当前位置: 首页 > 期刊 > 《干细胞学杂志》 > 2004年第1期 > 正文
编号:11342476
Limited Contribution of Circulating Cells to the Development and Maintenance of Nonhematopoietic Bovine Tissues
http://www.100md.com 《干细胞学杂志》
     a Division of Anatomy and

    b Division of Biochemistry, Department of Basic Veterinary Sciences, University of Helsinki, Helsinki, Finland

    Key Words. Stem cells ? Blood circulation ? Regeneration ? Chimera ? Freemartinism ? Y chromosome ? In situ hybridization

    Antti Iivanainen, D.V.M., Ph.D., Department of Basic Veterinary Sciences, FIN-00014 University of Helsinki, Helsinki, Finland. Telephone 358-9-191-49544; Fax: 358-9-191-49799; e-mail: Antti.Iivanainen@helsinki.fi

    ABSTRACT

    The differentiation potential of stem cells (SCs) in adult mammals is currently a topic of intense debate. Many recent reports have challenged the traditional dogma of strictly restricted differentiation options of adult cells. Bone marrow (BM) cells, hematopoietic stem cells (HSCs), or mesenchymal stem cells (MSCs) have been shown to differentiate into cells of the skeletal muscle , liver , epithelia of skin, lung, and gastrointestinal tract and even to cells of the central nervous system (CNS) . However, the physiological significance of the findings is uncertain. The cell culture techniques and transplantation methods applied are likely to induce changes both in the cells and recipient tissues . Also, heterogeneity of the transplanted cell populations , potential cell fusion events , and problems in reproducing some of the results complicate interpretation of transdifferentiation experiments.

    Circulating cells derived from BM or other sources may have a role in tissue repair . A traditional model of regenerating tissue is a subcutaneously implanted cellulose viscose sponge . The sponge is invaded by irregular, richly vascularized granulation tissue similar to that forming in a healing wound. Granulocytes and macrophages first enter the sponge, followed by fibroblasts, and a rapidly increasing collagen synthesis and angiogenesis in a few days . The origin of the granulation tissue producing cells is unclear, with several contradictory studies supporting either a local or systemic source .

    We have used freemartin cattle to monitor the contribution of prenatally circulating cells to the development, physiological maintenance, and repair of bovine tissues. Freemartin is a female calf born as a twin to a bull. About 90% of cattle twins are chimeric due to placental anastomoses that form early in the development and permit the exchange of blood and other cells between the fetuses . The twins permanently share identical composite blood types and are mutually tolerant to allografts from each other . The proportions of donor-derived nucleated cells in blood and BM of different animals are randomly distributed between less than 5% and more than 95% . The freemartins develop normally, apart from the inner sex organs, which are masculinized to a variable degree probably due to hormonal influences . Donor-derived cells in freemartin tissues can be readily identified by their Y chromosomes. Thus, the freemartin is a suitable model for studies of SC transfer in an unmanipulated large mammal, and also unique in allowing the investigation of prenatal events in a chimeric animal.

    Freemartin tissues have formerly been analyzed by cytogenetic techniques , but little is known about the contribution of donor-derived cells to the development, growth, and cell dynamics of the non-hematopoietic organs. We have previously characterized the freemartin immune system by Y-chromosome-targeted in situ hybridization (Y-ISH) , which allows the identification of bull-derived cells in intact tissue sections. Y-ISH can be combined with immunohistochemistry or other staining methods to investigate the phenotypes of cells. In this paper, we describe an extensive mapping of freemartin tissues using Y-ISH combined with CD45 immunohistochemistry and mistletoe lectin I (ML-I) histochemistry to exclude cells of the hematopoietic lineage. CD45 is a cell surface antigen exclusively expressed by all nucleated cells of the hematopoietic lineage . ML-I labels bovine microglia .

    MATERIALS AND METHODS

    We first performed a wide Y-ISH screening of tissues from 12 freemartins (ages: 20 days to 20 months). Samples from 20 to 74 different body sites per animal were analyzed. Y-chromosome positive (Y+) bull-derived cells were detected in all tissues examined in all 12 animals. The highest frequencies of Y+ cells were found in blood, BM, and lymphatic tissues. These tissues also exhibited considerable variation between individual animals. Among blood mononuclear cells, the proportion of Y+ cells varied from approximately 10% to 90%. In other organs, the variation was less pronounced.

    The distribution of Y+ cells suggested that a significant proportion of them were infiltrating leukocytes, which often are difficult to distinguish reliably from parenchymal cells in a standard histological staining. To label the leukocytes residing in the tissues, we combined Y-ISH and CD45 immunohistochemistry or, in the case of CNS, ML-I histochemistry for bovine microglial cells . We re-examined all major tissue types and derivatives of all embryonic layers. The double staining confirmed that most Y+ cells indeed were of the hematopoietic lineage (Fig. 1A–1G). CD45/ML-I-Y+ cells were identified in most tissues, but in 11 of 12 calves (208 succesfully double-stained sections from 14 different tissues) they were mostly sporadic. No groups of CD45/ML-I-Y+ cells indicating local clonal expansion were seen in these calves. However, one of the calves (FM10) was a striking exception, with high numbers of CD45/ML-I-Y+ cells in several tissues. This case is discussed more thoroughly below.

    Figure 1. Bull-derived (Y+) cells in freemartin tissues. Y-chromosome targeted in situ hybridization combined with anti-leukocyte staining (MLI histochemistry for brain, anti-CD45 immunohistochemistry for other tissues). A-G) physiological freemartin tissues: A) cerebral cortex; B) renal cortex; C) liver; D) mammary gland, duct epithelium; E) mammary gland, connective tissue around ducts; F) skeletal muscle; G) mucosa of the small intestine; H) intestinal mucosa from FM10, showing exceptionally strong donor contribution to intestinal epithelium; I) granulation tissue in a cellulose viscose sponge, incubation time 4 weeks. White arrowheads: CD45/ML-I-Y+ cells. Black arrowheads: CD45/ML-I+Y+ cells. Asterisk: a chimeric foreign body giant cell. Bars: 25 μm.

    The donor contribution to the nonhematopoietic cells in various tissues was estimated by counting the proportion of CD45/ML-I-Y+ cells in 4 to 5 calves per tissue (including the exceptional case). Altogether, several tissues were analyzed from all 12 freemartins. Apart from FM10, the calves did not differ markedly in the relative frequency of CD45/ML-I-Y+ cells, which constituted less than 1% of all CD45/ML-I- cells in most tissues investigated (Fig. 2). There was no obvious correlation between the frequency of Y+ cells in blood and in nonhematopoietic tissues, or between different nonhematopoietic tissues in an animal. Donor-derived nonhematopoietic cells were markedly rare in the ectoderm-derived epidermis and CNS. Otherwise, no consistent differences between derivatives of different embryonic layers were seen. In 11 of 12 calves, highest numbers of CD45/ML-I-Y+ cells were detected in the connective tissue around ducts of mammary glands (0.94 ± 0.67%), liver (0.85 ± 0.41%, primarily not hepatocytes), and skeletal muscle (0.83 ± 0.26%). In cardiac and smooth muscle, renal epithelia, and epidermis the average frequency of CD45-Y+ cells was 0.1% or less (in epidermis the interdigitating Langerhans cells make the identification of CD45- cells difficult). In the brain, very few ML-I- donor-derived cells were found, although donor contribution to the microglia was in most cases substantial (we note that in addition to microglial cells, the lectin labels most endothelia). In the cerebral cortex and the underlying white matter, 0.0020 ± 0.0023% of ML-I- cells were Y+. In the hippocampus and the OB, two regions involved in the generation of new neurons in adult mammals , no ML-I-Y+ cells were detected in approximately 2 x 105 cells screened in double-stained sections of each tissue. No Y+ cells with clearly neuronal morphology were seen in about 720 successfully hybridized sections from freemartin CNS, representing approximately 28 x 106 total cells and including samples from cerebral cortex, cerebellum, brain stem, corpus callosum, hippocampus, OB, thalamus, ventricular walls, hypophysis, and spinal cord.

    Figure 2. Proportions of bull-derived (Y+) cells in all nonhematopoietic (CD45/ML-I-) cells in 11 majority-type freemartins. Each tissue was analyzed from four majority-type calves (not the same calves for different tissues), with the exception of the mammary gland (three samples containing ducts available), and the regenerating tissue in implants (one sample per time point). Numbers were corrected with counts obtained from similar bull tissues as described in Materials and Methods. EP = epithelium, CT = connective tissue, MU = muscle tissue, NE = nervous tissue. For each tissue, individual samples are arranged in order of increasing animal age.

    In FM10, the distribution of CD45/ML-I-Y+ cells differed dramatically from the rest of the calves. Higher numbers of CD45-Y+ cells were found in the epithelium of small intestine (19%, Fig. 1H), connective tissue of mammary gland (15%), liver (4.3%), epithelium of mammary gland (4.0%), skeletal muscle (2.3%), cardiac muscle (1.0%), and smooth muscle (0.5%), whereas in brain and in renal epithelia the frequency did not differ notably from the other calves. In the villi of the intestinal mucosa, strands of CD45-Y+ cells were seen (Fig. 1H). The hematopoietic system of FM10 differed from the majority of calves in that the donor contribution to B cells was clearly weaker than to the total leukocyte pool (not shown).

    To investigate the role of donor-derived cells in tissue regeneration, we implanted cellulose viscose sponges in the subcutis of a 70-day-old freemartin and stained the invading granulation tissue for Y-chromosome and CD45. The regenerating tissue contained relatively high numbers of CD45-Y+ cells, with increasing numbers within the incubation period of 4 weeks (Figs. 1I, 2). At 4 weeks, the frequency of CD45-Y+ cells in the regenerating tissue was significantly higher than in most other nonhematopoietic tissues of the same animal (p = 0.12 for liver, p < 0.02 for any other tissue investigated). The frequencies of CD45/ML-I-Y+ cells in the tissues of the recipient did not differ markedly from those in the 10 other calves of the majority type. No clear Y+ cell clusters indicating local expansion were seen.

    DISCUSSION

    We thank M. Georges (University of Liège, Belgium) for the btDYZ clone, U. Pfüller (Witten/Herdecke University, Germany) for the biotinylated mistletoe lectin I, staff at the Saari unit (Dept. Clinical Veterinary Medicine, University of Helsinki) for providing blood samples of potential freemartins, K. Lahti, T. Pankasalo, H. Valtonen, and A. Koivunen for expert technical assistance, and L.C. Andersson and L.A. Lindberg for valuable comments about the work.

    REFERENCES

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

    LaBarge MA, Blau HM. Biological progression from adult bone marrow to mononucleate muscle stem cell to multinucleate muscle fiber in response to injury. Cell 2002;111:589–601.

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

    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.

    Okamoto R, Yajima T, Yamazaki M et al. Damaged epithelia regenerated by bone marrow-derived cells in the human gastrointestinal tract. Nat Med 2002;8:1011–1017.

    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.

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

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

    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.

    Priller J, Persons DA, Klett FF et al. Neogenesis of cerebellar Purkinje neurons from gene-marked bone marrow cells in vivo. J Cell Biol 2001;155:733–738.

    Morshead CM, Benveniste P, Iscove NN et al. Hematopoietic competence is a rare property of neural stem cells that may depend on genetic and epigenetic alterations. Nat Med 2002;8:268–273.

    Hagemann RF, Sigdestad CP, Lesher S. Intestinal crypt survival and total and per crypt levels of proliferative cellularity following irradiation: single x-ray exposures. Radiat Res 1971;46:533–546.

    d’Avella D, Cicciarello R, Albiero F et al. Quantitative study of blood-brain barrier permeability changes after experimental whole-brain radiation. Neurosurgery 1992;30:30–34.

    Orkin SH, Zon LI. Hematopoiesis and stem cells: plasticity versus developmental heterogeneity. Nat Immunol 2002;3:323–328.

    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.

    Castro RF, Jackson KA, Goodell MA et al. Failure of bone marrow cells to transdifferentiate into neural cells in vivo. Science 2002;297:1299.

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

    Forbes SJ, Vig P, Poulsom R et al. Adult stem cell plasticity: new pathways of tissue regeneration become visible. Clin Sci (Lond) 2002;103:355–369.

    Viljanto J, Kivikoski A. Local effect of neutral salt-soluble collagen on formation of granulation tissue—a histological study with viscose cellulose implants on rats. Ann Med Exp Biol Fenn 1962;40:118–127.

    Holund B, Junker P, Garbarsch C et al. Formation of granulation tissue in subcutaneously implanted sponges in rats. A comparison between granulation tissue developed in viscose cellulose sponges (Visella) and in polyvinyl alcohol sponges (Ivalon). Acta Pathol Microbiol Scand 1979;87:367–374.

    Lampiaho K, Kulonen E. Metabolic phases during the development of granulation tissue. Biochem J 1967;105:333–341.

    Ross R, Everett NB, Tyler R. Wound healing and collagen formation. VI. The origin of the wound fibroblast studied in parabiosis. J Cell Biol 1970;44:645–654.

    Bucala R, Spiegel LA, Chesney J et al. Circulating fibrocytes define a new leukocyte subpopulation that mediates tissue repair. Mol Med 1994;1:71–81.

    Lillie FR. The free-martin; a study of the action of sex hormones in the foetal life of cattle. J Exp Zool 1917;23:371–452.

    Nisbet NW. Parabiosis in immunobiology. Transplant Rev 1973;15:123–161.

    Owen RD. Immunogenetic consequenses of vascular anastomoses between bovine twins. Science 1945;102:400–401.

    Anderson D, Billingham RE, Lampkin GH et al. The use of skin grafting to distinguish between monozygotic and dizygotic twins in cattle. Heredity 1951;5:379–397.

    Billingham RE, Lampkin GH, Medawar PB et al. Tolerance to homografts, twin diagnosis, and the freemartin condition in cattle. Heredity 1952;6:201–212.

    Wilkes PR, Wijeratne WV, Munro IB. Reproductive anatomy and cytogenetics of freemartin heifers. Vet Rec 1981;108:349–353.

    Vigier B, Tran D, Legeai L et al. Origin of anti-Mullerian hormone in bovine freemartin fetuses. J Reprod Fertil 1984;70:473–479.

    Niku M, Pessa-Morikawa T, Andersson L et al. Oligoclonal Peyer’s patch follicles in the terminal small intestine of cattle. Dev Comp Immunol 2002;26:689–695.

    Pessa-Morikawa T, Niku M, Iivanainen A. Persistent differences in the level of chimerism in B versus T cells of freemartin cattle. Dev Comp Immunol 2004;28:77–87.

    Thomas ML. The leukocyte common antigen family. Annu Rev Immunol 1989;7:339–369.

    Hewicker-Trautwein M, Schultheis G, Trautwein G. Demonstration of amoeboid and ramified microglial cells in pre- and postnatal bovine brains by lectin histochemistry. Anat Anz 1996;178:25–31.

    Perret J, Shia YC, Fries R et al. A polymorphic satellite sequence maps to the pericentric region of the bovine Y chromosome. Genomics 1990;6:482–490.

    Modi WS, Gallagher DS, Womack JE. Molecular organization and chromosomal localization of six highly repeated DNA families in the bovine genome. Anim Biotechnol 1993;4:143–161.

    Skowronski J, Plucienniczak A, Bednarek A et al. Bovine 1.709 satellite. Recombination hotspots and dispersed repeated sequences. J Mol Biol 1984;177:399–416.

    Hopman AH, Ramaekers FC, Speel EJ. Rapid synthesis of biotin-, digoxigenin-, trinitrophenyl-, and fluorochrome-labeled tyramides and their application for In situ hybridization using CARD amplification. J Histochem Cytochem 1998;46:771–777.

    Altman J, Das GD. Autoradiographic and histological evidence of postnatal hippocampal neurogenesis in rats. J Comp Neurol 1965;124:319–335.

    Lois C, Alvarez-Buylla A. Long-distance neuronal migration in the adult mammalian brain. Science 1994;264:1145–1148.

    Morshead CM, Reynolds BA, Craig CG et al. Neural stem cells in the adult mammalian forebrain: a relatively quiescent subpopulation of subependymal cells. Neuron 1994;13:1071–1082.

    Palmer TD, Takahashi J, Gage FH. The adult rat hippocampus contains primordial neural stem cells. Mol Cell Neurosci 1997;8:389–404.

    Fukushima N, Yokouchi K, Kawagishi K et al. Differential neurogenesis and gliogenesis by local and migrating neural stem cells in the olfactory bulb. Neurosci Res 2002;44:467–473.

    Amano T, Inamura T, Wu CM et al. Effects of single low dose irradiation on subventricular zone cells in juvenile rat brain. Neurol Res 2002;24:809–816.

    Bissonnette TH. The development of the reproductive ducts and canals in the freemartin with comparison of the normal. Am J Anat 1924;33:267–345.

    Ohno S, Gropp A. Embryological basis for germ cell chimerism in mammals. Cytogenetics 1965;4:251–261.

    Rüsse I, Sinowatz F. In: Rüsse I, eds. Lehrbuch der Embryologie der Haustiere. Berlin: Parey, 1991:159–168.

    Butler H, Juurlink BHJ. An Atlas for Staging Mammalian and Chick Embryos. Boca Raton: CRC Press, 1987:1–218.

    Rangan SR. Origin of the fibroblastic growths in chicken buffy coat macrophage cultures. Exp Cell Res 1967;46:477–487.

    Petrakis NL, Lucia SP, Davis M. The in vivo differentiation of human leukocytes into histiocytes, fibroblasts and fat cells in subcutaneous diffusion chambers. Blood 1961;17:109–118.

    Abe R, Donnelly SC, Peng T et al. Peripheral blood fibrocytes: differentiation pathway and migration to wound sites. J Immunol 2001;166:7556–7562.

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

    K?rbling 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.

    Laflamme MA, Myerson D, Saffitz JE et al. Evidence for cardiomyocyte repopulation by extracardiac progenitors in transplanted human hearts. Circ Res 2002;90:634–640.

    Kleeberger W, Versmold A, Rothamel T et al. Increased chimerism of bronchial and alveolar epithelium in human lung allografts undergoing chronic injury. Am J Pathol 2003;162:1487–1494.

    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 USA 2003;100:2088–2093.

    Campagnoli C, Roberts IA, Kumar S et al. Identification of mesenchymal stem/progenitor cells in human first-trimester fetal blood, liver, and bone marrow. Blood 2001;98:2396–2402.

    Wright DE, Wagers AJ, Gulati AP et al. Physiological migration of hematopoietic stem and progenitor cells. Science 2001;294:1933–1936.

    Zvaifler NJ, Marinova-Mutafchieva L, Adams G et al. Mesenchymal precursor cells in the blood of normal individuals. Arthritis Res 2000;2:477–488.

    Gallacher L, Murdoch B, Wu D et al. Identification of novel circulating human embryonic blood stem cells. Blood 2000;96:1740–1747.

    Owen M. Marrow stromal stem cells. J Cell Sci Suppl 1988;10:63–76.

    Pittenger MF, Mackay AM, Beck SC et al. Multilineage potential of adult human mesenchymal stem cells. Science 1999;284:143–147.

    Friedenstein AJ, Petrakova KV, Kurolesova AI et al. Heterotopic transplants of bone marrow. Analysis of precursor cells for osteogenic and hematopoietic tissues. Transplantation 1968;6:230–247.

    Reyes M, Lund T, Lenvik T et al. Purification and ex vivo expansion of postnatal human marrow mesodermal progenitor cells. Blood 2001;98:2615–2625.

    Jiang Y, Vaessen B, Lenvik T et al. Multipotent progenitor cells can be isolated from postnatal murine bone marrow, muscle, and brain. Exp Hematol 2002;30:896–904.

    Simmons PJ, Przepiorka D, Thomas ED et al. Host origin of marrow stromal cells following allogeneic bone marrow transplantation. Nature 1987;328:429–432.

    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;5:309–313.

    Van den Heuvel RL, Versele SR, Schoeters GE et al. Stromal stem cells (CFU-f) in yolk sac, liver, spleen and bone marrow of pre- and postnatal mice. Br J Haematol 1987;66:15–20.

    Trier JS, Moxey PC. Morphogenesis of the small intestine during fetal development. Ciba Found Symp 1979;70:3–29.

    Ponder BA, Schmidt GH, Wilkinson MM et al. Derivation of mouse intestinal crypts from single progenitor cells. Nature 1985;313:689–691.

    Turner CW. The anatomy of the mammary gland of cattle. I. Embryonic development. Mo Agr Exp Sta Res Bul 1930;140:5–34.

    Hennighausen L, Robinson GW. Signaling pathways in mammary gland development. Dev Cell 2001;1:467–475.

    Kordon EC, Smith GH. An entire functional mammary gland may comprise the progeny from a single cell. Development 1998;125:1921–1930.

    Smith GH, Chepko G. Mammary epithelial stem cells. Microsc Res Tech 2001;52:190–203.(Mikael Nikua, Lotta Ilmon)