References

Monassier JP. Reperfusion injury in acute myocardial infarction: from bench to cath lab. Part I: Basic considerations. Arch Cardiovasc Dis. 2008; 101:(7-8)491-500

Pagliaro P, Penna C. Cardiac postconditioning. Antioxid Redox Signal. 2011; 14:(5)777-779

Deftereos S, Angelidis C, Bouras G Innate immune inflammatory response in the acutely ischemic myocardium. Med Chem. 2014; 10:(7)653-662

Martindale JJ, Metzger JM. Uncoupling of increased cellular oxidative stress and myocardial ischemia reperfusion injury by directed sarcolemma stabilization. J Mol Cell Cardiol. 2014; 67:26-37 https://doi.org/10.1016/j.yjmcc.2013.12.008

Yasuda S, Townsend D, Michele DE Dystrophic heart failure blocked by membrane sealant poloxamer. Nature. 2005; 436:(7053)1025-1029 https://doi.org/10.1038/nature03844

Markham BE, Kernodle S, Nemzek J Chronic dosing with membrane sealant poloxamer 188 NF improve respiratory dysfunction in dystrophic Mdx and Mdx/Utrophoin-/-mice. PLoS One. 2015; 10:(8) https://doi.org/10.1371/journal.pone.0134832

Townsend D, Turner I, Yasuda S Chronic administration of membrane sealant prevents severe cardiac injury and ventricular dilatation in dystrophic dogs. J Clin Invest. 2010; 120:(4)1140-1150 https://doi.org/10.1172/JCI41329

Spurney CF, Guerron AD, Yu Q, Sali A Membrane sealant poloxamer P188 protects against isoproterenol induced cardiomyopathy in dystrophin deficient mice. BMC Cardiovasc Disord. 2011; 11 https://doi.org/10.1186/1471-2261-11-20

Kahles T, Brandes RP. Which NADPH oxidase isoform is relevant for ischemic stroke? The case for nox 2. Antioxid Redox Signal. 2013; 18:(12)1400-1417 https://doi.org/10.1089/ars.2012.4721

Gu JH, Ge JB, Li M Poloxamer 188 protects neurons against ischemia/reperfusion injury through preserving integrity of cell membranes and blood brain barrier. PLoS One. 2013; 8:(4) https://doi.org/10.1371/journal.pone.0061641

Mbye LH, Keles E, Tao L Kollidon VA64, a membrane-resealing agent, reduces histopathology and improves functional outcome after controlled cortical impact in mice. J Cereb Blood Flow Metab. 2012; 32:(3)515-524 https://doi.org/10.1038/jcbfm.2011.158

Bao HJ, Wang T, Zhang MY Poloxamer-188 attenuates TBI-induced bloodbrain barrier damage leading to decreased brain edema and reduced cellular death. Neurochem Res. 2012; 37:(12)2856-2867 https://doi.org/10.1007/s11064-012-0880-4

Borgens RB, Bohnert D, Duerstock B Subcutaneous tri-block copolymer produces recovery from spinal cord injury. J Neurosci Res. 2004; 76:(1)141-154 https://doi.org/10.1002/jnr.20053

Shi R. Polyethylene glycol repairs membrane damage and enhances functional recovery: a tissue engineering approach to spinal cord injury. Neurosci Bull. 2013; 29:(4)460-466

Borgens RB. Cellular engineering: molecular repair of membranes to rescue cells of the damaged nervous system. Neurosurgery. 2001; 49:(2)370-378

Bor MV, Durmuş O, Bilgihan A The beneficial effect of 2’-deoxycoformycin in renal ischemia-reperfusion is mediated both by preservation of tissue ATP and inhibition of lipid peroxidation. Int J Clin Lab Res. 1999; 29:(2)75-79

Chakraborti T, Mandal A, Mandal M Complement activation in heart diseases. Role of oxidants. Cell Signal. 2000; 12:(9-10)607-617 https://doi.org/10.1016/S0898-6568(00)00111-X

Lee RC. Injury by electrical forces: pathophysiology, manifestations, and therapy. Curr Probl Surg. 1997; 34:(9)677-764

Collins JM, Despa F, Lee RC. Structural and functional recovery of electropermeabilized skeletal muscle in-vivo after treatment with surfactant poloxamer 188. Biochim Biophys Acta. 2007; 1768:(5)1238-1246 https://doi.org/10.1016/j.bbamem.2007.01.012

Matthews KL, Aarsvold JN, Mintzer RA Tc-99m pyrophosphate imaging of poloxamer-treated electroporated skeletal muscle in an in vivo rat model. Burns. 2006; 32:(6)755-764 https://doi.org/10.1016/j.burns.2006.01.011

Abreu-Blanco MT, Verboon JM, Parkhurst SM. Single cell wound repair: Dealing with life's little traumas. Bioarchitecture. 2011; 1:(3)114-121

McNeil PL, Steinhardt RA. Loss, restoration, and maintenance of plasma membrane integrity. J Cell Biol. 1997; 137:(1)1-4

McNeil PL, Steinhardt RA. Plasma membrane disruption: repair, prevention, adaptation. Annu Rev Cell Dev Biol. 2003; 19:697-731 https://doi.org/10.1146/annurev.cellbio.19.111301.140101

Sonnemann KJ, Bement WM. Wound repair: toward understanding and integration of single-cell and multicellular wound responses. Annu Rev Cell Dev Biol. 2011; 27:237-263 https://doi.org/10.1146/annurev-cellbio-092910-154251

Lee RC. Cytoprotection by stabilization of cell membranes. Ann N Y Acad Sci. 2002; 961:271-275

Matzinger P. Tolerance, danger, and the extended family. Annu Rev Immunol. 1994; 12:991-1045 https://doi.org/10.1146/annurev.iy.12.040194.005015

Kono H, Rock KL. How dying cells alert the immune system to danger. Nat Rev Immunol. 2008; 8:(4)279-289

Armstrong DG, Jude EB. The role of matrix metalloproteinases in wound healing. J Am Podiatr Med Assoc. 2002; 92:(1)12-18

Maskarinec SA, Wu G, Lee KY. Membrane sealing by polymers. Ann N Y Acad Sci. 2005; 1066:310-320

Lee RC, River LP, Pan FS Surfactant-induced sealing of electropermeabilized skeletal muscle membranes in vivo. Proc Natl Acad Sci U S A. 1992; 89:(10)4524-4528

Greenebaum B, Blossfield K, Hannig J Poloxamer c188 prevents acute necrosis of adult skeletal muscle cells following high-dose irradiation. Burns. 2004; 30:(6)539-547 https://doi.org/10.1016/j.burns.2004.02.009

Soneru AP, Beckett MA, Weichselbaum RR, Lee RC. Mg ATP and antioxidants augment the radioprotective effect of surfactant copolymers. Health Phys. 2011; 101:(6)731-738 https://doi.org/10.1097/HP.0b013e3182166759

Hannig J, Lee RC. Structural changes in cell membranes after ionizing electromagnetic field exposure. IEEE Trans Plasma Sci. 2000; 28:97-101

Houang EM, Haman KJ, Filareto A Membranestabilizing copolymers confer marked protection to dystrophic skeletal muscle in vivo. Mol Ther Methods Clin Dev. 2015; 2

Ng R, Metzger JM, Claflin DR, Faulkner JA. Poloxamer 188 reduces the contraction-induced force decline in lumbrical muscles from mdx mice. Am J Physiol Cell Physiol. 2008; 295:(1)C146-C150 https://doi.org/10.1152/ajp-cell.00017.2008

Phillips DM, Haut RC. The use of a non-ionic surfactant (P188) to save chondrocytes from necrosis following impact loading of chondral explants. J Orthop Res. 2004; 22:(5)1135-1142

Serbest G, Horwitz J, Barbee K. The effect of poloxamer-188 on neuronal cell recovery from mechanical injury. J Neurotrauma. 2005; 22:(1)119-132 https://doi.org/10.1089/neu.2005.22.119

Marks JD, Pan C, Bushell T Amphiphilic, tri-block copolymers provide potent, membrane-targeted neuroprotection. FASEB J. 2001; https://doi.org/10.1096fj.00-0547fje

Shelat PB, Plant LD, Wang JC The membraneactive tri-block copolymer pluronic F-68 profoundly rescues rat hippocampal neurons from oxygen-glucose deprivation-induced death through early inhibition of apoptosis. J Neurosci. 2013; 33:(30)12287-12299 https://doi.org/10.1523%2FJNEUROSCI.5731-12.2013

Mina EW, Lasagna-Reeves C, Glabe CG, Kayed R. Poloxamer 188 copolymer membrane sealant rescues toxicity of amyloid oligomers in vitro. J Mol Biol. 2009; 391:(3)577-585 https://doi.org/10.1016/j.jmb.2009.06.024

Lee RC, Astumian RD. The physicochemical basis for thermal and non-thermal ‘burn’ injuries. Burns. 1996; 22:(7)509-519

Wang C, Wang H, Wu D A novel perspective for burn-induced myopathy: Membrane repair defect. Sci Rep. 2016; 6 https://doi.org/10.1038/srep31409

Yuhua S, Ligen L, Jiake C, Tongzhu S. Effect of Poloxamer 188 on deepening of deep second-degree burn wounds in the early stage. Burns. 2012; 38:(1)95-101 https://doi.org/10.1016/j.burns.2010.06.002

Despa F, Orgill DP, Neuwalder J, Lee RC. The relative thermal stability of tissue macromolecules and cellular structure in burn injury. Burns. 2005; 31:(5)568-577 https://doi.org/10.1016/j.burns.2005.01.015

Merchant FA, Holmes WH, Capelli-Schellpfeffer M Poloxamer 188 enhances functional recovery of lethally heat-shocked fibroblasts. J Surg Res. 1998; 74:(2)131-140 https://doi.org/10.1006/jsre.1997.5252

Birchenough SA, Rodeheaver GT, Morgan RF Topical poloxamer-188 improves blood flow following thermal injury in rat mesenteric microvasculature. Ann Plast Surg. 2008; 60:(5)584-588 https://doi.org/10.1097/SAP.0b013e3181651661

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

Maskarinec SA, Hannig J, Lee RC, Lee KY. Direct observation of poloxamer 188 insertion into lipid monolayers. Biophys J. 2002; 82:(3)1453-1459 https://doi.org/10.1016/S0006-3495(02)75499-4

Maskarinec SA, Lee KY. Comparative study of poloxamer inertion into lipid monolayers. Langmuir. 2003; 19:1809-1815 https://doi.org/10.1021/la0626398

Firestone MA, Wolf AC, Seifert S. Small-angle X-ray scattering study of the interaction of poly(ethylene oxide)-b-poly(propylene oxide)-b-poly(ethylene oxide) triblock copolymers with lipid bilayers. Biomacromolecules. 2003; 4:(6)1539-1549 https://doi.org/10.1021/bm034134r

Wu G, Majewski J, Ege C Lipid corralling and poloxamer squeeze-out in membranes. Phys Rev Lett. 2004; 93:(2) https://doi.org/10.1103/PhysRev-Lett.93.028101

Wu G, Majewski J, Ege C Interaction between lipid monolayers and poloxamer 188: an X-ray reflectivity and diffraction study. Biophys J. 2005; 89:(5)3159-3173 https://doi.org/10.1529/biophysj.104.052290

Walsh AM, Mustafi D, Makinen MW, Lee RC. A surfactant copolymer facilitates functional recovery of heat-denatured lysozyme. Ann N Y Acad Sci. 2005; 1066:321-327 https://doi.org/10.1196/annals.1363.029

Lee RC, Despa F, Guo L Surfactant copolymers prevent aggregation of heat denatured lysozyme. Ann Biomed Eng. 2006; 34:(7)1190-1200 https://doi.org/10.1007/s10439-006-9139-z

Frey SL, Zhang D, Carignano MA Effects of block copolymer's architecture on its association with lipid membranes: experiments and simulations. J Chem Phys. 2007; 127:(11) https://doi.org/10.1063/1.2768947

Redhead M, Mantovani G, Nawaz S Relationship between the affinity of PEO-PPO-PEO block copolymers for biological membranes and their cellular effects. Pharm Res. 2012; 29:(7)1908-1918 https://doi.org/10.1007/s11095-012-0716-6

Houang EM, Haman KJ, Kim M Chemical end group modified diblock copolymers elucidate anchor and chain mechanism of membrane stabilization. Mol Pharm. 2017; 14:(7)2333-2339 https://doi.org/10.1021/acs.molpharmaceut.7b00197

Cheng CY, Wang JY, Kausik R Nature of interactions between PEO-PPO-PEO triblock copolymers and lipid membranes: (II) role of hydration dynamics revealed by dynamic nuclear polarization. Biomacromolecules. 2012; 13:(9)2624-2633 https://doi.org/10.1021/bm300848c

Mustafi D, Smith CM, Makinen MW, Lee RC. Multiblock poloxamer surfactants suppress aggregation of denatured proteins. Biochim Biophys Acta. 2008; 1780:(1)7-15 https://doi.org/10.1016/j.bbagen.2007.08.017

Chin J, Mustafi D, Poellmann MJ, Lee RC. Amphiphilic copolymers reduce aggregation of unfolded lysozyme more effectively than polyethylene glycol. Phys Biol. 2017; 14:(1) https://doi.org/10.1088/1478-3975/aa5788

Poellmann MJ, Sosnick TR, Meredith SC, Lee RC. The pentablock amphiphilic copolymer T1107 prevents aggregation of denatured and reduced lysozyme. Macromol Biosci. 2017; 17:(2) https://doi.org/10.1002/mabi.201600217

Palmer JS, Cromie WJ, Lee RC. Surfactant administration reduces testicular ischemia-reperfusion injury. J Urol. 1998; 159:(6)2136-2139

Hannig J, Yu J, Beckett M Poloxamine 1107 sealing of radiopermeabilized erythrocyte membranes. Int J Radiat Biol. 1999; 75:(3)379-385

Wang JY, Marks J, Lee KY. Nature of interactions between PEO-PPO-PEO triblock copolymers and lipid membranes: (I) effect of polymer hydrophobicity on its ability to protect liposomes from peroxidation. Biomacromolecules. 2012; 13:(9)2616-2623 https://doi.org/10.1021/bm300847x

Sandez-Macho I, Casas M, Lage EV Interaction of poloxamine block copolymers with lipid membranes: Role of copolymer structure and membrane cholesterol content. Colloids Surf B Biointerfaces. 2015; 133:270-277 https://doi.org/10.1016/j.colsurfb.2015.06.019

Schaer GL, Spaccavento LJ, Browne KF Beneficial effects of RheothRx injection in patients receiving thrombolytic therapy for acute myocardial infarction. Results of a randomized, double-blind, placebocontrolled trial. Circulation. 1996; 94:(3)298-307

Quinlan JG, Wong BL, Niemeier RT Poloxamer 188 failed to prevent exercise-induced membrane breakdown in mdx skeletal muscle fibers. Neuromuscul Disord. 2006; 16:(12)855-864 https://doi.org/10.1016/j.nmd.2006.09.016

Terry RL, Kaneb HM, Wells DJ. Poloxamer [corrected] 188 has a deleterious effect on dystrophic skeletal muscle function. PLoS One. 2014; 9:(3) https://doi.org/10.1371/journal.pone.0091221

Percival SL, Mayer D, Malone M Surfactants and their role in wound cleansing and biofilm management. J Wound Care. 2017; 26:(11)680-690 https://doi.org/10.12968/jowc.2017.26.11.680

Structure and properties of PluroGel: a new surfactant-based biomaterial. 2016. https://tinyurl.com/ybtxrbch (accessed 6 August 2018)

Ratliff CR. Case series of 18 patients with lower extremity wounds treated with a concentrated surfactant-based gel dressing. J Vasc Nurs. 2018; 36:(1)3-7 https://doi.org/10.1016/j.jvn.2017.09.001

Diegelmann RF. Excessive neutrophils characterize chronic pressure ulcers. Wound Repair Regen. 2003; 11:(6)490-495

Yager DR, Nwomeh BC. The proteolytic environment of chronic wounds. Wound Repair Regen. 1999; 7:(6)433-441

Baum CL, Arpey CJ. Normal cutaneous wound healing: clinical correlation with cellular and molecular events. Dermatol Surg. 2005; 31:(6)674-686

Mast BA, Schultz GS. Interactions of cytokines, growth factors, and proteases in acute and chronic wounds. Wound Repair Regen. 1996; 4:(4)411-420 https://doi.org/10.1046/j.1524-475X.1996.40404.x

Barrick B, Campbell EJ, Owen CA. Leukocyte proteinases in wound healing: roles in physiologic and pathologic processes. Wound Repair Regen. 1999; 7:(6)410-422

Nwomeh BC, Liang HX, Cohen IK, Yager DR. MMP-8 is the predominant collagenase in healing wounds and nonhealing ulcers. J Surg Res. 1999; 81:(2)189-195 https://doi.org/10.1006/jsre.1998.5495

Nwomeh BC, Yager DR, Cohen IK. Physiology of the chronic wound. Clin Plast Surg. 1998; 25:(3)341-356

Gefen A, Weihs D. Cytoskeleton and plasmamembrane damage resulting from exposure to sustained deformations: A review of the mechanobiology of chronic wounds. Med Eng Phys. 2016; 38:(9)828-833 https://doi.org/10.1016/j.medengphy.2016.05.014

James TJ, Hughes MA, Cherry GW, Taylor RP. Evidence of oxidative stress in chronic venous ulcers. Wound Repair Regen. 2003; 11:(3)172-176

Moseley R, Hilton JR, Waddington RJ Comparison of oxidative stress biomarker profiles between acute and chronic wound environments. Wound Repair Regen. 2004; 12:(4)419-429 https://doi.org/10.1111/j.1067-1927.2004.12406.x

Chen WY, Rogers AA. Recent insights into the causes of chronic leg ulceration in venous diseases and implications on other types of chronic wounds. Wound Repair Regen. 2007; 15:(4)434-449 https://doi.org/10.1111/j.1524475X.2007.00250.x

Gary Sibbald R, Woo KY. The biology of chronic foot ulcers in persons with diabetes. Diabetes Metab Res Rev. 2008; 24:S25-S30 https://doi.org/10.1002/dmrr.847

Hu SC, Lan CE. Highglucose environment disturbs the physiologic functions of keratinocytes: Focusing on diabetic wound healing. J Dermatol Sci. 2016; 84:(2)121-127 https://doi.org/10.1016/j.jdermsci.2016.07.008

Armstrong DG, Boulton AJ, Bus SA. Diabetic foot ulcers and their recurrence. N Engl J Med. 2017; 376:(24)2367-2375 https://doi.org/10.1056/NEJM-ra1615439

Fujiwara T, Dohi T, Maan ZN Age-associated intracellular superoxide dismutase deficiency potentiates dermal fibroblast dysfunction during wound healing. Exp Dermatol. 2017; https://doi.org/10.1111/exd.13404

Diegelmann RF, Evans MC. Wound healing: an overview of acute, fibrotic and delayed healing. Front Biosci. 2004; 9:283-289

Zölß C, Cech JD. Efficacy of a new multifunctional surfactant-based biomaterial dressing with 1% silver sulphadiazine in chronic wounds. Int Wound J. 2016; 13:(5)738-743 https://doi.org/10.1111/iwj.12361

Palumbo FP, Harding KG, Abbritti F New surfactant-based dressing product to improve wound closure rates of nonhealing wounds: a European multicenter study including 1036 patients. Wounds. 2016; 28:(7)233-240

Cell salvage in acute and chronic experimental data and early clinical results

02 February 2019
Volume 3 · Issue 1

Abstract

On 9 May 2018, the authors took part in a closed panel discussion on the impact of cell salvage in acute and chronic wounds. The goal was to deliberate the possible use of plurogel micelle matrix (PMM) as a new treatment strategy for wound healing and the authors openly shared their experiences, thoughts, experimental data and early clinical results. The outcome of the panel discussion has been abridged in this paper. The cell membrane consists of a lipid bilayer, which provides a diffusion barrier separating the inside of a cell from its environment. Cell membrane injury can result in acute cellular necrosis when defects are too large and cannot be resealed. There is a potential hazard to the body when these dying cells release endogenous alarm signals referred to as ‘damage (or danger) associated molecular patterns' (DAMPs), which trigger the innate immune system and modulate inflammation. Cell salvage by membrane resealing is a promising target to ensure the survival of the individual cell and prevention of further tissue degeneration by inflammatory processes. Non-ionic surfactants such as poloxamers, poloxamines and PMM have the potential to resuscitate cells by inserting themselves into damaged membranes and stabilising the unstable portions of the lipid bilayers. The amphiphilic properties of these molecules are amenable to insertion into cell wall defects and so can play a crucial, reparative role. This new approach to cell rescue or salvage has gained increasing interest as several clinical conditions have been linked to cell membrane injury via oxidative stress-mediated lipid peroxidation or thermal disruption. The repair of the cell membrane is an important step in salvaging cells from necrosis to prevent further tissue degeneration by inflammatory processes. This is applicable to acute burns and chronic wounds such as diabetic foot ulcers (DFUs), chronic venous leg ulcers (VLUs), and pressure ulcers (PUs). Experimental data shows that PMM is biocompatible and able to insert itself into damaged membranes, salvaging their barrier function and aiding cell survival. Moreover, the six case studies presented in this paper reveal the potential of this treatment strategy.

Injury to the body might macroscopically present as a disruption of tissues, but eventually it is the cells in this tissue that are damaged. With the emergence of molecular medicine, correction or reversal of pathological states at the cellular/molecular level now seems possible.

The concept of cell salvage has primarily been linked to damage like myocardial infarction,1,2,3,4 dystrophic heart failure,5,6,7,8 ischaemic stroke,910 traumatic brain injury,11,12 damage to the nervous system13,14,15 and renal failure,16 some of which represent ischaemia-reperfusion-mediated injuries. Under ischaemic conditions, cellular energy stores are rapidly depleted and degradation products quickly amount to toxic concentrations.16 Subsequent reperfusion, although necessary to restore oxygen and the supply of nutrients, has been associated with pathological inflammatory reactions.17 For example, following reperfusion of ischaemic myocardium, an increase in protease activity, the release of cytokines and other proinflammatory mediators (like IL-1β, TNF-α, and C5a), and excessive formation of reactive oxygen species (ROS) and reactive nitrogen species (RNS) have been reported.17 The oxidative stress that results when the production of free radicals exceeds the tissue's antioxidative capacity1,13 damages major cellular components such as DNA, proteins and lipids, causing cell death.3 There has been a steady increase in interest in cell membrane dysfunction associated with oxidative stress, particularly as traumatic mechanical or electrical injuries could also be linked to cell membrane integrity failure and subsequent cell death.10,11,12,13,14,15,18,19,20

Register now to continue reading

Thank you for visiting Wound Central and reading some of our peer-reviewed resources for wound care professionals. To read more, please register today. You’ll enjoy the following great benefits:

What's included

  • Access to clinical or professional articles

  • New content and clinical updates each month