673.
    1. Wetzler C, Kampfer H, Stallmeyer B, Pfeilschifter J, Frank S. Large and sustained induction of chemokines during impaired wound healing in the genetically diabetic mouse: prolonged persistence of neutrophils and macrophages during the late phase of repair. Journal of Investigative Dermatology. 2000;115:245–253.
    1. Hubner G, Brauchle M, Smola H, Madlener M, Fassler R, et al. Differential regulation of pro-inflammatory cytokines during wound healing in normal and glucocorticoid-treated mice. Cytokine. 1996;8:548–556.
    1. Cooper L, Johnson C, Burslem F, Martin P. Wound healing and inflammation genes revealed by array analysis of ‘macrophageless’ PU.1 null mice. Genome Biology. 2005;6:R5.
    1. Peters T, Sindrilaru A, Hinz B, Hinrichs R, Menke A, et al. Wound-healing defect of CD18(-/-) mice due to a decrease in TGF-beta1 and myofibroblast differentiation. EMBO Journal. 2005;24:3400–3410.
    1. Cohen JJ. Programmed cell death in the immune system. Advances in Immunology. 1991;50:55–85.
    1. 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. Journal of Experimental Medicine. 1996;184:429–440.
    1. Haslett C. Resolution of acute inflammation and the role of apoptosis in the tissue fate of granulocytes. Clin Sci (Lond) 1992;83:639–648.
    1. Lawrence T, Willoughby DA, Gilroy DW. Anti-inflammatory lipid mediators and insights into the resolution of inflammation. Nat Rev Immunol. 2002;2:787–795.
    1. Savill J. Apoptosis in resolution of inflammation. Kidney Blood Press Res. 2000;23:173–174.
    1. Manfredi AA, Iannacone M, D'Auria F, Rovere-Querini P. The disposal of dying cells in living tissues. Apoptosis. 2002;7:153–161.
    1. Ren Y, Savill J. Apoptosis: the importance of being eaten. Cell Death Differ. 1998;5:563–568.
    1. Savill J, Dransfield I, Gregory C, Haslett C. A blast from the past: clearance of apoptotic cells regulates immune responses. Nature Reviews Immunology. 2002;2:965–975.
    1. Fadok VA, Bratton DL, Konowal A, Freed PW, Westcott JY, et al. 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.
    1. Voll RE, Herrmann M, Roth EA, Stach C, Kalden JR, et al. Immunosuppressive effects of apoptotic cells. Nature. 1997;390:350–351.
    1. Huynh ML, Fadok VA, Henson PM. Phosphatidylserine-dependent ingestion of apoptotic cells promotes TGF-beta1 secretion and the resolution of inflammation. Journal of Clinical Investigation. 2002;109:41–50.
    1. Stein M, Keshav S, Harris N, Gordon S. Interleukin 4 potently enhances murine macrophage mannose receptor activity: a marker of alternative immunologic macrophage activation. Journal of Experimental Medicine. 1992;176:287–292.
    1. Benoit M, Desnues B, Mege JL. Macrophage polarization in bacterial infections. Journal of Immunology. 2008;181:3733–3739.
    1. Porcheray F, Viaud S, Rimaniol AC, Leone C, Samah B, et al. Macrophage activation switching: an asset for the resolution of inflammation. Clinical & Experimental Immunology. 2005;142:481–489.
    1. Michlewska S, Dransfield I, Megson IL, Rossi AG. Macrophage phagocytosis of apoptotic neutrophils is critically regulated by the opposing actions of pro-inflammatory and anti-inflammatory agents: key role for TNF-alpha. FASEB Journal. 2009;23:844–854.
    1. Bystrom J, Evans I, Newson J, Stables M, Toor I, et al. Resolution-phase macrophages possess a unique inflammatory phenotype that is controlled by cAMP. Blood. 2008;112:4117–4127.
    1. Sen CK, Khanna S, Babior BM, Hunt TK, Ellison EC, et al. Oxidant-induced vascular endothelial growth factor expression in human keratinocytes and cutaneous wound healing. J Biol Chem. 2002;277:33284–33290.
    1. Roy S, Khanna S, Nallu K, Hunt TK, Sen CK. Dermal wound healing is subject to redox control. Mol Ther. 2006;13:211–220.
    1. Roy S, Khanna S, Rink C, Biswas S, Sen CK. Characterization of the acute temporal changes in excisional murine cutaneous wound inflammation by screening of the wound-edge transcriptome. Physiol Genomics 2008
    1. Albina JE, Mills CD, Barbul A, Thirkill CE, Henry WL, Jr, et al. Arginine metabolism in wounds. American Journal of Physiology. 1988;254:E459–467.
    1. Efron DT, Barbul A. Subcutaneous sponge models. Methods in Molecular Medicine. 2003;78:83–93.
    1. Schaffer MR, Tantry U, Barbul A. Wound fluid inhibits wound fibroblast nitric oxide synthesis. Journal of Surgical Research. 2004;122:43–48.
    1. Depraetere V. “Eat me” signals of apoptotic bodies. Nat Cell Biol. 2000;2:E104.
    1. Devitt A, Gregory CD. Measurement of apoptotic cell clearance in vitro. Methods Mol Biol. 2004;282:207–221.
    1. Underwood RA, Gibran NS, Muffley LA, Usui ML, Olerud JE. Color subtractive-computer-assisted image analysis for quantification of cutaneous nerves in a diabetic mouse model. Journal of Histochemistry & Cytochemistry. 2001;49:1285–1291.
    1. Matalka KZ, Tutunji MF, Abu-Baker M, Abu Baker Y. Measurement of protein cytokines in tissue extracts by enzyme-linked immunosorbent assays: application to lipopolysaccharide-induced differential milieu of cytokines. Neuroendocrinology Letters. 2005;26:231–236.
    1. Khanna S, Roy S, Park HA, Sen CK. Regulation of c-SRC activity in glutamate-induced neurodegeneration. J Biol Chem. 2007;Jun 14; [Epub ahead of print]
    1. Roy S, Khanna S, Bickerstaff AA, Subramanian SV, Atalay M, et al. Oxygen sensing by primary cardiac fibroblasts: a key role of p21(Waf1/Cip1/Sdi1). Circ Res. 2003;92:264–271.
    1. Roy S, Khanna S, Rink T, Radtke J, Williams WT, et al. P21waf1/cip1/sdi1 as a central regulator of inducible smooth muscle actin expression and differentiation of cardiac fibroblasts to myofibroblasts. Molecular Biology of the Cell. 2007;18:4837–4846.

Source: PubMed

3
Tilaa