当前位置: 首页 > 期刊 > 《干细胞学杂志》 > 2004年第6期 > 正文
编号:11340461
a Department of Hepatology, Chancellor’s Building, University of Edinburgh, Edinburgh, Scotland;
http://www.100md.com 《干细胞学杂志》
     Potential of Hematopoietic Stem Cell Therapy in Hepatology: A Critical Review

    Steven Massona, David J. Harrisonb, John N. Plevrisa, Philip N. Newsomea,bKey Words. Bone marrow ? Cell differentiation ? Cell fusion ? Hematopoietic stem cells Hepatocytes ? Liver regeneration ? Stem cells

    Correspondence: Philip Newsome, M.D., Ph.D., Department of Hepatology, Chancellor’s Building, University of Edinburgh, Edinburgh EH16 4SB, U.K. Telephone: 0044-131-242-1712; Fax: 0044-131-242-1633; e-mail: P.Newsome@ed.ac.uk

    ABSTRACT

    Presently, orthotopic liver transplantation is the major therapeutic option for patients with acute and chronic end-stage liver disease. However, a shortage of suitable donor organs and requirement for immunosuppression restrict its usage, highlighting the need to find suitable alternatives.

    A novel and exciting approach could be offered through the current developments in the field of stem cell biology. In the past few years, multiple studies have demonstrated that adult stem cell plasticity is far greater and complex than previously thought, raising expectations that it could lay the foundations for new cellular therapies in regenerative medicine. In this review, the evidence for adult stem cell plasticity will be discussed with respect to hepatology, covering experimental models in animal and human tissues along with a discussion of the factors, putative mechanisms involved, current controversies, and potential clinical implications.

    HEMATOPOIETIC STEM CELLS: MULTILINEAGE PLASTICITY

    The adult liver has a remarkable regenerative capacity, with mature hepatocytes proliferating rapidly in response to most mild-to-moderate liver injuries and an intrahepatic stem cell compartment contributing in more severe injury. Experimental models of partial hepatectomy or carbon tetrachloride (CCl4) toxicity indicate that regeneration can occur wholly through proliferation of mature hepatocytes. However, in models of more severe injury, such as CCl4 combined with the hepatocarcinogen 2-acetylaminofluorene (2-AAF), which inhibits mature hepatocyte proliferation, additional cellular components contribute. After such injury, large numbers of small oval cells appear, which are able to differentiate into both hepatocytes and ductular cells . Similarly, addition of the DNA alkylating agent retrorsine to CCl4 gives rise to progenitors that express phenotypic characteristics of, but are morphologically distinct from, oval cells . This has led investigators to establish the concept that liver regeneration occurs on three separate levels: hepatocytes, intrahepatic stem cells, and extrahepatic stem cells .

    In the first account of hepatic transdifferentiation, lethally irradiated rats underwent cross-sex or cross-strain bone marrow transplantation followed by administration of 2-AAF to suppress hepatocyte proliferation and CCl4 to induce hepatic injury. Examination of the host liver demonstrated hepatic cells that were donor derived, as detected by expression of dipeptidyl peptidase IV enzyme (DPPIV+) in DPPIV– rats or the Y chromosome in female animals . This led to the conclusion that marrow-derived cells could act as progenitors for hepatic cells, albeit in a model of injury in which the replicative capacity of mature host hepatocytes was impaired.

    However, liver repopulation by marrow-derived cells has also been seen in the absence of any intentional liver injury. Bone marrow from male donors was infused into irradiated female mice, and the liver tissue was analyzed by fluorescent in situ hybridization (FISH) for the Y chromosome and albumin mRNA, demonstrating significant levels of donor-derived hepatocytes . Further work, again in mice, indicated that a single male HSC transplanted into an irradiated female recipient demonstrated diverse differentiative potential. In addition to hepatic engraftment, epithelial cells from throughout the gastrointestinal tract, bronchus, and skin were donor derived . Notably, in multiorgan engraftment, highest levels and most diffuse clustering of donor cells were seen in alveolar epithelium. The authors postulated that the observed differences in engraftment may relate to the injury induced by irradiation, because lung tissue is known to be more radiosensitive . It remains unclear whether irradiation injury is relevant in the hepatic models, because although radiation-induced liver damage is known to occur , it is usually seen with larger doses than those used in such preparative regimens. Additionally, there was no histological evidence of tissue damage in either study, raising the possibility that a contribution of bone marrow cells may occur even in physiological maintenance or minimal liver injury.

    Other groups have been unable to reproduce these findings. Chimeric animals were generated by transplantation of a single green fluorescent protein (GFP)–marked HSC into sublethally irradiated mice in the absence of specific tissue injury. Single HSCs resulted in significant hematopoietic engraftment, but hepatocytes were produced at a frequency of only approximately 1 in 70,000 cells . Furthermore, GFP+:GFP– parabiotic mice with a common circulatory system were created, surgically enabling evaluation of circulating stem cells and HSC engraftment in a model that does not even require irradiation. Despite successful hematopoietic cross-engraftment, there was no engraftment of non-hematopoietic tissue, leading the authors to conclude that HSCs played no role in the production of nonhematopoietic cells under physiological conditions. A different group used various liver injury models to assess hepatic regeneration after gender-mismatched bone marrow transplantation. No significant contribution was demonstrated .

    Nevertheless, several other studies seem to corroborate adult stem cell plasticity, with moderate to severe injury increasing the level of hepatic transdifferentiation, a finding augmented additionally in model systems in which the donor HSCs have a survival advantage . The significance of such a survival advantage is confirmed in a transgenic model based on the protective effect of the antiapoptotic gene, Bcl-2, against Fas-mediated cell death. Bone marrow from mice expressing this transgene, under the control of a liver-specific promoter, was infused into normal mice. Some mice underwent repeated injections with a Fas-agonist antibody to induce liver injury, whereas others did not. Only those that had received antibody injections showed mature hepatocytes expressing Bcl-244, implying that transdifferentiation is inefficient under physiological conditions and that tissue injury such as accumulation of toxic catabolites or apoptotic challenge is required to generate a more robust response. It is interesting to note that a recent study of the contribution of HSCs in a model of liver fibrosis demonstrated higher than previously reported levels of marrow-derived hepatocytes. Transgenic mice expressing GFP were used as a source of bone marrow, and up to 26% of the recipient liver was repopulated by 4 weeks . For the first time, donor bone marrow cells without a survival advantage resulted in robust and efficient regeneration, although there has been some concern raised regarding ambiguous identification and unusual architecture of the reporter cells .

    Although the presence and severity of liver injury may be important in regulating the extent of stem cell plasticity and engraftment, the reported variation in the literature is still marked (Table 1). Detailed analysis of the models used demonstrates that different subpopulations of stem cells may have different levels of functional plasticity. Although initial studies mainly used unfractionated bone marrow, subsequent work highlighted the capacity of CD34+ cells for hepatic engraftment . More recently, the SP fraction of marrow cells was demonstrated to contain cells with similar ability , and yet it is rich in CD34– stem cells . Indeed, the authors of these many studies have recognized that supposedly minimal differences in experimental methods may be responsible for the observed discrepancies. Such factors have been discussed in more detail in recent commentaries .

    Table 1. Stem cell–derived hepatocytic differentiation in animal models

    DO STEM CELL–DERIVED HEPATOCYTES HAVE FUNCTIONAL SIGNIFICANCE?

    The characterization of stem cell biology in animal work is clearly important, but clinical application requires convincing evidence that human stem cells also share the properties demonstrated by adult rodent stem cells. Cellular phenotypes of human hepatic stem cells and tissue reactions similar to those seen in animal models have been described in a variety of human acute and chronic liver diseases .

    The first reports implying transdifferentiation in human cells used archival biopsies, in which liver specimens from recipients of sex-mismatched bone marrow or liver transplants were analyzed for marrow-derived hepatic cell types. Using immunohistochemistry with FISH staining for the Y chromosome, analysis of liver and bone marrow samples revealed that hepatic cell types had arisen from a marrow-derived population . Such differentiation was also reported in patients who had undergone transfusion of peripheral-blood stem cells for the treatment of hematological malignancy, suggesting that such progenitor cells circulate in the blood . In this study, epithelial cells of the skin and gastrointestinal tract were also seen to derive from cells of donor origin.

    There is some concern that Y chromosome–positive cells identified in the female livers in these studies could occur as a result of the transplacental passage of male fetal blood cells during pregnancy , because fetal–maternal microchimerism has previously been documented . However, this cannot be entirely responsible, because these male cells have been seen in at least one human liver from a nulliparous female along with a female with no history of male childbearing . In addition, in the murine models, female recipients were all nulliparous .

    Additional analysis of this archival work allows interesting comparisons with the animal models. For example, specimens from human liver allografts revealed varying degrees of injury, with mild biliary obstruction in most and fibrosing cholestatic hepatitis in one. Injury severity correlated directly with hepatic engraftment frequency , yet recipients of bone marrow transplantation also demonstrated significant hepatic engraftment despite the absence of overt hepatic injury, in keeping with the murine model previously discussed . Additionally, histology revealed that in most allografts, hepatocyte engraftment was isolated and scattered, whereas the liver with severe recurrent disease showed more extensive ductular clustering. Again, this is analogous to the animal data suggesting that, as in animals, there may be several modes of human hepatic regeneration with a potential role for adult stem cells in both physiological maintenance and acute injury.

    Indeed, this highlights the need for an in vivo system in which to study human stem cells directly (Table 2). Human cord blood cells were infused into sublethally irradiated nonobese diabetic/severe combined immunodeficient (NOD/SCID) mice, and at euthanasia, murine livers were analyzed for the presence of human hepatic cells. Despite the absence of intentional liver damage, human-derived hepatocytes were detected in murine liver . Using a similar xenogeneic transplantation model, engrafted human cells were seen to produce human albumin mRNA, as detected by reverse transcription–polymerase chain reaction .

    Table 2. Hepatocyte differentiation of hematopoietic cells in human cord blood in immunodeficient mice

    The role of liver injury in such models has also been reported. Hepatic injury, induced by one-third partial hepatectomy and 2-AAF after infusion of human cord blood cells, led to the identification of functional hepatocytes . Notably, in another xenogeneic model comparing injured and noninjured mice, only those who had been administered CCl4 were seen to express human albumin . Additionally, this study demonstrated that it was CD34+ HSCs that gave rise to this phenomenon. However, as in murine models, this remains controversial. In another study, human stem cells expressing the receptor for the complement molecule C1q (C1qRp) were infused . This is a human homologue of a mouse stem cell antigen, AA4, recognized by a monoclonal antibody used to define murine multipotent hematopoietic progenitors . Notably, C1qRp is expressed on both CD34+ and CD34– cells, suggesting that this marker may define an even more primitive subpopulation. Perhaps differences in the differentiative potential of the two populations are responsible for the conflicting data regarding the role of injury.

    Furthermore, recent studies in the NOD/SCID model demonstrate the role of the stromal cell–derived factor-1 (SDF-1)/CXC chemokine receptor 4 (CXCR4) axis in regulating the migration of HSCs to damaged liver . Intriguingly, hepatocyte growth factor, which is upregulated in liver injury and has been shown to augment hepatocytic differentiation of engrafted HSCs , was shown to play a key role in recruiting stem cells via its interaction with SDF-1 . Some of the engrafted human cells differentiated into albumin-producing hepatocyte-like cells. Another group has used the SCID model to examine different mechanistic factors. Consistent downregulation of ?2 microglobluin (?2M), an integral part of the major histocompatibility complex, was observed early after stem cell transplantation in liver tissue in which human albumin expression was upregulated . It was suggested that this switching off of ?2M may be an important mechanism in escaping the host immune system. Although there is clearly much to be established regarding these mechanistic factors, human blood-derived cells can, under strictly defined circumstances perhaps similar to previous animal studies, transdifferentiate into hepatic cell types, and liver injury influences the observed differentiation. As yet, there are no data to demonstrate that human stem cells have therapeutic potential, but this xenogeneic transplantation model will be useful in studying human adult stem cell biology further.

    THE ROLE OF CELL FUSION

    Despite uncertainty surrounding the mechanisms underlying adult stem cell plasticity, there is much speculation regarding potential clinical implications. Enthusiasm has been heightened by pioneering clinical trials in other disciplines. For example, in cardiology, the delivery of marrow-derived cells into the coronary circulation of human subjects was reported to improve blood flow and cardiac function in ischemic myocardium . In hepatology, the data presented here provide hope that somatic stem cells could eventually be used in tissue replacement protocols for the treatment of inherited and acquired end-stage liver diseases.

    Use of adult stem cells overcomes many of the moral and ethical barriers of ES cell manipulation, and if somatic cells genuinely can switch lineage barriers, then HSCs are an ideal source. There is already considerable experience in their handling, and they are relatively accessible. Additionally, their administration may induce immunological tolerance, because they may potentially induce hematopoietic microchimerism, thus obviating the need for immunosuppression. Rather than relying on cadaveric organs from deceased donors who are often immunologically disparate, HSCs offer ready availability of liver-repopulating stem cells or progenitors obtained from living donors.

    Despite such enthusiasm, at present there remains significant uncertainty as to what such cells would accomplish in the clinical setting, and there are many issues to be addressed before translation into clinical practice. First, a better understanding of the mechanisms that modulate the role of HSCs in physiological maintenance and liver injury is required. Specifically, more work is required to establish whether HSCs may play a therapeutic role in chronic liver disease, which is a more clinically relevant target. Second, the mechanism of genomic plasticity needs to be further defined. If transdifferentiation truly is responsible, then there could be wide-ranging utility for a range of acquired liver diseases, whereas if fusion is responsible, then this could be exploited to deliver corrective genes for hepatic metabolic disorders, as long as genetic stability in the reprogrammed cells could be assured. However, given the great disparity in levels of transdifferentiation reported and controversy regarding the mechanism responsible, these issues seem far from resolved and bring into question therapeutic strategies based on this idea. Additionally, much work is needed to assess practical issues; if HSCs are capable of engrafting directly as hepatocytes, then direct intraorgan injection may facilitate a more robust response and obviate the need for irradiation to deplete the recipient’s bone marrow. Alternatively, if marrow engraftment is a prerequisite, then peripheral administration with marrow preparation and manipulation of the hepatic microenvironment to maximally attract HSCs will likely be required.

    In conclusion, the new findings in adult stem cell biology are transforming our understanding of tissue repair with promising hopes of regenerative hepatology. However, perhaps we should remain cautious at present. Adult stem cell plasticity does occur, but it is a rare event even under selective pressure, and it remains to be seen whether this will be clinically significant in the human context.

    REFERENCES

    Loeffler M, Roeder I. Tissue stem cells: definition, plasticity, heterogeneity, self-organization and models: a conceptual approach. Cells Tissues Organs 2002;171:8–26.

    Thomson JA, Itskovitz-Eldor J, Shapiro SS et al. Embryonic stem cell lines derived from human blastocysts. Science 1998;282:1145–1147.

    Shamblott MJ, Axelman J, Wang S et al. Derivation of pluripotent stem cells from cultured human primordial germ cells. Proc Natl Acad Sci U S A 1998;95:13726–13731.

    Booth C, Potten CS. Gut instincts: thoughts on intestinal epithelial stem cells. J Clin Invest 2000;105:1493–1499.

    Gage FH. Mammalian neural stem cells. Science 2000;287:1433–1438.

    Beauchamp JR, Heslop L, Yu DS et al. Expression of CD34 and Myf5 defines the majority of quiescent adult skeletal muscle satellite cells. J Cell Biol 2000;151:1221–1234.

    Taylor G, Lehrer MS, Jensen PJ et al. Involvement of follicular stem cells in forming not only the follicle but also the epidermis. Cell 2000;102:451–461.

    Prockop DJ. Marrow stromal cells as stem cells for non-hematopoietic tissues. Science 1997;276:71–74.

    Bianco P, Riminucci M, Gronthos S et al. Bone marrow stromal stem cells: nature, biology, and potential applications. STEM CELLS 2001;19:180–192.

    Osawa M, Hanada K, Hamada H et al. Long-term lympho-hematopoietic reconstitution by a single CD34-low/negative hematopoietic stem cell. Science 1996;273:242–245.

    Goodell MA, Rosenzweig M, Kim H et al. Dye efflux studies suggest that hematopoietic stem cells expressing low or undetectable levels of CD34 antigen exist in multiple species. Nat Med 1997;3:1337–1345.

    Yin AH, Miraglia S, Zanjani ED et al. AC133, a novel marker for human hematopoietic stem and progenitor cells. Blood 1997;90:5002–5012.

    Goodell MA, Brose K, Paradis G et al. Isolation and functional properties of murine hematopoietic stem cells that are replicating in vivo. J Exp Med 1996;183:1797–1806.

    Zhou S, Schuetz JD, Bunting KD et al. The ABC transporter Bcrp1/ABCG2 is expressed in a wide variety of stem cells and is a molecular determinant of the side-population phenotype. Nat Med 2001;7:1028–1034.

    Scharenberg CW, Harkey MA, Torok-Storb B. The ABCG2 transporter is an efficient Hoechst 33342 efflux pump and is preferentially expressed by immature human hematopoietic progenitors. Blood 2002;99:507–512.

    Shimano K, Satake M, Okaya A et al. Hepatic oval cells have the side population phenotype defined by expression of ATP-binding cassette transporter ABCG2/BCRP1. Am J Pathol 2003;163:3–9.

    Ros JE, Libbrecht L, Geuken M et al. High expression of MDR1, MRP1, and MRP3 in the hepatic progenitor cell compartment and hepatocytes in severe human liver disease. J Pathol 2003;200:553–560.

    Ferrari G, Cusella-De Angelis G, Coletta M et al. Muscle regeneration by bone marrow-derived myogenic progenitors. Science 1998;279:1528–1530.

    Orlic D, Kajstura J, Chimenti S et al. Bone marrow cells regenerate infarcted myocardium. Nature 2001;410:701–705.

    Jackson KA, Majka SM, Wang H et al. Regeneration of ischemic cardiac muscle and vascular endothelium by adult stem cells. J Clin Invest 2001;107:1395–1402.

    Orlic D, Kajstura J, Chimenti S et al. Mobilized bone marrow cells repair the infarcted heart, improving function and survival. Proc Natl Acad Sci U S A 2001;98:10344–10349.

    Jackson KA, Mi T, Goodell MA. Hematopoietic potential of stem cells isolated from murine skeletal muscle. Proc Natl Acad Sci U S A 1999;96:14482–14486.

    Gussoni E, Soneoka Y, Strickland CD et al. Dystrophin expression in the mdx mouse restored by stem cell transplantation. Nature 1999;401:390–394.

    Mezey E, Chandross KJ, Harta G et al. Turning blood into brain: cells bearing neuronal antigens generated in vivo from bone marrow. Science 2000;290:1779–1782.

    Brazelton TR, Rossi FM, Keshet GI et al. From marrow to brain: expression of neuronal phenotypes in adult mice. Science 2000;290:1775–1779.

    Bjornson CR, Rietze RL, Reynolds BA et al. Turning brain into blood: a hematopoietic fate adopted by adult neural stem cells in vivo. Science 1999;283:534–537.

    Clarke DL, Johansson CB, Wilbertz J et al. Generalized potential of adult neural stem cells. Science 2000;288:1660–1663.

    Wagers AJ, Sherwood RI, Christensen JL et al. Little evidence for developmental plasticity of adult hematopoietic stem cells. Science 2002;297:2256–2259.

    Weimann JM, Charlton CA, Brazelton TR et al. Contribution of transplanted bone marrow cells to Purkinje neurons in human adult brains. Proc Natl Acad Sci U S A 2003;100:2088–2093.

    Alvarez-Dolado M, Pardal R, Garcia-Verdugo JM et al. Fusion of bone-marrow-derived cells with Purkinje neurons, cardiomyocytes and hepatocytes. Nature 2003;425:968–973.

    Rabes HM, Wirsching R, Tuczek HV et al. Analysis of cell cycle compartments of hepatocytes after partial hepatecomy. Cell Tissue Kinet 1976;9:517–532.

    Stowell RE, Less CS. Histochemical studies of mouse liver after single feeding of carbon tetrachloride. Arch Pathol 1950;50:519–537.

    Farber E. Similarities in the sequence of early histological changes induced in the liver of the rat by ethionine, 2-acetylamino-fluorene, and 3'-methyl-4-dimethylaminoazobenzene. Cancer Res 1956;16:142–148.

    Yin L, Lynch D, Ilic Z et al. Proliferation and differentiation of ductular progenitor cells and littoral cells during the regeneration of the rat liver to CCl4/2-AAF injury. Histol Histopathol 2002;17:65–81.

    Gordon GJ, Coleman WB, Hixson DC et al. Liver regeneration in rats with retrorsine-induced hepatocellular injury proceeds through a novel cellular response. Am J Pathol 2000;156:607–619.

    Sell S. Heterogeneity and plasticity of hepatocyte lineage cells. Hepatology 2001;33:738–750.

    Petersen BE, Bowen WC, Patrene KD et al. Bone marrow as a potential source of hepatic oval cells. Science 1999;284:1168–1170.

    Theise ND, Badve S, Saxena R et al. Derivation of hepatocytes from bone marrow cells in mice after radiation-induced myeloablation. Hepatology 2000;31:235–240.

    Krause DS, Theise ND, Collector MI et al. Multi-organ, multi-lineage engraftment by a single bone marrow-derived stem cell. Cell 2001;105:369–377.

    Travis EL, Peters LJ, McNeill J et al. Effect of dose-rate on total body irradiation: lethality and pathologic findings. Radiother Oncol 1985;4:341–351.

    Lawrence TS, Robertson JM, Anscher MS et al. Hepatic toxicity resulting from cancer treatment. Int J Radiat Oncol Biol Phys 1995;31:1237–1248.

    Kanazawa Y, Verma IM. Little evidence of bone marrow-derived hepatocytes in the replacement of injured liver. Proc Natl Acad Sci U S A 2003;100(suppl 1):11850–11853.

    Lagasse E, Connors H, Al Dhalimy M et al. Purified hematopoietic stem cells can differentiate into hepatocytes in vivo. Nat Med 2000;6:1229–1234.

    Mallet VO, Mitchell C, Mezey E et al. Bone marrow transplantation in mice leads to a minor population of hepatocytes that can be selectively amplified in vivo. Hepatology 2002;35:799–804.

    Terai S, Sakaida I, Yamamoto N et al. An in vivo model for monitoring transdifferentiation of bone marrow cells into functional hepatocytes. J Biochem (Tokyo) 2003;134:551–558.

    McTaggart RA, Feng S. An uncomfortable silence em leader while we all search for a better reporter gene in adult stem cell biology. Hepatology 2004;39:1143–1146.

    Wulf GG, Luo KL, Jackson KA et al. Cells of the hepatic side population contribute to liver regeneration and can be replenished with bone marrow stem cells. Haematologica 2003;88:368–378.

    Theise ND, Krause DS, Sharkis S. Comment on "Little evidence for developmental plasticity of adult hematopoietic stem cells" Science 2003;299:1317.

    Wagers AJ, Sherwood RI, Christensen JL et al. Response to comment on "little evidence for developmental plasticity of adult hematopoietic stem cells." Science 2003;299:1317b.

    Dahlke MH, Popp FC, Bahlmann FH et al. Liver regeneration in a retrorsine/CCl4-induced acute liver failure model: do bone marrow-derived cells contribute? J Hepatol 2003;39:365–373.

    Lowes KN, Brennan BA, Yeoh GC et al. Oval cell numbers in human chronic liver diseases are directly related to disease severity. Am J Pathol 1999;154:537–541.

    Crosby HA, Hubscher S, Fabris L et al. Immunolocalization of putative human liver progenitor cells in livers from patients with end-stage primary biliary cirrhosis and sclerosing cholangitis using the monoclonal antibody OV-6. Am J Pathol 1998;152:771–779.

    Theise ND, Saxena R, Portmann BC et al. The canals of Hering and hepatic stem cells in humans. Hepatology 1999;30:1425–1433.

    Theise ND, Nimmakayalu M, Gardner R et al. Liver from bone marrow in humans. Hepatology 2000;32:11–16.

    Alison MR, Poulsom R, Jeffery R et al. Hepatocytes from non-hepatic adult stem cells. Nature 2000;406:257.

    Korbling M, Katz RL, Khanna A et al. Hepatocytes and epithelial cells of donor origin in recipients of peripheral-blood stem cells. N Engl J Med 2002;346:738–746.

    McDonnell WM. Liver stem cells from bone marrow. Hepatology 2000;32:1181.

    Nelson JL, Furst DE, Maloney S et al. Microchimerism and HLA-compatible relationships of pregnancy in scleroderma. Lancet 1998;351:559–562.

    Tanaka A, Lindor K, Gish R et al. Fetal microchimerism alone does not contribute to the induction of primary biliary cirrhosis. Hepatology 1999;30:833–838.

    Newsome PN, Johannessen I, Boyle S et al. Human cord blood-derived cells can differentiate into hepatocytes in the mouse liver with no evidence of cellular fusion. Gastroenterology 2003;124:1891–1900.

    Ishikawa F, Drake CJ, Yang S et al. Transplanted human cord blood cells give rise to hepatocytes in engrafted mice. Ann N Y Acad Sci 2003;996:174–185.

    Kakinuma S, Tanaka Y, Chinzei R et al. Human umbilical cord blood as a source of transplantable hepatic progenitor cells. STEM CELLS 2003;21:217–227.

    Wang X, Ge S, McNamara G et al. Albumin-expressing hepatocyte-like cells develop in the livers of immune-deficient mice that received transplants of highly purified human hematopoietic stem cells. Blood 2003;101:4201–4208.

    Danet GH, Luongo JL, Butler G et al. C1qRp defines a new human stem cell population with hematopoietic and hepatic potential. Proc Natl Acad Sci U S A 2002;99:10441–10445.

    Petrenko O, Beavis A, Klaine M et al. The molecular characterization of the fetal stem cell marker AA4. Immunity 1999;10:691–700.

    Kollet O, Shivtiel S, Chen YQ et al. HGF, SDF-1, and MMP-9 are involved in stress-induced human CD34+ stem cell recruitment to the liver. J Clin Invest 2003;112:160–169.

    Armbrust T, Batusic D, Xia L et al. Early gene expression of hepatocyte growth factor in mononuclear phagocytes of rat liver after administration of carbon tetrachloride. Liver 2002;22:486–494.

    Beerheide W, von Mach MA, Ringel M et al. Downregulation of beta2-microglobulin in human cord blood somatic stem cells after transplantation into livers of SCID-mice: an escape mechanism of stem cells? Biochem Biophys Res Commun 2002;294:1052–1063.

    Terada N, Hamazaki T, Oka M et al. Bone marrow cells adopt the phenotype of other cells by spontaneous cell fusion. Nature 2002;416:542–545.

    Ying QL, Nichols J, Evans EP et al. Changing potency by spontaneous fusion. Nature 2002;416:545–548.

    Vassilopoulos G, Wang PR, Russell DW. Transplanted bone marrow regenerates liver by cell fusion. Nature 2003;422:901–904.

    Wang X, Willenbring H, Akkari Y et al. Cell fusion is the principal source of bone-marrow-derived hepatocytes. Nature 2003;422:897–901.

    Sigal SH, Rajvanshi P, Gorla GR et al. Partial hepatectomy-induced polyploidy attenuates hepatocyte replication and activates cell aging events. Am J Physiol 1999;276:G1260–G1272.

    Guha C, Sharma A, Gupta S et al. Amelioration of radiation-induced liver damage in partially hepatectomized rats by hepatocyte transplantation. Cancer Res 1999;59:5871–5874.

    Vignery A. Osteoclasts and giant cells: macrophage-macrophage fusion mechanism. Int J Exp Pathol 2000;81:291–304.

    Camargo FD, Chambers SM, Goodell MA. Stem cell plasticity: from transdifferentiation to macrophage fusion. Cell Prolif 2004;37:55–65.

    Camargo FD, Finegold M, Goodell MA. Hematopoietic myelomonocytic cells are the major source of hepatocyte fusion partners. J Clin Invest 2004;113:1266–1270.

    Jang YY, Collector MI, Baylin SB et al. Hematopoietic stem cells convert into liver cells within days without fusion. Nat Cell Biol 2004;6:532–539.

    Ianus A, Holz GG, Theise ND et al. In vivo derivation of glucose-competent pancreatic endocrine cells from bone marrow without evidence of cell fusion. J Clin Invest 2003;111:843–850.

    Tran SD, Pillemer SR, Dutra A et al. Differentiation of human bone marrow-derived cells into buccal epithelial cells in vivo: a molecular analytical study. Lancet 2003;361:1084–1088.

    Jiang Y, Jahagirdar BN, Reinhardt RL et al. Pluripotency of mesenchymal stem cells derived from adult marrow. Nature 2002;418:41–49.

    Grompe M, Lindstedt S, Al Dhalimy M et al. Pharmacological correction of neonatal lethal hepatic dysfunction in a murine model of hereditary tyrosinaemia type I. Nat Genet 1995;10:453–460.

    Blau HM. A twist of fate. Nature 2002;419:437.

    Schwartz RE, Reyes M, Koodie L et al. Multipotent adult progenitor cells from bone marrow differentiate into functional hepatocyte-like cells. J Clin Invest 2002;109:1291–1302.

    Assmus B, Schachinger V, Teupe C et al. Transplantation of progenitor cells and regeneration enhancement in acute myocardial infarction (TOPCARE-AMI). Circulation 2002;106:3009–3017.

    Tse HF, Kwong YL, Chan JK et al. Angiogenesis in ischaemic myocardium by intramyocardial autologous bone marrow mononuclear cell implantation. Lancet 2003;361:47–49.(b Centre for Inflammation)