Skip to main content
Log in

Surfactant Copolymer Annealing of Chemically Permeabilized Cell Membranes

  • Published:
Regenerative Engineering and Translational Medicine Aims and scope Submit manuscript

Abstract

Structural breakdown of the cell membrane is a primary mediator in trauma-induced tissue necrosis. When membrane disruption exceeds intrinsic membrane sealing processes, biocompatible multi-block amphiphilic copolymer surfactants such as Poloxamer 188 (P188) have been found to be effective in catalyze or augment sealing. Although in living cells copolymer-induced sealing of membrane defects has been detected by changes in membrane transport properties, it has not been directly imaged. In this project, we used Atomic force microscopy (AFM) to directly image saponin permeabilized and poloxamer-sealed plasma membranes of monolayer-cultured MDCK and 3T3 fibroblasts. AFM image analysis resulted in the density and diameter ranges for membrane indentations per 5 × 5 μm area. For control, saponin lysed, and P188 treatment of saponin-lysed membranes, the supra-threshold indentation density was 3.6 ± 2.8, 13.8 ± 6.7, and 4.9 ± 3.3/cell, respectively. These results indicated that P188 catalyzed reduction in size of AFM indentations which correlated with increase cell survival. This evidence confirms that biocompatible surfactant P188 augments natural cell membrane sealing capability when intrinsic processes are incapable alone.

Lay Summary and Future Studies

The fundamental purpose of this study was to compare the topography of living mammalian cell membranes before and after detergent disruption using direct atomic force microscope (AFM) measurement membrane surface topography. Because increased roughness was observed, as reflected in greater depth and diameter of AFM tip indentations, the effect of Poloxamer 188 (P188, ~ 9 kDa) on the membrane was investigated. P188 is a copolymer surfactant that is known to seal disrupted pure lipid bilayer membranes. We observed that P188 reduced the roughness disrupted cell membranes consistent with defect sealing. The pure hydrophilic (10 kDa) neutral polyethylene glycol did not induce sealing.

Future investigation will focus on optimizing the molecular design of block copolymer surfactants to maximize their membrane sealing efficacy. The optimal structure or membrane sealing is likely to be cell phenotype specific.

This is a preview of subscription content, log in via an institution to check access.

Access this article

Price excludes VAT (USA)
Tax calculation will be finalised during checkout.

Instant access to the full article PDF.

Fig. 1
Fig. 2
Fig. 3
Fig. 4
Fig. 5
Fig. 6
Fig. 7

Similar content being viewed by others

References

  1. Feury K. Injury prevention: where are the resources? Orthopaedic nursing. Orthop Nurs. 2003;22(2):124–30. https://doi.org/10.1097/00006416-200303000-00010.

    Article  Google Scholar 

  2. Kendrick D, Young B, Mason-Jones A, Ilyas N, Achana F, Cooper N, et al. Home safety education and provision of safety equipment for injury prevention. Cochrane Database Sytematic Rev. 2012;CD005014.

  3. Bischof JC, Padanilam J, Holmes WH, Ezzell RM, Lee R, Tompkins RG, et al. Dynamics of cell membrane permeability changes at supraphysiological temperatures. Biophys J. 1995;68(6):2608–14. https://doi.org/10.1016/S0006-3495(95)80445-5.

    Article  Google Scholar 

  4. Bullock R, Zauner A, Woodward JJ, Myseros J, Choi SC, Ward JD, et al. Factors affecting excitatory amino acid release following severe human head injury. J Neurosurg. 1998;89(4):507–18. https://doi.org/10.3171/jns.1998.89.4.0507.

    Article  Google Scholar 

  5. Stark G. Functional consequences of oxidative membrane damage. J Membr Biol. 2005;205(1):1–16. https://doi.org/10.1007/s00232-005-0753-8.

    Article  Google Scholar 

  6. Maskarinec SA, Wu G, Lee KYC. Membrane sealing by polymers. Ann N Y Acad Sci. 2006;1066:310–20.

    Article  Google Scholar 

  7. Dalal N, Lee R. Treatment of burn injury by cellular repair : journal of craniofacial surgery. J Craniofac Surg. 2008;19(4):903–6. https://doi.org/10.1097/SCS.0b013e318175b541.

    Article  Google Scholar 

  8. Greenebaum B, Blossfield K, Hannig J, Carrillo CS, Beckett MA, Weichselbaum RR, et al. Poloxamer 188 prevents acute necrosis of adult skeletal muscle cells following high-dose irradiation. Burns. 2004;30(6):539–47. https://doi.org/10.1016/j.burns.2004.02.009.

    Article  Google Scholar 

  9. Lee RC, Hannig J, Matthews KL, Myerov A, Chen C-T. Pharmaceutical therapies for sealing of Permeabilized cell membranes in electrical Injuriesa. Ann N Y Acad Sci. 1999;888(1 OCCUPATIONAL):266–73. https://doi.org/10.1111/j.1749-6632.1999.tb07961.x.

    Article  Google Scholar 

  10. Serbest G, Horwitz J, Jost M, Barbee K. Mechanisms of cell death and neuroprotection by poloxamer 188 after mechanical trauma. FASEB J. 2006;20(2):308–10. https://doi.org/10.1096/fj.05-4024fje.

    Article  Google Scholar 

  11. Ng R, Metzger J, Claflin D, Faulkner J. Poloxamer 188 reduces the contraction-induced force decline in lumbrical muscles from mdx mice. Am J Physiol Cell Physiol. 2008;295(1):C146–50. https://doi.org/10.1152/ajpcell.00017.2008.

    Article  Google Scholar 

  12. Townsend D, Yasuda S, Metzger J. Cardiomyopathy of Duchenne muscular dystrophy: pathogenesis and prospect of membrane sealants as a new therapeutic approach. Expert Rev Cardiovasc Ther. 2007;5(1):99–109. https://doi.org/10.1586/14779072.5.1.99.

    Article  Google Scholar 

  13. Yasuda S, Townsend D, Michele DE, Favre EG, Day SM, Metzger JM. Dystrophic heart failure blocked by membrane sealant poloxamer. Nature. 2005;436(7053):1025–9. https://doi.org/10.1038/nature03844.

    Article  Google Scholar 

  14. Wu G, Majewski J, Ege C, Kjaer K, Weygand MJ, Lee KYC. Lipid corralling and Poloxamer squeeze-out in membranes. Phys Rev Lett. 2004;93(2):028101. https://doi.org/10.1103/PhysRevLett.93.028101.

    Article  Google Scholar 

  15. Maskarinec SA, Hannig J, Lee RC, Lee KYC. Direct observation of Poloxamer 188 insertion into lipid monolayers. Biophys J. 2002;82(3):1453–9. https://doi.org/10.1016/S0006-3495(02)75499-4.

    Article  Google Scholar 

  16. Raucher D, Sheetz MP. Characteristics of a membrane reservoir buffering membrane tension. Biophys J. 1999;77(4):1992–2002. https://doi.org/10.1016/S0006-3495(99)77040-2.

    Article  Google Scholar 

  17. Glauert AM, Dingle JT, Lucy JA. Action of Saponin on biological cell membranes. Nature. 1962;196(4858):953–5. https://doi.org/10.1038/196953a0.

    Article  Google Scholar 

  18. Schulz I. Permeabilizing cells: some methods and applications for the study of intracellular processes. In: Enzymology B-M in, editor. Academic press; 1990. p. 280–300.

  19. Gissel H. The role of Ca2+ in muscle cell damage. Ann N Y Acad Sci. 2006;1066:166–80.

    Article  Google Scholar 

  20. LaPlaca MC, Simon CM, Prado GR, Cullen DK. CNS. Injury biomechanics and experimental models. In: Maas JTW and AIR, editor. Prog Brain Res Elsevier; 2007. p 13–26.

  21. Francis G, Kerem Z, Makkar HP, Becker K. The biological action of saponins in animal systems: a review. Br J Nutr. 2002;88(06):587–605. https://doi.org/10.1079/BJN2002725.

    Article  Google Scholar 

  22. Binnig G, Quate CF, Gerber C. Atomic force microscope. Phys Rev Lett. 1986;56(9):930–3. https://doi.org/10.1103/PhysRevLett.56.930.

    Article  Google Scholar 

  23. Radmacher M, Tillmann RW, Fritz M, Gaub HE. From molecules to cells: imaging soft samples with the atomic force microscope. Science. 1992;257(5078):1900–6. https://doi.org/10.1126/science.1411505.

    Article  Google Scholar 

  24. Schaus SS, Henderson ER. Cell viability and probe-cell membrane interactions of XR1 glial cells imaged by atomic force microscopy. Biophys J. 1997;73(3):1205–14. https://doi.org/10.1016/S0006-3495(97)78153-0.

    Article  Google Scholar 

  25. Dai J, Sheetz MP. Mechanical properties of neuronal growth cone membranes studied by tether formation with laser optical tweezers. Biophys J. 1995;68(3):988–96. https://doi.org/10.1016/S0006-3495(95)80274-2.

    Article  Google Scholar 

  26. Brooks JC, Carmichael SW. Scanning electron microscopy of chemically skinned bovine adrenal medullary chromaffin cells. Mikroskopie. 1983;40(11-12):347–56.

    Google Scholar 

  27. Lee R, River L, Pan F, Ji L, Wollmann R. Surfactant-induced sealing of electropermeabilized skeletal muscle membranes in vivo. Proc Natl Acad Sci. 1992;89(10):4524–8. https://doi.org/10.1073/pnas.89.10.4524.

    Article  Google Scholar 

  28. Lee R, Zhang D, Hannig J. Biophysical injury mechanisms in electrical shock trauma. Annu Rev Biomed Eng. 2000;2(1):477–509. https://doi.org/10.1146/annurev.bioeng.2.1.477.

    Article  Google Scholar 

  29. Baskaran H, Toner M, Yarmush ML, Berthiaume F. Poloxamer-188 improves capillary blood flow and tissue viability in a cutaneous burn wound. J Surg Res. 2001;101(1):56–61. https://doi.org/10.1006/jsre.2001.6262.

    Article  Google Scholar 

  30. Merchant FA, Holmes WH, Capelli-Schellpfeffer M, Lee RC, Toner M. Poloxamer 188 enhances functional recovery of lethally heat-shocked fibroblasts. J Surg Res. 1998;74(2):131–40. https://doi.org/10.1006/jsre.1997.5252.

    Article  Google Scholar 

  31. Collins J, Despa F, Lee R. Structural and functional recovery of electropermeabilized skeletal muscle in-vivo after treatment with surfactant poloxamer 188. Biochim Biophys Acta BBA Biomembr. 1768;2007:1238–46.

    Google Scholar 

  32. Curry DJ, Wright DA, Lee RC, Kang UJ, Frim DM. Surfactant poloxamer 188—related decreases in inflammation and tissue damage after experimental brain injury in rats. J Neurosurg Pediatr. 2004;101(2):91–6. https://doi.org/10.3171/ped.2004.101.2.0091.

    Article  Google Scholar 

  33. Borgens RB, Bohnert D, Duerstock B, Spomar D, Lee RC. Subcutaneous tri-block copolymer produces recovery from spinal cord injury. J Neurosci Res. 2004;76(1):141–54. https://doi.org/10.1002/jnr.20053.

    Article  Google Scholar 

  34. Williams IA, Allen DG. Intracellular calcium handling in ventricular myocytes from mdx mice. Am J Physiol Heart Circ Physiol. 2007;292:H846–55.

    Article  Google Scholar 

  35. Diederichs FA. Decrease of both [Ca2+]e and [H+]e produces cell damage in the perfused rat heart. Cell Calcium. 1997;22(6):487–96. https://doi.org/10.1016/S0143-4160(97)90076-2.

    Article  Google Scholar 

  36. Adams-Graves P, Kedar A, Koshy M, Steinberg M, Veith R, Ward D, et al. RheothRx (Poloxamer 188) injection for the acute painful episode of sickle cell disease: a pilot study. Blood. 1997;90(5):2041–6.

    Google Scholar 

Download references

Acknowledgments

This work was financially supported by National Institutes of Health (GM 64757) grants to R Lee and in part by grant from the Office of Naval Research (MC). The authors would like to thank the University of Chicago Flow Cytometry Core Facility for supporting the flow cytometry experiments and Dr. Karl. S. Matlin of the University Of Chicago Department Of Surgery for the MDCK cell line.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Raphael Lee.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Chen, H., McFaul, C., Titushkin, I. et al. Surfactant Copolymer Annealing of Chemically Permeabilized Cell Membranes. Regen. Eng. Transl. Med. 4, 1–10 (2018). https://doi.org/10.1007/s40883-017-0044-9

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s40883-017-0044-9

Keywords

Navigation