doi: 10.3967/bes2018.110
Biocompatibility and Immunotoxicology of the Preclinical Implantation of a Collagen-based Artificial Dermal Regeneration Matrix
-
Abstract:
Objective Graft rejection, with the possibility of a violent immune response, may be severe and life threatening. Our aim was to thoroughly investigate the biocompatibility and immunotoxicology of collagen-based dermal matrix (DM) before assessment in clinical trials. Methods DM was subcutaneously implanted in BALB/c mice in two doses to induce a potential immune response. The spleen and lymph nodes were assessed for shape, cell number, cell phenotype via flow cytometry, cell activation via CCK8 kit, Annexin V kit, and Ki67 immunostaining. Serum samples were used to measure antibody concentration by enzyme-linked immunosorbent assay. Local inflammation was analyzed by histology and immunohistochemistry staining. Data analysis was performed by one-way ANOVA and non-parametric tests. Results Our data illustrate that the spleen and lymph node sizes were similar between the negative control mice and mice implanted with DM. However, in the high-dose DM (DM-H) group, the total cell populations in the spleen and lymph nodes, T cells and B cells in the spleen had slight increases in prophase, and the low-dose DM (DM-L) group did not display gross abnormities. Moreover, DM-H initiated moderate cell activation and proliferation in the early phase post-immunization, whereas DM-L did not. Neither DM-H nor DM-L implantation noticeably increased IgM and IgG serum concentrations. Examination of the local cellular response revealed only benign cell infiltration and TNF-α expression in slides of DM in the early phase. Conclusion Overall, DM-H may have induced a benign temporary acute immune response post-implantation, whereas DM-L had quite low immunogenicity. Thus, this DM can be regarded as a safe product. -
Key words:
- Collagen /
- Lymphocytes /
- Immunogenicity /
- Flow cytometry
-
Figure 1. Images of the spleen (A) and lymph nodes (B) on days 7 and 60, as well as their cell populations (C, D) at different harvesting time points. In each image, the spleens and lymph nodes are placed in the order of PC, DM, and NC from left to right. Data are expressed as the mean ± SEM, n ≥ 3. Spl: spleen; LN: lymph nodes.
Figure 2. Splenic TCR+, CD19+, and CD11b+ cell subpopulations on days 7 (A) and 60 (B) following three immunizations. Data were calculated from the results of cell counting and flow cytometry plots. Data are expressed as the mean ± SEM, n ≥ 3. *P < 0.05, **P < 0.01, and ***P < 0.005, indicate significant differences from the NC group.
Figure 3. TCR+ and CD19+ cell subpopulations in lymph nodes on days 7 (A) and 60 (B) following three immunizations. Data were calculated from the results of cell counting and flow cytometry plots. Data are expressed as the mean ± SEM, n ≥ 3. *P < 0.05 and **P < 0.01 indicate significant differences from the NC group.
Supplementary Figure S2. Splenic lymphocytes activation after three immunizations was accessed by Ki67+ proportion in CD4+ (a), CD8+ (b), and CD19+ (c) lymphocytes in spleen. Splenic lymphocytes subpopulations were incubated with anti-Ki67 PE and analyzed by flow cytometry. Data are present as mean ± SEM of at least 3 samples in every group. Difference is considered significant if **P < 0.01, ***P < 0.005, compared to NC group.
Figure 4. Splenic cell activation determined by Ki67 expression. Ki67 antigen expression was detected by cell immunofluorescence staining with anti-Ki67 PE. Representative flow cytometry plots of Ki67 staining showing the Ki67+ cell percentage in CD8+, CD4+, and CD19+ lymphocytes on day 60 post-implantation.
Figure 5. Splenic lymphocyte activation level after three immunizations by assessing cell proliferative ability and apoptosis level in vitro. Splenic lymphocyte proliferation in vitro on days 7 (A), 14 (B), 60 (C), and 90 (D) post-implantation and apoptosis in vitro on day 7 (e) post-implantation were determined following stimulation with PMA and Iono for 3 days and quantification with a CCK8 kit and Annexin-V kit. Data are shown as the mean ± SEM of at least three samples. ***P < 0.005 indicates a significant difference from the NC group.
Figure 6. Humoral immune response of mice to implants following three immunizations. Double antibody sandwich ELISA was used for IgM (A) and IgG (B) concentration quantification in the peripheral blood of mice. Data are expressed as the mean ± SEM from at least three sera samples per group per time point.
Figure 7. Cell infiltration around implants. Skin samples from implantation sites were cut out, and cell infiltration was determined by H & E staining. Representative images of cell infiltration around implants on days 7 (A-D) and 60 (E-H) after three immunizations, indicating inflammation intensity and duration. Nuclei are stained blue, and the cytoplasm, connective tissues, and muscles are stained red by H & E staining. All sections were examined with an optical microscope. BT: bovine tendon, DM: dermal matrix.
Supplementary Figure S3. Cell infiltration around implants. Representative H & E staining images of at least 3 samples in every group at all sampling time points are shown to analyze local inflammation reaction. Nucleus are stained blue, cytoplasm, connective tissues and muscles are stained red by H & E staining. Images were captured on an optical microscope. BT: bovine tendon, DM: dermal matrix.
Figure 8. Effects of DM implantation on proinflammatory cytokine (IFN-γ) expression. IFN-γ expression at implantation sites was determined by immunohistochemistry. Representative images of IFN-γ immunostaining around implants on days 7 (A-D) and 60 (E-H) post-surgeries are displayed. All sections were imaged with an optical microscope.
Figure 9. Effects of DM implantation on proinflammatory cytokine (TNF-α) expression. TNF-α expression at implantation sites was determined by immunohistochemistry. Representative images of TNF-α immunostaining around implants on days 7 (A-D) and 60 (E-H) post-immunizations are displayed. All sections were examined with an optical microscope. Positive staining is marked with blue arrows.
-
[1] Colvin RB, Smith RN. Antibody-mediated organ-allograft rejection. Nat Rev Immunol, 2005; 5, 807-17. doi: 10.1038/nri1702 [2] Smolenski RT, Forni M, Maccherini M, et al. Reduction of hyperacute rejection and protection of metabolism and function in hearts of human decay accelerating factor (hdaf)-expressing pigs. Cardiovasc Res, 2007; 73, 143-52. doi: 10.1016/j.cardiores.2006.10.027 [3] Puckett FA, Stahlfeld KR, DiMarco RF. Hyperacute rejection of a bovine pericardial prosthesis. Tex Heart Inst J, 2006; 33, 260-1. http://d.old.wanfangdata.com.cn/OAPaper/oai_pubmedcentral.nih.gov_1524681 [4] Benzimra M, Calligaro GL, Glanville AR. Acute Rejection. J Thorac Dis, 2017; 9, 5440-57. http://jtd.amegroups.com/article/view/17414 [5] Lauro A, Oltean M, Marino IR. Chronic rejection after intestinal transplant: Where are we in order to avert it? Dig Dis Sci, 2018; 63, 551-62. doi: 10.1007/s10620-018-4909-7 [6] Khaireddin R, Wachtlin J, Hopfenmüller W, et al. Hla-a, hla-b and hla-dr matching reduces the rate of corneal allograft rejection. Graefe's Arch Clin Exp Ophthalmol, 2003; 241, 1020-8. doi: 10.1007/s00417-003-0759-9 [7] Taylor AL, Watson CJ, Bradley JA. Immunosuppressive agents in solid organ transplantation: Mechanisms of action and therapeutic efficacy. Crit Rev Oncol Hematol, 2005; 56, 23-46. doi: 10.1016/j.critrevonc.2005.03.012 [8] Eisen HJ, Tuzcu EM, Dorent R, et al. Everolimus for the prevention of allograft rejection and vasculopathy in cardiac-transplant recipients. N Engl J Med, 2003; 349, 847-58. doi: 10.1056/NEJMoa022171 [9] Soulillou JP. Immune monitoring for rejection of kidney transplants. N Engl J Med, 2001; 344, 1006-7. doi: 10.1056/NEJM200103293441309 [10] Lee CH, Singla A, Lee Y. Biomedical applications of collagen. Int J Pharm, 2001; 221, 1-22. doi: 10.1016/S0378-5173(01)00691-3 [11] Gaspar A, Moldovan L, Constantin D, et al. Collagen-based scaffolds for skin tissue engineering. J Med Life, 2011; 4, 172-7. http://d.old.wanfangdata.com.cn/OAPaper/oai_pubmedcentral.nih.gov_3124265 [12] Powell HM, Supp DM, Boyce ST. Influence of electrospun collagen on wound contraction of engineered skin substitutes. Biomaterials, 2008; 29, 834-43. doi: 10.1016/j.biomaterials.2007.10.036 [13] Peng YY, Glattauer V, Ramshaw JA, et al. Evaluation of the immunogenicity and cell compatibility of avian collagen for biomedical applications. J Biomed Mater Res, Part A, 2010; 93, 1235-44. http://www.ncbi.nlm.nih.gov/pubmed/19777573 [14] Lynn AK, Yannas IV, Bonfield W. Antigenicity and immunogenicity of collagen. J Biomed Mater Res, Part B, 2004; 71, 343-54. doi: 10.1002-jbm.b.30096/ [15] Sundback CA, Shyu JY, Wang Y, et al. Biocompatibility analysis of poly(glycerol sebacate) as a nerve guide material. Biomaterials, 2005; 26, 5454. doi: 10.1016/j.biomaterials.2005.02.004 [16] Hassanbhai AM, Lau CS, Wen F, et al. In vivo immune responses of cross-linked electrospun tilapia collagen membrane. Tissue Eng Part A, 2017; 23. doi: 10.1089/ten.tea.2016.0504. [17] Wang X, Tian J, Yong KT, et al. Immunotoxicity assessment of cdse/zns quantum dots in macrophages, lymphocytes and balb/c mice. J Nanobiotechnol, 2016; 14, 10. doi: 10.1186/s12951-016-0162-4 [18] Pati F, Datta P, Adhikari B, et al. Collagen scaffolds derived from fresh water fish origin and their biocompatibility. J Biomed Mater Res, Part A, 2012; 100, 1068-79. http://d.old.wanfangdata.com.cn/NSTLQK/NSTL_QKJJ0226309575/ [19] Bornapour M, Muja N, Shum-Tim D, et al. Biocompatibility and biodegradability of mg-sr alloys: The formation of sr-substituted hydroxyapatite. Acta Biomater, 2012; 9, 5319-30. http://www.sciencedirect.com/science/article/pii/S1742706112003650 [20] Elsabahy M, Wooley KL. Cytokines as biomarkers of nanoparticle immunotoxicity. Chemical Society Reviews, 2013; 42, 5552. doi: 10.1039/c3cs60064e [21] Tamaddon M, Walton RS, Brand DD, et al. Characterisation of freeze-dried type ii collagen and chondroitin sulfate scaffolds. Journal of Materials Science Materials in Medicine, 2013; 24, 1153-65. doi: 10.1007/s10856-013-4882-9 [22] Kwon HJ, Han Y. Chondroitin sulfate-based biomaterials for tissue engineering, 2016; 40, 290-9. [23] Kim J, Dadsetan M, Ameenuddin S, et al. In vivo biodegradation and biocompatibility of peg/sebacic acid-based hydrogels using a cage implant system. J Biomed Mater Res, Part A, 2010; 95, 191-7. http://www.ncbi.nlm.nih.gov/pubmed/20574982 [24] Liu H, Wise SG, Rnjak-Kovacina J, et al. Biocompatibility of silk-tropoelastin protein polymers. Biomaterials, 2014; 35, 5138-47. doi: 10.1016/j.biomaterials.2014.03.024 [25] Fang JJ, Zhu ZY, Dong H, et al. Effect of spleen lymphocytes on the splenomegaly in hepatocellular carcinoma-bearing mice. Biomed Environ Sci, 2014; 27, 17-26. http://www.wanfangdata.com.cn/details/detail.do?_type=perio&id=bes201401004 [26] Bao LQ, Dang MN, Huy NT, et al. Splenic cd11c(+) cells derived from semi-immune mice protect naïve mice against experimental cerebral malaria. Malar J, 2015; 14, 23. doi: 10.1186/s12936-014-0533-y [27] O'Donnell H, Pham OH, Li LX, et al. Toll-like receptor and inflammasome signals converge to amplify the innate bactericidal capacity of t helper 1 cells. Immunity, 2014; 40, 213-24. doi: 10.1016/j.immuni.2013.12.013 [28] Ge J, Liu Y, Li Q, et al. Resveratrol induces apoptosis and autophagy in t-cell acute lymphoblastic leukemia cells by inhibiting akt/mtor and activating p38-mapk. Biomed Environ Sci, 2013; 26, 902-11. http://www.wanfangdata.com.cn/details/detail.do?_type=perio&id=bes201311005 [29] Duan ZH, Lin ZA, Yao HR, et al. Preparation of artificial antigen and egg yolk-derived immunoglobulin (iγg) of citrinin for enzyme-linked immunosorbent assay. Biomed Environ Sci, 2009; 22, 237-43. doi: 10.1016/S0895-3988(09)60051-9 [30] Welch RJ, Litwin CM. A comparison of brucella igg and igm elisa assays with agglutination methodology. J Clin Lab Anal, 2010; 24, 160-2. doi: 10.1002/jcla.v24:3 [31] Ivana M, Mhfuod EMAA, Jelena D, et al. Pulmonary immune responses to aspergillus fumigatus in rats. Biomed Environ Sci, 2014; 27, 684-94. http://d.wanfangdata.com.cn/Periodical_bes201409004.aspx [32] Wang QT, Wu YJ, Huang B, et al. Etanercept attenuates collagen-induced arthritis by modulating the association between baffr expression and the production of splenic memory b cells. Pharmacol Res, 2013; 68, 38-45. doi: 10.1016/j.phrs.2012.11.003 [33] Rosenberg GA, Cunningham LA, Wallace J, et al. Immunohistochemistry of matrix metalloproteinases in reperfusion injury to rat brain: Activation of mmp-9 linked to stromelysin-1 and microglia in cell cultures. Brain Res, 2001; 893, 104-12. doi: 10.1016/S0006-8993(00)03294-7 [34] Liu PM, Zou L, Sadhu C, et al. Comparative immunogenicity assessment: A critical consideration for biosimilar development. Bioanalysis, 2015; 7, 373-81. doi: 10.4155/bio.14.311 [35] Iwasaki A, Medzhitov R. Regulation of adaptive immunity by the innate immune system. Science, 2010; 327, 291-5. doi: 10.1126/science.1183021 [36] Shen Y, Redmond SL, Papadimitriou JM, et al. The biocompatibility of silk fibroin and acellular collagen scaffolds for tissue engineering in the ear. Biomed Mater, 2014; 9, 015015. doi: 10.1088/1748-6041/9/1/015015 [37] Xiao X, Pan S, Liu X, et al. In vivo study of the biocompatibility of a novel compressed collagen hydrogel scaffold for artificial corneas. J Biomed Mater Res, Part A, 2014; 102, 1782-7. doi: 10.1002/jbm.a.34848 [38] Wufuer M, Lee G, Hur W, et al. Skin-on-a-chip model simulating inflammation, edema and drug-based treatment. Sci Rep, 2016; 6, 37471. doi: 10.1038/srep37471