Can Cytoprotective Cobalt Protoporphyrin Protect Skeletal Muscle and Muscle-derived Stem Cells From Ischemic Injury
详细信息    查看全文
  • 作者:Heather-Marie P. Wilson PhD…
  • 刊名:Clinical Orthopaedics and Related Research?
  • 出版年:2015
  • 出版时间:September 2015
  • 年:2015
  • 卷:473
  • 期:9
  • 页码:2908-2919
  • 全文大小:3,766 KB
  • 参考文献:1.Abraham NG, Kappas A. Pharmacological and clinical aspects of heme oxygenase. Pharmacol Rev. 2008;60:79′7.PubMed View Article
    2.Bilban M, Haschemi A, Wegiel B, Chin BY, Wagner O, Otterbein LE. Heme oxygenase and carbon monoxide initiate homeostatic signaling. J Mol Med. 2008;86:267?9.PubMed View Article
    3.Blaisdell FW. The pathophysiology of skeletal muscle ischemia and the reperfusion syndrome: a review. Cardiovasc Surg. 2002;10:620″0.PubMed View Article
    4.Cao B, Zheng B, Jankowski RJ, Kimura S, Ikezawa M, Deasy B, Cummins J, Epperly M, Qu-Petersen Z, Huard J. Muscle stem cells differentiate into haematopoietic lineages but retain myogenic potential. Nat Cell Biol. 2003;5:640?6.PubMed View Article
    5.Chan RK, Austen WG Jr, Ibrahim S, Ding GY, Verna N, Hechtman HB, Moore FD Jr. Reperfusion injury to skeletal muscle affects primarily type II muscle fibers. J Surg Res. 2004;122:54‰.PubMed View Article
    6.Chen XK, Rathbone CR, Walters TJ. Treatment of tourniquet-induced ischemia reperfusion injury with muscle progenitor cells. J Surg Res. 2011;170:e65″.PubMed View Article
    7.Chung SW, Liu X, Macias AA, Baron RM, Perrella MA. Heme oxygenase-1-derived carbon monoxide enhances the host defense response to microbial sepsis in mice. J Clin Invest. 2008;118:239?7.PubMed Central PubMed View Article
    8.Crawford RS, Hashmi FF, Jones JE, Albadawi H, McCormack M, Eberlin K, Entabi F, Atkins MD, Conrad MF, Austen WG Jr, Watkins MT. A novel model of acute murine hindlimb ischemia. Am J Physiol Heart Circ Physiol. 2007;292:H830″7.PubMed View Article
    9.Eliason JL, Wakefield TW. Metabolic consequences of acute limb ischemia and their clinical implications. Semin Vasc Surg. 2009;22:29″.PubMed View Article
    10.Gozzelino R, Jeney V, Soares MP. Mechanisms of cell protection by heme oxygenase-1. Annu Rev Pharmacol Toxicol. 2010;50:323‵4.PubMed View Article
    11.Hamer PW, McGeachie JM, Davies MJ, Grounds MD. Evans blue dye as an in vivo marker of myofibre damage: optimising parameters for detecting initial myofibre membrane permeability. J Anat. 2002;200:69?.PubMed Central PubMed View Article
    12.Katori M, Buelow R, Ke B, Ma?J, Coito AJ, Iyer S, Southard D, Busuttil RW, Kupiec-Weglinski JW. Heme oxygenase-1 overexpression protects rat hearts from cold ischemia/reperfusion injury via an antiapoptotic pathway. Transplantation. 2002;73:287?2.PubMed View Article
    13.Kawamoto S, Flynn JP, Shi Q, Sakr SW, Luo J, Allen MD. Heme oxygenase-1 induction enhances cell survival and restores contractility to unvascularized three-dimensional adult cardiomyocyte grafts implanted in vivo. Tissue Eng Part A. 2011;17:1605?14.PubMed Central PubMed View Article
    14.Konrad FM, Braun S, Ngamsri KC, Vollmer I, Reutershan J. Heme oxygenase-1 attenuates acute pulmonary inflammation by decreasing the release of segmented neutrophils from the bone marrow. Am J Physiol Lung Cell Mol Physiol. 2014;307:L707?7.PubMed View Article
    15.Lakkisto P, Siren JM, Kyto V, Forsten H, Laine M, Pulkki K, Tikkanen I. Heme oxygenase-1 induction protects the heart and modulates cellular and extracellular remodelling after myocardial infarction in rats. Exp Biol Med (Maywood). 2011;236:1437?48.PubMed View Article
    16.Lawendy AR, Bihari A, Sanders DW, Potter RF, Cepinskas G. The severity of microvascular dysfunction due to compartment syndrome is diminished by the systemic application of CO-releasing molecule (CORM-3). J Orthop Trauma. 2014;28:e263?8.PubMed View Article
    17.Lin YT, Chen YH, Yang YH, Jao HC, Abiko Y, Yokoyama K, Hsu C. Heme oxygenase-1 suppresses the infiltration of neutrophils in rat liver during sepsis through inactivation of p38 MAPK. Shock. 2010;34:615′1.PubMed View Article
    18.Luo J, Weaver MS, Cao B, Dennis JE, Van Biber B, Laflamme MA, Allen MD. Cobalt protoporphyrin pretreatment protects human embryonic stem cell-derived cardiomyocytes from hypoxia/reoxygenation injury in vitro and increases graft size and vascularization in vivo. Stem Cells Trans Med. 2014;3:734?4.View Article
    19.MacGarvey NC, Suliman HB, Bartz RR, Fu P, Withers CM, Welty-Wolf KE, Piantadosi CA. Activation of mitochondrial biogenesis by heme oxygenase-1-mediated NF-E2-related factor-2 induction rescues mice from lethal Staphylococcus aureus sepsis. Am J Respir Crit Care Med. 2012;185:851?1.PubMed Central PubMed View Article
    20.McArdle F, Spiers S, Aldemir H, Vasilaki A, Beaver A, Iwanejko L, McArdle A, Jackson MJ. Preconditioning of skeletal muscle against contraction-induced damage: the role of adaptations to oxidants in mice. J Physiol. 2004;561:233?4.PubMed Central PubMed View Article
    21.Melo LG, Agrawal R, Zhang L, Rezvani M, Mangi AA, Ehsan A, Griese DP, Dell’Acqua G, Mann MJ, Oyama J, Yet SF, Layne MD, Perrella MA, Dzau VJ. Gene therapy strategy for long-term myocardial protection using adeno-associated virus-mediated delivery of heme oxygenase gene. Circulation. 2002;105:602?
  • 作者单位:Heather-Marie P. Wilson PhD (1)
    Robert E. Welikson PhD (1) (2)
    Jun Luo MD, PhD (1) (2)
    Thomas J. Kean PhD (1) (3)
    Baohong Cao MD, PhD (4)
    James E. Dennis PhD (1) (3)
    Margaret D. Allen MD (1) (5)

    1. Benaroya Research Institute at Virginia Mason, 1201 9th Avenue, Seattle, WA, 98101, USA
    2. University of Washington, Seattle, WA, USA
    3. Department of Orthopedic Surgery, Baylor College of Medicine, Houston, TX, USA
    4. Shanghai Chempartner, Shanghai, China
    5. Department of Surgery, University of Washington, Seattle, WA, USA
  • 刊物主题:Orthopedics; Surgical Orthopedics; Medicine/Public Health, general; Surgery; Sports Medicine; Conservative Orthopedics;
  • 出版者:Springer US
  • ISSN:1528-1132
文摘
Background Extremity trauma is the most common injury seen in combat hospitals as well as in civilian trauma centers. Major skeletal muscle injuries that are complicated by ischemia often result in substantial muscle loss, residual disability, or even amputation, yet few treatment options are available. A therapy that would increase skeletal muscle tolerance to hypoxic damage could reduce acute myocyte loss and enhance preservation of muscle mass in these situations. Questions/purposes In these experiments, we investigated (1) whether cobalt protoporphyrin (CoPP), a pharmacologic inducer of cytoprotective heme oxygenase-1 (HO-1), would upregulate HO-1 expression and activity in skeletal muscle, tested in muscle-derived stem cells (MDSCs); and (2) whether CoPP exposure would protect MDSCs from cell death during in vitro hypoxia/reoxygenation. Then, using an in vivo mouse model of hindlimb ischemia/reperfusion injury, we examined (3) whether CoPP pharmacotherapy would reduce skeletal muscle damage when delivered after injury; and (4) whether it would alter the host inflammatory response to injury. Methods MDSCs were exposed in vitro to a single dose of 25μΜ CoPP and harvested over 24 to 96hours, assessing HO-1 protein expression by Western blot densitometry and HO-1 enzyme activity by cGMP levels. To generate hypoxia/reoxygenation stress, MDSCs were treated in vitro with phosphate-buffered saline (vehicle), CoPP, or CoPP plus an HO-1 inhibitor, tin protoporphyrin (SnPP), and then subjected to 5hours of hypoxia (<0.5% O2) followed by 24hours of reoxygenation and evaluated for apoptosis. In vivo, hindlimb ischemia/reperfusion injury was produced in mice by unilateral 2-hour tourniquet application followed by 24hours of reperfusion. In three postinjury treatment groups (n=7 mice/group), CoPP was administered intraperitoneally during ischemia, at the onset of reperfusion, or 1hour later. Two control groups of mice with the same injury received phosphate-buffered saline (vehicle) or the HO-1 inhibitor, SnPP. Myocyte damage in the gastrocnemius and tibialis anterior muscles was determined by uptake of intraperitoneally delivered Evans blue dye (EBD), quantified by image analysis. On serial sections, inflammation was gauged by the mean myeloperoxidase staining intensity per unit area over the entirety of each muscle. Results In MDSCs, a single exposure to CoPP increased HO-1 protein expression and enzyme activity, both of which were sustained for 96hours. CoPP treatment of MDSCs reduced apoptotic cell populations by 55% after in vitro hypoxia/reoxygenation injury (from a mean of 57.3% apoptotic cells in vehicle-treated controls to 25.7% in CoPP-treated cells, mean difference 31.6%; confidence interval [CI], 28.1‵.0; p<0.001). In the hindlimb ischemia/reperfusion model, CoPP delivered during ischemia produced a 38% reduction in myocyte damage in the gastrocnemius muscle (from 86.4%±7% EBD+ myofibers in vehicle-treated, injured controls to 53.2% EBD+ in CoPP-treated muscle, mean difference 33.2%; 95% CI, 18.3, 48.4; p<0.001). A 30% reduction in injury to the gastrocnemius was seen with drug delivery at the onset of reperfusion (to 60.6%±13% EBD+ with CoPP treatment, mean difference 25.8%; CI, 12.2.4; p<0.001). In the tibialis anterior, however, myocyte damage was decreased only when CoPP was given at the onset of reperfusion, resulting in a 27% reduction in injury (from 78.8%±8% EBD+ myofibers in injured controls to 58.3%±14% with CoPP treatment, mean difference 20.5%; CI, 6.1‵.0; p=0.004). Delaying CoPP delivery until 1hour after tourniquet release obviated the protective effect in both muscles. Mean MPO staining intensity per unit area, indicating the host inflammatory response, decreased by 27% across both the gastrocnemius and tibialis anterior muscles when CoPP was given either during ischemia or at the time of reperfusion. Delaying drug delivery until 1hour after the start of reperfusion abrogated this antiinflammatory effect. Conclusions CoPP can decrease skeletal muscle damage when given early in the course of ischemia/reperfusion injury and also provide protection for regenerative stem cell populations. Clinical Relevance Pharmacotherapy with HO-1 inducers, delivered in the field, on hospital arrival, or during trauma surgery, may improve preservation of muscle mass and muscle-inherent stem cells after severe ischemic limb injury.

© 2004-2018 中国地质图书馆版权所有 京ICP备05064691号 京公网安备11010802017129号

地址:北京市海淀区学院路29号 邮编:100083

电话:办公室:(+86 10)66554848;文献借阅、咨询服务、科技查新:66554700