Evolving paradigms for repair of tissues by adult stem/progenitor cells (MSCs)

Darwin J Prockop, Daniel J Kota, Nikolay Bazhanov, Roxanne L Reger, Darwin J Prockop, Daniel J Kota, Nikolay Bazhanov, Roxanne L Reger

Abstract

In this review, we focus on the adult stem/progenitor cells that were initially isolated from bone marrow and first referred to as colony forming units-fibroblastic, then as marrow stromal cells and subsequently as either mesenchymal stem cells or multipotent mesenchymal stromal cells (MSCs). The current interest in MSCs and similar cells from other tissues is reflected in over 10,000 citations in PubMed at the time of this writing with 5 to 10 new publications per day. It is also reflected in over 100 registered clinical trials with MSCs or related cells (http//www.clinicaltrials.gov). As a guide to the vast literature, this review will attempt to summarize many of the publications in terms of three paradigms that have directed much of the work: an initial paradigm that the primary role of the cells was to form niches for haematopoietic stem cells (paradigm I); a second paradigm that the cells repaired tissues by engraftment and differentiation to replace injured cells (paradigm II); and the more recent paradigm that MSCs engage in cross-talk with injured tissues and thereby generate microenvironments or 'quasi-niches' that enhance the repair tissues (paradigm III).

© 2010 The Authors Journal compilation © 2010 Foundation for Cellular and Molecular Medicine/Blackwell Publishing Ltd.

Figures

Fig 1
Fig 1
Schematic summarizing three evolving paradigms for the repair of tissues by MSCs. The morphology of a small number of adherent cells from bone marrow suggested the paradigm that the cells served as a niche for haematopoietic cells (paradigm I). The ready differentiation of the cells in culture suggested that the cells could repair tissues by engrafting and differentiating (paradigm II). Clinical trials using the cells to improve bone marrow transplants unexpectedly demonstrated that they improved graft-versus-host diseases in a few patients and thereby drew attention to their immune modulatory properties. Functional improvement without significant engraftment in animal models and a few patients suggested that MSCs enhanced repair by forming microenvironments or ‘quasi-niches’ (paradigm III).
Fig 2
Fig 2
Effects of human MSCs and recombinant TSG-6 in mice (NOD/scid) with myocardial infarcts (MI). (A) Schematic illustrating the progressive damage to the myocardium following myocardial infarction. The ischemia triggers invasion by inflammatory cells. The inflammatory cells and the matrix metalloproteinases they release accentuate damage to the myocardium. TSG-6 synthesized by MSCs or recombinant TSG-6 limits the injury and thereby enhances repair. Reproduced with permission and modified from [100]. (B) Protective/reparative properties of MSCs and TSG-6 in MI. Three weeks after permanent ligation of the anterior descending coro nary artery in mice to produce MI, each heart was cut from the apex through base into over 400 sequential 5 μm sections and stained with Masson Trichrome. Every 20th section is shown. Symbols: Normal, naïve mice; –, MI only; hMSCs, 2 × 106 hMSCs infused intravenously (i.v.) 1 hr after MI; scr siRNA, 2 × 106 hMSCs transduced with scrambled siRNA infused i.v. 1 hr after MI; TSG-6 siRNA, 2 × 106 hMSCs transduced with TSG-6 siRNA infused i.v. 1 hr after MI; rhTSG-6, 30 μg recombinant TSG-6 protein infused i.v. 1 hr and again 24 hrs after MI. Reproduced with permission from [62].
Fig 3
Fig 3
Schematic for MSCs providing a niche for haematopoietic stem cells as in paradigm I and modulating excessive inflammatory and immune responses as in paradigm III. One effect of administered MSCs is to introduce a negative feedback into the excessive responses of tissues to sterile injury. They may also enhance repair by increasing propagation and differentiation of tissue endogenous stem/progenitor cells (not shown). Some of the therapeutic effects of MSCs may require direct cell-to-cell contact and transfer of components such as mitochondria.

References

    1. Owen M, Friedenstein AJ. Stromal stem cells: marrow-derived osteogenic precursors. Ciba Found Symp. 1988;136:42–60.
    1. Allen TD, Dexter TM. Long term bone marrow cultures: an ultrastructural review. Scan Electron Microsc. 1983;4:1851–66.
    1. Caplan AI. Mesenchymal stem cells. J Orthop Res. 1991;9:641–50.
    1. Dominici M, Le Blanc K, Mueller I, et al. Minimal criteria for defining multipotent mesenchymal stromal cells. The International Society for Cellular Therapy position statement. Cytotherapy. 2006;8:315–7.
    1. Eaves CJ, Cashman JD, Sutherland HJ, et al. Molecular analysis of primitive hematopoietic cell proliferation control mechanisms. Ann NY Acad Sci. 1991;628:298–306.
    1. Whetton AD, Dexter TM. Influence of growth factors and substrates on differentiation of haemopoietic stem cells. Curr Opin Cell Biol. 1993;5:1044–9.
    1. Sacchetti B, Funari A, Michienzi S, et al. Self-renewing osteoprogenitors in bone marrow sinusoids can organize a hematopoietic microenvironment. Cell. 2008;131:324–36.
    1. Koç ON, Gerson SL, Cooper BW, et al. Rapid hematopoietic recovery after coinfusion of autologous-blood stem cells and culture-expanded marrow mesenchymal stem cells in advanced breast cancer patients receiving high-dose chemotherapy. J Clin Oncol. 2000;18:307–16.
    1. Prockop DJ. Marrow stromal cells as stem cells for nonhematopoietic tissue. Science. 1997;276:71–4.
    1. Pittenger MF, Mackay AM, Beck SC, et al. Multilineage potential of adult human mesenchymal stem cells. Science. 1999;284:143–7.
    1. Colter DC, Class R, DiGirolamo CM, et al. Rapid expansion of recycling stem cells in cultures of plastic-adherent cells from human bone marrow. Proc Natl Acad Sci USA. 2000;97:3213–8.
    1. Bruder SP, Fink DJ, Caplan AI. Mesenchymal stem cells in bone development, bone repair, and skeletal regeneration therapy. J Cell Biochem. 1994;56:283–94.
    1. Pereira RF, Halford KW, O’Hara MD, et al. Cultured adherent cells from marrow can serve as long-lasting precursor cells for bone, cartilage, and lung in irradiated mice. Proc Natl Acad Sci USA. 1995;92:4857–61.
    1. Pereira RF, O’Hara MD, Laptev AV, et al. Marrow stromal cells as a source of progenitor cells for nonhematopoietic tissues in transgenic mice with a phenotype of osteogenesis imperfecta. Proc Natl Acad Sci USA. 1998;95:1142–7.
    1. Kopen GC, Prockop DJ, Phinney DG. Marrow stromal cells migrate throughout forebrain and cerebellum, and they differentiate into astrocytes after injection into neonatal mouse brains. Proc Natl Acad Sci USA. 1999;96:10711–6.
    1. Horwitz EM, Prockop DJ, Fitzpatrick LA, et al. Transplantability and therapeutic effects of bone marrow-derived mesenchymal cells in children with osteogenesis imperfecta. Nat Med. 1999;5:309–13.
    1. Koç ON, Peters C, Aubourg P, et al. Bone marrow-derived mesenchymal stem cells remain host-derived despite successful hematopoietic engraftment after allogeneic transplantation in patients with lysosomal and peroxisomal storage diseases. Exp Hematol. 2000;27:1675–81.
    1. Lee RH, Hsu SC, Munoz J, et al. A subset of human rapidly self-renewing marrow stromal cells preferentially engraft in mice. Blood. 2006;107:2153–61.
    1. Krause DS. Bone marrow-derived lung epithelial cells. Proc Am Thorac Soc. 2008;5:699–702.
    1. Kuhn HG, Cooper-Kuhn CM. Bromodeoxyuridine and the detection of neurogenesis. Curr Pharm Biotechnol. 2007;8:127–31.
    1. Coyne TM, Marcus AJ, Woodbury D, et al. Marrow stromal cells transplanted to the adult brain are rejected by an inflammatory response and transfer donor labels to host neurons and glia. Stem Cells. 2006;24:2483–92.
    1. Rubin H. Multistage carcinogenesis in cell culture. Dev Biol. 2001;106:61–6.
    1. Tolar J, Nauta AJ, Osborn MJ, et al. Sarcoma derived from cultured mesenchymal stem cells. Stem Cells. 2007;25:371–9.
    1. Foudah D, Redaelli S, Donzelli E, et al. Monitoring the genomic stability of in vitro cultured rat bone-marrow-derived mesenchymal stem cells. Chromosome Res. 2009;17:1025–39.
    1. Furlani D, Li W, Pittermann E, et al. A transformed cell population derived from cultured mesenchymal stem cells has no functional effect after transplantation into the injured heart. Cell Transplant. 2008;18:319–31.
    1. Serhan CN, Chiang N, Van Dyke TE. Resolving inflammation: dual anti-inflammatory and pro-resolution lipid mediators. Nat Rev Immunol. 2008;8:349–61.
    1. Mestas J, Hughes CC. Of mice and not men: differences between mouse and human immunology. J Immunol. 2008;172:2731–8.
    1. Ban TA. The role of serendipity in drug discovery. Dialogues Clin Neurosci. 2006;8:335–44.
    1. Morris CDW. Acetylsalicyclic acid and platelet. Sickness. Lancet. 1967;289:279–80.
    1. Patrono C, Rocca B. Aspirin, 110 years later. J Thromb Haemost. 2009;7:258–61.
    1. Thomas ED, Blume KG. Historical markers in the development of allogeneic hematopoietic cell transplantation. Biol Blood Marrow Transplant. 1999;5:341–6.
    1. Fleisch H. Development of bisphosphonates. Breast Cancer Res. 2002;4:30–4.
    1. Bryan NS, Bian K, Murad F. Discovery of the nitric oxide signaling pathway and targets for drug development. Front Biosci. 2009;14:1–18.
    1. Ghofrani HA, Osterloh IH, Grimminger F. Sildenafil: from angina to erectile dysfunction to pulmonary hypertension and beyond. Nat Rev Drug Discov. 2006;5:689–702.
    1. Mankani MH, Kuznetsov SA, Wolfe RM, et al. In vivo bone formation by human bone marrow stromal cells: reconstruction of the mouse calvarium and mandible. Stem Cells. 2006;24:2140–9.
    1. Horie M, Sekiya I, Muneta T, et al. Intra-articular injected synovial stem cells differentiate into meniscal cells directly and promote meniscal regeneration without mobilization to distant organs in rat massive meniscal defect. Stem Cells. 2009;27:878–87.
    1. Deschaseaux F, Pontikoglou C, Sensébé L. Bone regeneration: the stem/progenitor cells point of view. J Cell Mol Med. 2010;14:103–15.
    1. Krause U, Harris S, Green A, et al. Pharmaceutical modulation of canonical Wnt signaling in multipotent stromal cells for improved osteoinductive therapy. Proc Natl Acad Sci USA. 2010;107:4147–52.
    1. Oshima Y, Harwood FL, Coutts RD, et al. Variation of mesenchymal cells in polylactic acid scaffold in an osteochondral repair model. Tissue Eng C Methods. 2009;15:595–604.
    1. Fukuda K. Regeneration of cardiomyocytes from bone marrow: use of mesenchymal stem cell for cardiovascular tissue engineering. Cytotechnology. 2003;41:165–75.
    1. Quevedo HC, Hatzistergos KE, Oskouei BN, et al. Allogeneic mesenchymal stem cells restore cardiac function in chronic ischemic cardiomyopathy via trilineage differentiating capacity. Proc Natl Acad Sci USA. 2009;106:14022–7.
    1. Stamm C, Nasseri B, Choi YH, et al. Cell therapy for heart disease: great expectations, as yet unmet. Heart Lung Circ. 2009;18:245–56.
    1. McBride C, Gaupp D, Phinney DG. Quantifying levels of transplanted murine and human mesenchymal stem cells in vivo by real-time PCR. Cytotherapy. 2003;5:7–18.
    1. Muñoz-Elias G, Marcus AJ, Coyne TM, et al. Adult bone marrow stromal cells in the embryonic brain: engraftment, migration, differentiation, and long-term survival. J Neurosci. 2004;24:4585–95.
    1. Tatard VM, D’Ippolito G, Diabira S, et al. Neurotrophin-directed differentiation of human adult marrow stromal cells to dopaminergic-like neurons. Bone. 2007;40:360–73.
    1. Phinney DG, Prockop DJ. Concise review: mesenchymal stem/multipotent stromal cells: the state of transdifferentiation and modes of tissue repair – current views. Stem Cells. 2007;25:2896–902.
    1. Parr AM, Tator CH, Keating A. Bone marrow-derived mesenchymal stromal cells for the repair of central nervous system injury. Bone Marrow Transplant. 2007;40:609–19.
    1. Munoz JR, Stoutenger BR, Robinson AP, et al. Human stem/progenitor cells from bone marrow promote neurogenesis of endogenous neural stem cells in the hippocampus of mice. Proc Natl Acad Sci USA. 2005;102:18171–6.
    1. Ohtaki H, Ylostalo JH, Foraker JE, et al. Stem/progenitor cells from bone marrow decrease neuronal death in global ischemia by modulation of inflammatory/immune responses. Proc Natl Acad Sci USA. 2008;105:14638–43.
    1. Chopp M, Zhang XH, Li Y, et al. Spinal cord injury in rat: treatment with bone marrow stromal cell transplantation. Neuroreport. 2000;11:3001–5.
    1. Hofstetter CP, Schwarz EJ, Hess D, et al. Marrow stromal cells form guiding strands in the injured spinal cord and promote recovery. Proc Natl Acad Sci USA. 2002;99:2199–204.
    1. Abrams MB, Dominguez C, Pernold K, et al. Multipotent mesenchymal stromal cells attenuate chronic inflammation and injury-induced sensitivity to mechanical stimuli in experimental spinal cord injury. Restor Neurol Neurosci. 2007;27:307–21.
    1. Pereira RF, O’Hara MD, Laptev AV, et al. Marrow stromal cells as a source of progenitor cells for nonhematopoietic tissues in transgenic mice with a phenotype of osteogenesis imperfecta. Proc Natl Acad Sci USA. 1998;95:1142–7.
    1. Chopp M, Li Y, Zhang ZG. Mechanisms underlying improved recovery of neurological function after stroke in the rodent after treatment with neurorestorative cell-based therapies. Stroke. 2009;40:S143–5.
    1. Krause U, Harter C, Seckinger A, et al. Intravenous delivery of autologous mesenchymal stem cells limits infarct size and improves left ventricular function in the infarcted porcine heart. Stem Cells Dev. 2007;16:31–7.
    1. Tögel F, Westenfelder C. Stem cells in acute kidney injury repair. Minerva Urol Nefrol. 2009;61:205–13.
    1. Lee RH, Seo MJ, Reger RL, et al. Multipotent stromal cells from human marrow home to and promote repair of pancreatic islets and renal glomeruli in diabetic NOD/scid mice. Proc Natl Acad Sci USA. 2006;103:17438–43.
    1. Ezquer F, Ezquer M, Simon V, et al. Endovenous administration of bone-marrow-derived multipotent mesenchymal stromal cells prevents renal failure in diabetic mice. Biol Blood Marrow Transplant. 2009;15:1354–65.
    1. Gao J, Dennis JE, Muzic RF, et al. The dynamic in vivo distribution of bone marrow-derived mesenchymal stem cells after infusion. Cells Tissues Organs. 2001;169:12–20.
    1. Schrepfer S, Deuse T, Reichenspurner H, et al. Stem cell transplantation: the lung barrier. Transplant Proc. 2007;39:573–6.
    1. Kidd S, Spaeth E, Dembinski JL, et al. Direct evidence of mesenchymal stem cell tropism for tumor and wounding microenvironments using in vivo bioluminescent imaging. Stem Cells. 2009;27:2614–23.
    1. Lee RH, Pulin AA, Seo MJ, et al. Intravenous hMSCs improve myocardial infarction in mice because cells embolized in lung are activated to secrete the anti-inflammatory protein TSG-6. Cell Stem Cell. 2009;5:54–63.
    1. Lee JW, Fang X, Gupta N, et al. Allogeneic human mesenchymal stem cells for treatment of E. coli endotoxin-induced acute lung injury in the ex vivo perfused human lung. Proc Natl Acad Sci USA. 2009;106:16357–62.
    1. Oh JY, Kim MK, Shin MS, et al. The anti-inflammatory and anti-angiogenic role of mesenchymal stem cells in corneal wound healing following chemical injury. Stem Cells. 2008;26:1047–55.
    1. Timmers L, Lim SK, Arslan F, et al. Reduction of myocardial infarct size by human mesenchymal stem cell conditioned medium. Stem Cell Res. 2007;1:129–37.
    1. Haynesworth SE, Baber MA, Caplan AI. Cytokine expression by human marrow-derived mesenchymal progenitor cells in vitro: effects of dexamethasone and IL-1 alpha. J Cell Physiol. 1996;166:585–92.
    1. Caplan AI, Dennis JE. Mesenchymal stem cells as trophic mediators. J Cell Biochem. 2006;98:1076–84.
    1. Ylöstalo J, Bazhanov N, Prockop DJ. Reversible commitment to differentiation by human multipotent stromal cells in single-cell-derived colonies. Exp Hematol. 2008;36:1390–402.
    1. Gunn WG, Conley A, Deininger L, et al. A crosstalk between myeloma cells and marrow stromal cells stimulates production of DKK1 and interleukin-6: a potential role in the development of lytic bone disease and tumor progression in multiple myeloma. Stem Cells. 2006;24:986–91.
    1. Ren G, Zhang L, Zhao X, et al. Mesenchymal stem cell-mediated immunosuppression occurs via concerted action of chemokines and nitric oxide. Cell Stem Cell. 2008;2:141–50.
    1. Romieu-Mourez R, François M, Boivin MN, et al. Cytokine modulation of TLR expression and activation in mesenchymal stromal cells leads to a proinflammatory phenotype. J Immunol. 2009;18212:963–73.
    1. Wisniewski HG, Vilcek J. Cytokine-induced gene expression at the crossroads of innate immunity, inflammation and fertility: TSG-6 and PTX3/TSG-14. Cytokine Growth Factor Rev. 2004;15:129–46.
    1. Mahoney DJ, Mikecz K, Ali T, et al. TSG-6 regulates bone remodeling through inhibition of osteoblastogenesis and osteoclast activation. J Biol Chem. 2008;283:25952–62.
    1. Halkos ME, Zhao ZQ, Kerendi F, et al. Intravenous infusion of mesenchymal stem cells enhances regional perfusion and improves ventricular function in a porcine model of myocardial infarction. Basic Res Cardiol. 2008;103:525–36.
    1. Iso Y, Spees JL, Serrano C, et al. Multipotent human stromal cells improve cardiac function after myocardial infarction in mice without long-term engraftment. Biochem Biophys Res Commun. 2007;354:700–6.
    1. Wolf D, Reinhard A, Seckinger A, et al. Regenerative capacity of intravenous autologous, allogeneic and human mesenchymal stem cells in the infarcted pig myocardium-complicated by myocardial tumor formation. Scand Cardiovasc J. 2009;43:39–45.
    1. Mirotsou M, Zhang Z, Deb A, et al. Secreted frizzled related protein 2 (Sfrp2) is the key Akt-mesenchymal stem cell-released paracrine factor mediating myocardial survival and repair. Proc Natl Acad Sci USA. 2007;104:1643–8.
    1. Hung SC, Pochampally RR, Chen SC, et al. Angiogenic effects of human multipotent stromal cell conditioned medium activate the PI3K-Akt pathway in hypoxic endothelial cells to inhibit apoptosis, increase survival, and stimulate angiogenesis. Stem Cells. 2007;25:2363–70.
    1. Block GJ, Ohkouchi S, Fung F, et al. Multipotent stromal cells (MSCs) are activated to reduce apoptosis in part by upregulation and secretion of stanniocalcin-1 (STC-1) Stem Cells. 2009;27:670–81.
    1. Wang Y, Huang L, Abdelrahim M, et al. Stanniocalcin-1 suppresses superoxide generation in macrophages through induction of mitochondrial UCP2. J Leukoc Biol. 2009;86:981–8.
    1. Huang L, Garcia G, Lou Y, et al. Anti-inflammatory and renal protective actions of stanniocalcin-1 in a model of anti-glomerular basement membrane glomerulonephritis. Am J Pathol. 2009;174:1368–78.
    1. Le Blanc K, Rasmusson I, Sundberg B, et al. Treatment of severe acute graft-versus-host disease with third party haploidentical mesenchymal stem cells. Lancet. 2004;363:1439–41.
    1. Uccelli A, Moretta L, Pistoia V. Mesenchymal stem cells in health and disease. Nat Rev Immunol. 2008;8:726–36.
    1. Bernardo ME, Locatelli F, Fibbe WE. Mesenchymal stromal cells. Ann NY Acad Sci. 2009;1176:101–17.
    1. Ren G, Helwani FM, Verma S, et al. Species variation in the mechanisms of mesenchymal stem cell-mediated immunosuppression. Stem Cells. 2009;27:1954–62.
    1. Rafei M, Helwani FM, Verma S, et al. Mesenchymal stromal cells ameliorate experimental autoimmune encephalomyelitis by inhibiting CD4 Th17 T cells in a CC chemokine ligand 2-dependent manner. J Immunol. 2009;182:5994–6002.
    1. François M, Romieu-Mourez R, Stock-Martineau S, et al. Mesenchymal stromal cells cross-present soluble exogenous antigens as part of their antigen-presenting cell properties. Blood. 2009;114:2632–8.
    1. English K, Ryan JM, Tobin L, et al. Cell contact, prostaglandin E(2) and transforming growth factor beta 1 play non-redundant roles in human mesenchymal stem cell induction of CD4+CD25(High) forkhead box P3+ regulatory T cells. Clin Exp Immunol. 2009;156:149–60.
    1. Shoelson SE, Herrero L, Naaz A. Obesity, inflammation, and insulin resistance. Gastroenterology. 2007;132:2169–80.
    1. Theuma P, Fonseca VA. Inflammation, insulin resistance, and atherosclerosis. Metab Syndr Relat Disord. 2004;2:105–13.
    1. Shi S, Gronthos S. Perivascular niche of postnatal mesenchymal stem cells in human bone marrow and dental pulp. J Bone Miner Res. 2003;18:696–704.
    1. Crisan M, Yap S, Casteilla L, et al. A perivascular origin for mesenchymal stem cells in multiple human organs. Cell Stem Cell. 2008;3:301–13.
    1. Díaz-Flores L, Gutiérrez R, Madrid JF, et al. Pericytes. Morphofunction, interactions and pathology in a quiescent and activated mesenchymal cell niche. Histol Histopathol. 2009;24:909–69.
    1. Krueger M, Bechmann I. CNS pericytes: concepts, misconceptions, and a way out. Glia. 2010;58:1–10.
    1. Spees JL, Olson SD, Whitney MJ, et al. Mitochondrial transfer between cells can rescue aerobic respiration. Proc Natl Acad Sci USA. 2006;103:1283–8.
    1. Park IH, Daley GQ. Human iPS cell derivation/reprogramming. Curr Protoc Stem Cell Biol. 2009;4:1–8.
    1. Kidd S, Spaeth E, Klopp A, et al. The (in) auspicious role of mesenchymal stromal cells in cancer: be it friend or foe. Cytotherapy. 2008;10:657–67.
    1. Garcia S, Martìn MC, De La Fuente R, et al. Pitfalls in spontaneous in vitro transformation of human mesenchymal stem cells. Exp Cell Res. 2010;316:1648–50.
    1. Fang L, Gao XM, Moore XL, et al. Differences in inflammation, MMP activation and collagen damage account for gender difference in murine cardiac rupture following myocardial infarction. J Mol Cell Cardiol. 2007;43:535–44.
    1. Prockop DJ. Repair of tissues by adult stem/progenitor cells (MSCs): controversies, myths, and changing paradigms. Mol Ther. 2009;17:939–46.

Source: PubMed

3
Subscribe