You are on page 1of 28

15524892, 2004, 1, Downloaded from https://anatomypubs.onlinelibrary.wiley.com/doi/10.1002/ar.a.10134 by Cochrane Romania, Wiley Online Library on [31/10/2022].

See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
THE ANATOMICAL RECORD PART A 276A:75–102 (2004)

Adult Stem Cells


HENRY E. YOUNG1,2* AND ASA C. BLACK, JR.1,3
1
Division of Basic Medical Sciences, Mercer University School of Medicine,
Macon, Georgia
2
Department of Pediatrics, Mercer University School of Medicine, Macon, Georgia
3
Department of Obstetrics and Gynecology, Mercer University School of Medicine,
Macon, Georgia

ABSTRACT
Development of a multicellular organism is accomplished through a series of events that
are preprogrammed in the genome. These events encompass cellular proliferation, lineage
commitment, lineage progression, lineage expression, cellular inhibition, and regulated ap-
optosis. The sequential progression of cells through these events results in the formation of
the differentiated cells, tissues, and organs that constitute an individual. Although most cells
progress through this sequence during development, a few cells leave the developmental
continuum to become reserve precursor cells. The reserve precursor cells are involved in the
continual maintenance and repair of the tissues and organs throughout the life span of the
individual. Until recently it was generally assumed that the precursor cells in postnatal
individuals were limited to lineage-committed progenitor cells specific for various tissues.
However, studies by Young, his colleagues, and others have demonstrated the presence of two
categories of precursor cells that reside within the organs and tissues of postnatal animals.
These two categories of precursor cells are lineage-committed (multipotent, tripotent, bipo-
tent, and unipotent) progenitor cells and lineage-uncommitted pluripotent (epiblastic-like,
ectodermal, mesodermal, and endodermal) stem cells. These reserve precursor cells provide
for the continual maintenance and repair of the organism after birth. Anat Rec Part A 276A:
75–102, 2004. © 2004 Wiley-Liss, Inc.

Key words: adult; pluripotent; stem cells; mammals; humans; embryonic; mes-
enchymal; epiblastic

The formation of tissues and organs occurs naturally dur- As development proceeds from the single-celled zygote,
ing embryogenesis (review, Moore and Persaud, 1998; Carl- proliferation of the blastomeres forms a solid sphere,
son, 1999; Kacsoh, 2000). Development of a multicellular termed the morula. Further proliferation and lineage com-
organism follows multiple predetermined molecular and cel- mitment results in formation of a blastocyst, a hollow
lular pathways that culminate in the formation of an indi- sphere of cells composed of the trophoblast, germ cells,
vidual composed of billions of cells with well-defined respon- and the inner cell mass. The trophoblast continues to
sibilities. These responsibilities include provision for develop by proliferation and segregation into the cytotro-
structural support and mobility, electrical and chemical con- phoblast and syncytiotrophoblast. These two layers even-
trol of body functions, protection through controlled interac- tually form the embryonic portion of the placenta (Fig. 1).
tions with the surrounding environment, nutrition, removal The germ cells, precursors to oogonia (female) or sper-
of waste products, and procreation. Moreover, the cells, tis-
sues, and organs must be maintained throughout the life
span of the individual as well as repaired following trauma
Grant sponsor: Rubye Ryle Smith Charitable Trust; Grant
or disease. Cellular development is accomplished through sponsor: MEDCEN Community Health Foundation; Grant spon-
the genome as a series of preprogrammed events. These sor: MorphoGen Pharmaceuticals, Inc.
events include cellular proliferation, lineage commitment, *Correspondence to: Dr. Henry E. Young, Division of Basic
lineage progression, lineage expression, cellular inhibition, Medical Science, Mercer University School of Medicine, 1550
and regulated apoptosis. Sequential progression through College St., Macon, GA 31207. Fax: 478-301-5489.
these programmed events results in the formation of the E-mail: young_he@mercer.edu
differentiated cells, tissues, and organs that fashion an indi- Received 27 January 2003; Accepted 21 February 2003
vidual. These preprogrammed processes are initiated follow- DOI 10.1002/ar.a.10134
ing fusion of the male and female pronuclei to form a totipo- Published online 00 Month 2004 in Wiley InterScience
tent zygote (Fig. 1). (www.interscience.wiley.com).

© 2004 WILEY-LISS, INC.


15524892, 2004, 1, Downloaded from https://anatomypubs.onlinelibrary.wiley.com/doi/10.1002/ar.a.10134 by Cochrane Romania, Wiley Online Library on [31/10/2022]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License

Lineage flow chart for mammalian embryogenetic development.


YOUNG AND BLACK

Fig. 1.
76
15524892, 2004, 1, Downloaded from https://anatomypubs.onlinelibrary.wiley.com/doi/10.1002/ar.a.10134 by Cochrane Romania, Wiley Online Library on [31/10/2022]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
ADULT STEM CELLS 77
matogonia (male), undergo proliferation and subsequent within the postnatal individual contains only its own
migration through various regions within the developing unique contingent of precursor cells specific for that tis-
conceptus, eventually reaching the gonadal ridges. Prolif- sue. It is further assumed that such lineage-committed
eration, lineage commitment, and segregation of cells progenitor cells function solely in the maintenance and
within the inner cell mass lead to formation of two pluri- repair of the tissue in which they reside (review, Donovan
potent layers, the epiblast and hypoblast. The epiblast and Gearhart, 2001; Forbes et al., 2002; Poulsom et al.,
and hypoblast develop further by proliferation, lineage 2002; Tsai et al., 2002).
commitment, and segregation into the primary pluripo- Young and his colleagues have been characterizing the
tent germ layers: ectoderm, mesoderm, and endoderm identities of precursor cells within the tissues of animals
(Fig. 1). The ectodermal germ layer continues its develop- of all ages, including humans. These studies began with
ment by proliferation, lineage commitment, and segrega- initial observations on the origins of the blastema cells
tion to form the surface ectoderm and neural ectoderm. during limb regeneration in terrestrial amphibians
The neural ectoderm segregates into neural tube and neu- (Young, 1977a, 1977b, 1977c, 1983; Young et al., 1983a,
ral crest. Similarly, the mesodermal germ layer continues 1983b, 1983c, 1983d, 1985, 1989a, 1989b). The studies
its development by proliferation, lineage commitment, have progressed through precursor cell identification in
and segregation to form the somitic mesoderm, interme- the connective tissues of avians (Bowerman et al., 1991;
diate mesoderm, lateral plate somatic mesoderm, and lat- Young et al., 1991, 1992a, 1992b, 1992c, 1993, 1995,
eral plate splanchnic mesoderm. The endodermal germ 1998a; Lucas et al., 1994a; Dixson et al., 1996; Young and
layer also continues its development by proliferation, lin- Lucas, 1998; Young, 2000), mammals (Shoptaw et al.,
eage commitment, and segregation. The cells of each of 1991; Lucas et al., 1992, 1993, 1994a, 1994b, 1995; Pate et
these germ layer lineages undergo further rounds of pro- al., 1993; Grande et al., 1995; Rogers et al., 1995; Ware-
liferation and segregation through lineage commitment to jcka et al., 1996; Young and Lucas, 1998; Young et al.,
form multipotent, tripotent, bipotent, and unipotent pro- 1998b, 2001a, 2004a, 2004b; Romero-Ramos et al., 2002;
genitor cell lineages that finally form the differentiated Young, 2004), and humans (Young et al., 1999, 2001b,
cells, tissues, and organs that are the end products of Young, 2004). These studies have utilized numerous tech-
these pathways of differentiation (Fig. 1). niques, including histology, energy-dispersive spectral
Although a majority of the cells progress through this analysis coupled with scanning electron microscopy, gly-
preprogrammed developmental sequence to form an indi- coconjugate histochemistry, immunocytochemistry, quali-
vidual, a few cells leave this developmental continuum to tative and quantitative enzyme-linked immunoculture as-
become reserve precursor cells. These reserve precursor says, differential isolation and plating, differential
cells are involved in the continual maintenance and repair cryopreservation, differential centrifugation, repetitive
of tissues and organs throughout the life span of the limiting serial dilution clonogenic analysis, cluster of dif-
individual. It has long been assumed (and reiterated con- ferentiation (CD) marker analysis, fluorescent-activated
tinually in review articles, most recently those concerning cell sorting, magnetic bead cell sorting, and molecular
embryonic stem cells) that the particular cells leaving the analyses for telomerase activity and gene expression.
developmental continuum to become reserve precursor These techniques have been employed in studies of prolif-
cells are limited solely to lineage-committed progenitor eration, lineage commitment, lineage progression, and lin-
cells specific for particular tissues. This assumption is eage inhibition induced by bioactive factors. These studies
based in part on the previous identification of tissue-spe- have led to the discovery that two general categories of
cific lineage-committed progenitor cells. Mauro (1961) reserve precursor cells exist within the body and are in-
identified the myosatellite myoblast, a precursor cell for volved in the maintenance and repair of tissues in adults:
the maintenance and repair of adult skeletal muscle. This lineage-committed progenitor cells and lineage-uncommit-
quiescent myogenic precursor cell is located between the ted pluripotent stem cells.
sarcolemma of the skeletal muscle cell and its adjoining Lineage-committed progenitor stem cells display cer-
basement membrane. Chondrogenic precursor cells for the tain characteristics in common (Tables 1 and 2) (Young et
maintenance and repair of cartilage were identified in the al., 1991, 1992a, 1992b, 1993, 1995, 1998a, 1998b, 1999,
perichondrial connective tissue surrounding cartilage 2001a, 2001b, 2004a; Rogers et al., 1995; Young and Lu-
(Cruess, 1982). Osteogenic precursor cells for the mainte- cas, 1998; Young, 2000, 2004). Progenitor cells may be
nance and repair of bone were identified in the outer committed to one or more specific tissue lineages. Progen-
periosteal connective tissues covering bone (Cruess, 1982), itor cells may be unipotent, forming a single cell type;
along the inner endosteal surface (Cruess, 1982), and bipotent, forming two cell types; tripotent, forming three
within the stroma of adjacent bone marrow (Owen and cell types; or multipotent, forming four or more discrete
Friedenstein, 1988). Adipogenic precursor cells for the cell types. Each progenitor cell for a particular tissue
maintenance and generation of adipose tissue have been lineage has a unique profile of cell surface cluster of dif-
identified within adult adipose tissue (Aihaud et al., ferentiation (CD) markers. Progenitor cells conform to
1992). More recently, tissue-specific precursor cells have Hayflick’s limit of 50 –70 population doublings (Hayflick,
been identified for other tissue types. Neurogenic precur- 1965) before they undergo programmed senescence and
sor cells have been located within tissues of the central cellular death. Progenitor cells are unresponsive to lin-
nervous system (Gage et al., 1995); epidermal precursor eage induction agents that have actions outside their re-
cells have been identified within the interfollicular epider- spective tissue lineage(s). For example, unipotent myo-
mis and bulge region of the hair follicle (Janes et al., blast progenitor cells are unresponsive to bone
2002); and basal epithelial cells have been identified as morphogenetic protein-2 (BMP-2), which induces cartilage
the precursor cells for the gastrointestinal mucosa and bone development. Progenitor cells are responsive to
(Bjerknes and Cheng, 2002). Based on these and other proliferation agents such as platelet-derived growth fac-
reports, it has been assumed that each tissue and/or organ tors (i.e., PDGF-BB, PDGF-AA, PDGF-AB). Progenitor
15524892, 2004, 1, Downloaded from https://anatomypubs.onlinelibrary.wiley.com/doi/10.1002/ar.a.10134 by Cochrane Romania, Wiley Online Library on [31/10/2022]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
78 YOUNG AND BLACK

TABLE 1. Postnatal precursor cells


Proliferation
Name Species Characteristics potential Differentiation potential References
Adult neuronal Rat Dependent on FGF-2 for survival, 1 year Astroglial, oligodendral Gage et al.,
progenitor cells Nestin⫹, A2B5⫹, O4⫹, GAD⫹, through 1995
NSE⫹, MAP2⫹, MAP5⫹, multiple
neuN⫺, tau⫺ passages
Adult liver- Mouse Ly5.1⫹, c-Kit⫹, Sca-1⫹, linlow/⫺ Not stated in lethally irradiated Taniguchi et
derived mice, replaced all al., 1996
hematopoietic blood cells (B cells, T
stem cells cells, granulocytes,
macrophages,
erythrocytes)
Adipo-fibroblasts Sheep Isolated by Percoll gradient Less than 2 Mature adipocytes Vierck et al.,
centrifugation weeks 1996
Mesenchymal Rat Selectively plated, released, ? 4–5 weeks Skeletal myotubes, Warejcka et
stem cells filtered, frozen and relplated adipocytes, endothelial al., 1996
cells, chondrocytes,
osteoblasts, smooth
muscle cells
Pancreatic- Mouse Dispersed islets grown to Not stated Pancreatic islets Cornelius et
derived “ . . . round cells budding containing ␣-cells, ␤- al., 1997
pluripotent upward from the epithelial cells, and ␦-cells
islet-producing monolayers . . . ”
stem cells
Neural stem cells Mouse Transgenic for lacZ, clonally Continuously B-lymphocytes, T- Bjornson et
derived (2H1), react strongly to expanded lymphocytes, myeloid al., 1999
X-Gal for cells
extended
periods of
time
SVZ astrocytes Mouse GFAP⫹, S100?⫹, vimentin⫹, Not stated TuJ1⫹, type A cells, type Doetsch et al.,
(neural stem labeled with 3H, DAP, GFP C cells, neurospheres, 1999
cells) neuroblasts
Multipotent stem Mouse Methylcellulose clonal spheres; 18–21 Form neuronal, Gritti et al.,
cell from FGFR1 & EGFR1 passages, astroglial, and 1999
subventricular ⱕ12 hour oligodendroglial cells
forebrain doubling
region time
Bone Marrow Mouse WT dystrophin gene ⫹ Not stated ⫹ Dystrophin fibers in Gussoni et al.,
Cells recipients 1999
Hematopoietic Mouse Sca-1⫹, c-Kit⫹, CD43⫹, CD45⫹, Not stated Dystrophin fibers in Gussoni et al.,
stem cells lin⫺ (B220⫺, Mac-1⫺, Gr1⫺, recipients 1999
CD4⫺, CD5⫺, CD8⫺)
Muscle side Mouse Sca-1⫹, lin⫺, c-Kit⫺, CD45⫺, Not stated myoblasts, fibroblasts, Gussoni et al.,
population CD43⫺ CD43⫹, CD45⫹, 1999
cells dystrophin ⫹
myofibers
Muscle main Mouse lin⫹ (CD11⫹, Gr1⫹, CD5⫹) Not stated Desmin ⫹ Gussoni et al.,
population 1999
cells
Ependymal- Rat, Notch-1, Decreased Olfactory bulb neurons, Johansson et
derived neural Mouse with neurons, astrocytes, al., 1999
stem cell increasing oligodendrocytes,
time after
injury
Marrow Stromal Mouse CD11b⫺, CD45⫺, BrdUrd labeled Not stated GFAP⫹, ORO⫹, Kopen et al.,
Cells BrdUrd⫹, 1999
Neurofilament⫹, Type
I & II collagen⫹,
Alizarin Red S⫹
Bone Marrow Rat Male, DPPIV⫹ Not stated Male DPPIV⫹ Petersen et
Stem Cells hepatocytes cells (also al., 1999
H4⫹, C-CAM⫹) found
in female DPPIV-
recipent
Mesenchymal Human SH2⫹, SH3⫹, CD29⫹, CD44⫹, Proliferate adipocytes, chondrocytes, Pittenger et
stem cells CD71⫹, CD90⫹, CD106⫹, extensively osteocytes al., 1999
CD120a⫹, CD124⫹, CD14⫺,
CD34⫺, CD45⫺, can be
subcloned at 4 cells per well
15524892, 2004, 1, Downloaded from https://anatomypubs.onlinelibrary.wiley.com/doi/10.1002/ar.a.10134 by Cochrane Romania, Wiley Online Library on [31/10/2022]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
ADULT STEM CELLS 79
TABLE 1. Postnatal precursor cells (continued)
Proliferation
Name Species Characteristics Potential Differentiation Potential References
Mesenchymal Rat top 2/3 of Percoll gradient Not stated Muscle, tendon, Tomita et al.,
stem cells cartilage, bone, fat, 1999
cardiomyocytes
Pluripotent Human Sorted, CD10⫹, CD13⫹, CD34⫹, ⱕ 400 Mesodermal only: Muscle Young et al.,
Mesenchymal CD56⫹, CD90⫹, MHC-I⫹ population (3), adipose (2), 1999,
Stem Cells doublings Cartilage (5), bone (2), 2001b,
Connective tissue (4), 2004a;
Endothelial cells, Young,
Hematopoietic cells 2000, 2004
Skeletal muscle Mouse CD34⫹, Myf-5⫹, M-Cadherin⫹ Not stated Skeletal muscle Beauchamp et
satellite cells al., 2000
Neural stem cells Mouse b-Gal⫹, express neomycin Not stated Integrate into developing Clarke et al.,
resistance gene, Desmin⫹, chick embryos and 2000
Myosin heavy chain⫹, clonal contribute to all germ
culture layers
Bone marrow- Human FLK-1⫹, TRK⫹, TransferrinR⫹, “Rapidly Osteogenesis, Colter et al.,
derived: small Annexin-II⫹, CD44⫹, CD49e⫹, dividing adipogenesis, 2000, 2001
agranular CD59⫹, cells” chondrogenesis
recycling cells
(RS-1)
Bone marrow- Human FLK-1⫾, TRK⫾, CD49e⫹ “Rapidly Osteogenesis, Colter et al.,
derived: small dividing adipogenesis, 2000, 2001
gran. recycling cells” chondrogenesis
cells (RS-2)
Bone marrow- Human FLK-1⫺, TRK⫺, TransferrinR⫺, “Rapidly Osteogenesis, Colter et al.,
derived: large Annexin-II⫺, CD10⫹, CD147⫹, dividing adipogenesis, 2000, 2001
and more CD44⫹, CD49e⫹, CD59⫹, cells” chondrogenesis
mature cells CD81⫹, CD90⫹
(mMSCs)
Neural stem cells Mice Clonally derived, do not express 7–8 months MyHC⫹, MyoD⫹, Galli et al.,
and any myogenic markers, Myosin⫹, Laminin⫹ 2000
Human MLC3F/nlacZ⫺,
Muscle-derived Mouse Sca-1⫹, desmin⫹, CD34⫹, Bcl- 25 passages Muscle regeneration, Lee et al.,
stem cell clone, 2⫹, Flk-1⫹, c-met⫹, MNF⫹ osteoblasts, improve 2000
MC13 bone healing
Bone marrow- Human Clones - 3 types: osteogenic, 22–23 Bone; bone and cartilage; Muraglia et
derived osteo-chondrogenic; osteo- population and bone, cartilage, al.,2000
mesenchymal chondro-adipogenic doublings and adipocytes
progenitor cells
Islet-producing Mouse Insulin I⫹, Insulin II⫹, Insulin 3 years ⫹ Islet like structures that Ramiya et al.,
stem cells receptor⫹, hepatocyte growth secrete insulin 2000
factor⫹, C-MET⫹, glucagons⫹,
somatostatin⫹, glucose
transporter-2 receptor⫹,
glutamic acid decarboxylase-
67⫹, insulin-like growth factors
I & II⫹
Bone marrow Rat CD44⫹, CD71⫹, CD90⫹ ⬎ 20 Neuron-specific enolase Woodbury et
stromal cells passages (NSE⫹), NeuN⫹, al., 2000
nestin⫹, trkA⫹,
neurofilament-M⫹,
tau⫹
Bone marrow Human Not stated Passage 2 NSE⫹, NF-M⫹, growth Woodbury et
stromal cells cones, terminal bulbs al., 2000
Blood-derived Human Buffy coat isola: CD105⫹, SDF- Not stated Fibroblasts, adipocytes, Zvaifler et al.,
Mesenchymal 1⫹ osteoblasts/bone, 2000
precursor cells osteoclasts
Bone marrow- Mouse CD34⫺/low, c-Kit⫹, Sca-1⫹ Not stated Cardiac myocytes and Jackson et al.,
derived side endothelial cells 2001
population
cells
Muscle Precursor Mouse Sca-1⫹, CD34low/⫺, desmin⫹ Not stated dystrophin expressing Jankowski et
cells myofibers al., 2001
Bone marrow- Rat CD90⫹ 4,500 cells Osteogenesis Javazon et al.,
derived: small per cell 2001
recycling stem plated
cells (refed)
15524892, 2004, 1, Downloaded from https://anatomypubs.onlinelibrary.wiley.com/doi/10.1002/ar.a.10134 by Cochrane Romania, Wiley Online Library on [31/10/2022]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
80 YOUNG AND BLACK

TABLE 1. Postnatal precursor cells (continued)


Proliferation
Name Species Characteristics Potential Differentiation Potential References
Bone marrow- Rat CD90⫹, CD59⫹ “Rapidly CFU formation, Javazon et al.,
derived: dividing osteogenic 2001
mature MSCs cells” differentiation
Marrow derived Mouse Fr25lin⫺, PKH26 labeled Not stated Incorporated onto bone Krause et al.,
stem cells marrow, stomach, 2001
esophagus, small
intestine, large
intestine, liver, lungs,
and skin
Circulating Mouse Blood via cardiac puncture, CFU Not stated Osteogenesis, Kuznetsov et
skeletal stem per 106 cells - 0.93 Adipogenesis al., 2001
cells
Circulating Rabbit Blood via cardiac puncture, CFU Not stated Osteogenesis, Kuznetsov et
skeletal stem per 106 cells - 0.18 Adipogenesis al., 2001
cells
Circulating Guinea Blood via cardiac puncture, CFU Not stated Osteogenesis, Kuznetsov et
skeletal stem Pig per 106 cells - 2.7 Adipogenesis al., 2001
cells
Circulating Human Whole blood & buffy coat conc., Not stated Osteogenesis, Kuznetsov et
skeletal stem CFU per 106 cells - rare, Adipogenesis al., 2001
cells
Neural Human Not stated 30–70⫹ cell neurons, astrocytes Palmer et al.,
progenitor cells doublings 2001
before
senescence
Multipotent Human long telomeres, CD45⫺, 70⫹ cell mesenchymal cell types, Reyes and
Adult glycophorin-A-, can be doublings cells of Verfaillie,
Progenitor subcloned at 10 cells per well neuroectodermal 2001
Cells lineage
Mesodermal Human CD45⫺, glycophorin-A⫺, CD10⫺, more than all mesodermal Reyes et al.,
progenitor cells CD31⫺, CD34⫺, CD36⫺, 50 cell phenotypes, 2001
CD38⫺, CD50⫺, CD62e⫺, doublings osteoblasts,
CD106⫺, CD117⫺, H1P12⫺, chondroblasts,
Fibroblast⫺, HLA-DR⫺, class 1 adipocytes, cells will
HLA⫺, Tie⫺, Tek⫺, b2- support hematopoiesis,
microglobulin low, CD44low, skeletal muscle,
CDw90low, KDRlow, Fltlow, endothelial cells
CD13⫹, CD49b⫹, average
telomere length of 11–15 kb,
normal karyotype
Human Human Nucleated Not stated Bone, cartilage Turgeman et
Mesenchymal al., 2001
Stem Cells
Spinal cord Rat Spinal cord gray matter isolate 24–30 hour Neurons (160 kDa-NF⫹) Vacanti et al.,
progenitor cells doubling and glia (GFAP⫹) 2001a
time, 4
weeks in
culture
Spore-like cells Rat 5 ␮m, pericellular mucopolysac- 12–36 hour “ . . . cells appear to Vacanti et al.,
charides and glycolipids doubling differentiate into cells 2001b
time specific to organ from
which they were
isolated, . . . ”
Pluripotent Rat Cloned, can be subcloned at 1 cell ⬎ 400 Mesodermal only: Muscle Young et al.,
Mesenchymal per well, Telomerase ⫹ population (3), adipose (2), 2001a,
Stem Cells doublings Cartilage (5), bone (2) 2004a;
Connective tissue (4), Young,
Endothelial cells 2000, 2004
Pluripotent Human CD10⫹, CD13⫹, CD34⫹, CD56⫹, ⬎ 400 Mesodermal only: Muscle Young et al.,
Mesenchymal CD90⫹, MHC-I⫹ population (3), adipose (2), 2001b,
Stem Cells doublings Cartilage (5), bone (2) 2004a;
Connective tissue (4), Young,
Endothelial cells, 2000, 2004
Hematopoietic cells
Adipose-derived Human Lipoaspirates: AS02⫹ (fibroblasts ⬍ 25 Adipogenic, Zuk et al.,
multi-lineage & mesenchymal cells); population chondrogenic, 2001
cells vimentin⫹ doublings myogenic, and
osteogenic cells
15524892, 2004, 1, Downloaded from https://anatomypubs.onlinelibrary.wiley.com/doi/10.1002/ar.a.10134 by Cochrane Romania, Wiley Online Library on [31/10/2022]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
ADULT STEM CELLS 81
TABLE 1. Postnatal precursor cells (continued)
Proliferation
Name Species Characteristics Potential Differentiation Potential References
Nestin-positive Rat Nestin-positive, CK19-negative Longevity in Liver cells, pancreatic Zulewski et
islet-derived culture ⬎ acinar cells, ductal al., 2001
multipotential 8 months cells, ␣-cells, ␤-cells
stem cells
(NIP)
Nestin-positive Human Nestin-positive CK19-negative Longevity in Liver cells, pancreatic Zulewski et
islet-derived culture ⬎ acinar cells, ductal al., 20010
multipotential 8 months cells, ␣-cells, ␤-cells
stem cells
(NIP)
Multipotential Mice Induced somatic mutated cells Long lived, Form parietal, zymogen, Bjerknes and
stem cells 48⫹ enteroendocrine, and Cheng, 2002
weeks mucus cell types
Muscle-derived Mouse Sca-1⫹, slowly adhering, Not stated Muscle regeneration, Deasy et al.,
stem cells desmin⫹, osteoblasts, improve 2002
bone healing,
reconstitute
hematopoietic system
Human Stem Human CD45⫺, CD31⫺ Not stated CD90⫹, CD105⫹, Dell’Agnola et
Cells CD106⫹, CD133⫺, al., 2002
CD135⫺, HER2/neu⫹,
P1H12⫹, c-kit⫹,
CXCR4⫹,
Multipotent Mouse CD44⫺, CD45⫺, MHC-I⫺, MHC- 75 CD31⫹, Tau⫹, HNF-1⫺, Jiang et al.,
Adult II⫺, c-kit⫺, Ter1119⫺, CD13⫹, population GFAP⫹, CK18⫹, 2002
Progenitor Flk1dim, Oct4⫹, Rex1⫹, CD31⫺, doublings Flk1⫹, NF-200⫹,
Cells CD62E⫺, Tek⫺, vWF⫺ NSE⫹, vWF⫹,
Albumin⫹:
Endothelial,
astrocytes, neurons,
epithelioid
Cementum- Mouse Low levels or no alkaline Not stated Bone-like tissue with Krebsbach
derived cells phosphatase activity osteocytes/cemento- and Robey,
cyte-like cells 2002
Cementum- Human Low levels or no alkaline Not stated Bone-like tissue with Krebsbach
derived cells phosphatase activity osteocytes/cemento- and Robey,
cyte-like cells 2002
Dental pulp stem Human Collagen type XVIII ␣1, IGF-2, Not stated Dentin lined with Krebsbach
cells discordin domain tyrosine odontoblasts-like cells, and Robey,
kinease-2, cyclin-dependent dental pulp-like tissue 2002
kinase6
Neural Crest Rat p75 ⫹ Self- neurons, glia, Kruger et al.,
Stem Cells Renewal myofibroblasts 2002
declined
with
increasing
age
Bone marrow Mouse Unfractionated bone marrow Not stated Diploid, desmin, Myf-5, LaBarge and
derived cell isolate cMet-R, ␣7-integren, Blau, 2002
Satellite cell to muscle
repair
Progenitor Cells Rat Cells from low-buoyancy fraction Not stated NeuN⫹, Rip ⫹/⫺, Lie et al.,
of Percoll, BrdU⫹, GFP⫹, NG2⫹, APC⫹, 2002
NG2⫹ S100b⫹, GFAP⫹,
A2B5⫹, BrdU⫹,
Ox42⫹/⫺, Nestin⫹
TH⫹, b-Tubulin III⫹/

Muscle-derived Mouse Sca-1⫹, CD45⫹, CD34⫺, c-kit⫺ Not stated Hematopoietic cells: McKinney-
hematopoietic Gr.1⫹, Mac-1⫹, Freeman et
stem cells Thy1⫹, B220⫹ al., 2002
Muscle-derived Mouse CD45⫺, Sca-1⫹ & CD45⫺, Sca- Not stated Fibroblastic-like cells McKinney-
myogenic stem 1⫺ and twitching skeletal Freeman et
cells muscle myofibers al., 2002
15524892, 2004, 1, Downloaded from https://anatomypubs.onlinelibrary.wiley.com/doi/10.1002/ar.a.10134 by Cochrane Romania, Wiley Online Library on [31/10/2022]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
82 YOUNG AND BLACK

TABLE 1. Postnatal precursor cells (continued)


Proliferation
Name Species Characteristics Potential Differentiation Potential References
Neural Stem Mouse Nestin⫹, EGFP⫹, b-Tubulin Self- Neurospheres Murayama et
Cells III⫹, GFAP⫺, O4⫹ Renewal al., 2002
of sorted
neurospher
e-
initiating
cells
Neural Stem Cell Mouse c-Kit⫹, Sca1⫺, CD45⫺, Notch Not stated Unknown Murayama et
Side 1⫹, Nestin⫺, EGFP⫺ al., 2002
Population
Bone Marrow- Mouse CD3⫺, CD11⫺, CD45⫺, Ter- Not stated CD31⫹, c-kit⫹, Sca1⫹, Otani et al.,
derived stem 199⫺, Ly-6G⫺ Flk1⫹ 2002
cells
Bone Marrow Human ␣-LNGFR ⫹, CD45⫺, a- High CD45⫺, TE7⫹, CD14⫹, Quirici et al.,
Mesenchymal glycophorin-A⫺, SH2⫹, proliferative CD34⫺, CD133⫺, 2002
Stem Cells ␣- CD34⫹, CD133⫹ capacity ORO⫹, Alizarin Red S
LNGFR⫹ ⫹
Bone Marrow Human ␣-LNGFR⫺, SH2⫺, CD45⫺, a- High CD45⫺, TE7⫹, CD14⫹, Quirici et al.,
Mesenchymal glycophorin-A⫺ proliferative Alizarin Red S ⫹ 2002
Stem Cells ␣- capacity
LNGFR⫺
Pluripotent Stem Rat Vimentin⫹, Pax6⫹, CD34 varied, Not stated Nestin⫹, Pax6⫹, Oct4⫹, Romero-
Cells CD45⫺, CD90⫹, Oct4 low, MBP⫹, NG2⫹, Ramos et
Myo-D⫺, myogenin⫺ NF145⫹, CNPase⫹, al., 2002
Tuj1⫹, NF68⫹,
GFAP⫹, MOSP⫹,
Tau⫹, NeuN⫺
Smooth Muscle Human CD34⫹, CD31⫺, Tie2⫺, Flk1⫹, Not stated Smooth muscle cells Simper et al.,
Outgrowth Flt1⫹, VE-Cadherin⫺, vWF⫺, 2002
Cells VEGF1⫹,2⫹, Integrin␣5␤1⫹,
␣SMA⫹, MHC⫹, Calponin⫹
Endothelial Human CD31⫹, vWF⫹, VE-Cadherin⫹, Not stated Endothelial cells Simper et al.,
Outgrowth Flt1⫹, Flk1⫹, ␣SMA⫺, Tie-2⫹ 2002
Cells
Bone marrow Rat Ring cloning; express germline, 15 to ⬎20 Induced neurons: Tau⫹, Woodbury et
stromal cells ectodermal, mesodermal, & passages TOAD-64⫹, b-Tubulin al., 2002
endodermal genes prior to III⫹, synaptophysis⫹,
differentiation ChAT⫹, TH⫹,
GFAP⫹, NF-M⫹,
vK⫹, NeuroD⫺
Adult hepatic Rat Two-step collagenase-perfusion, Long term Induced pancreatic Yang et al.,
oval stem cells cell sorting with Thy-1.1: AFP, culture for endocrine hormone 2002
albumin, ␥-glutamyl- ⬎ 1 year producing cells:
transpeptidase CK19, OV6 by serial insulin-1, insulin-2,
1:2 splits glucagon, somatostatin
Neural stem cells Human Not stated 60 cell Not stated Johe, 2003
doublings
Muscle SP cells Mouse SP⫹ (side population⫹), CD34⫺, Not stated Endothelium Majka et al.,
c-met⫹, CD45⫹, Sca-1⫹, PE- 2003
CAM⫹, Tie-2⫹
Muscle non-SP Mouse SP⫺, c-met⫹, CD45⫹, Sca-1⫹, PE- Not Stated Smooth muscle Majka et al.,
cells CAMlow, Tie-2low, CD34low, SM- 2003
␣-actin⫹, PDGFR␤⫹, Calponin-
mRNA
Hepatic stellate Mouse Anti-␣-smooth muscle actin⫹, ? 7 days When sitmulated with Oben et al.,
cells GFAP⫹ serum and PDGF the 2003
cells increased
proliferation and
increased expression of
collagen 1␣2
Adipose tissue- Human From low density region of Ficoll- 2 weeks Adipogenic, osteogenic, Winter et al.,
derived Paque Plus density gradient chondrogenic 2003
Stromal cells
Pluripotent Rat Cloned, can be subcloned at 1 cell ⬎ 400 Ectoderm: 7⫹ cell types; Young, 2004;
Epiblastic-Like per well, Oct-4⫹, Telomerase⫹ population Mesoderm: 18⫹ cell Young et
Stem Cells doublings types; Endoderm: 11⫹ al., 2004b
cell types
15524892, 2004, 1, Downloaded from https://anatomypubs.onlinelibrary.wiley.com/doi/10.1002/ar.a.10134 by Cochrane Romania, Wiley Online Library on [31/10/2022]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
ADULT STEM CELLS 83
TABLE 1. Postnatal precursor cells (continued)
Proliferation
Name Species Characteristics Potential Differentiation Potential References
Pluripotent Human Sorted, CD10⫹, CD66e⫹ SSEA- ⬎ 400 Ectoderm: 7⫹ cell types; Young, 2004;
Epiblastic-Like 1⫹, SSEA-3⫹, SSEA-4⫹, population Mesoderm: 18⫹ cell Young et
Stem Cells CEA⫹, HCEA⫹, CEA-CAM1⫹ doublings types; Endoderm: 11⫹ al., 2004a
cell types
Adult pluripotent Human CD14, CD34, CD45 Not stated Macrophages, T- Zhao et al.,
stem cells lymphocytes, epithelial 2003
cells, endothelial cells,
neuronal cells, liver
cells

TABLE 2. Precursor cell characteristics


Progenitor Cells PPGLSCs1 PPELSCs2
Serum-Free Defined Medium3 Quiescence Quiescence Quiescence
Commitment Lineage-Specific Germ Layer-Specific Uncommitted
Life Span4 Hayflick’s Limit5 Extended6 Extended
Telomerase Absent Present Present
Cell Growth at Confluence Contact Inhibited Contact Inhibited Non-Contact Inhibited
Proliferation7 Responsive Responsive Responsive
Induction8 Unresponsive Responsive, Germ Layer-specific Responsive, unlimited
Progression9 Responsive Unresponsive Unresponsive
Inhibitory10 Responsive Responsive Responsive
Antibodies11 Cell-Specific Germ Layer-Specific Embryonic
CD Markers12 Cell-Specific CD10, CD13, CD34, CD56, CD90, CD10, CD66e
MHC1 (PPMSCs only)
Cells Formed Lineage-Specific Germ Layer-Specific Ectoderm, Mesoderm,
Endoderm
1
PPGLSCs, pluripotent germ layer stem cells, i.e., pluripotent ectodermal stem cells (PPEctoSCs), pluripotent mesodermal
stem cells (PPMSCs), pluripotent endodermal stem cells (PPEndoSCs).
2
PPELSCs, pluripotent epiblastic-like stem cells.
3
Serum-Free Defined Medium, medium in the absence of proliferation factors, induction factors, progression factors, and
inhibitory factors (i.e., leukemia inhibitory factor, fibroblast feeder layer, anti-differentiation factor, scar inhibitory factor, etc.)
or their activities.
4
Life Span, population-doubling number.
5
Hayflick’s Limit, up to 50-70 population doublings before cell senescence and cellular death (Hayflick, 1965).
6
Extended, population doublings greater than Hayflick’s limit of 70. In this instance both PPMSC clone and PPELSC clone
were tested to 400 population doublings. Both clones retained their respective pluripotent potentials throughout maximum
population doublings examined (Young, 2004).
7
Proliferation, initiation of cell proliferation in response to proliferation agents such as platelet-derived growth factors
(PDGF)-AA, PDGF-BB, PDGF-AB, etc.
8
Induction, commitment to a specific germ layer or tissue/cell lineage in response to a specific (i.e., bone morphogenetic
protein-2, skeletal muscle morphogenetic protein, etc.) or a general (dexamethasone, etc.) lineage-tissue-cell inductive agent.
9
Progression, acceleration of expression of specific phenotypic marker(s) in response to progression agents such as insulin,
insulin-like growth factor (IGF)-I, IGF-II, etc.
10
Inhibitory, inhibition of commitment to and/or expression of specific phenotypic marker(s) in response to a general (i.e.,
leukemia inhibitory factor, LIF; ESGRO, murine LIF; fibroblast feeder layer; anti-differentiation factor, ADF; etc.) or a specific
(scar inhibitory factor, SIF; etc.) inhibitory agent.
11
Antibodies, antibodies to phenotypic expression markers, can be cell specific (i.e., OP137, F5D, MF-20, MY-32, ALD-58, and
A4.74 for skeletal muscle), germ layer specific (i.e., FORSE-1, Rat-401, HNES, and MAB353 for neural ectoderm), or embryonic
(i.e., SSEA-1, SSEA-3, SSEAS-4, CEA, HCEA, CD66e, and CEA-CAM-1 for embryonic stem cells) (see Table 3 for further
examples).
12
CD Markers, cluster of differentiation makers.

cells exhibit contact inhibition at confluence. Progenitor grown in the absence of IGF-I. Progenitor cells remain
cells are responsive to progression agents (i.e., insulin, quiescent in a serum-free environment lacking progres-
insulin-like growth factor-I (IGF-I), insulin-like growth sion agents, proliferation agents, and inhibitory factors
factor-II (IGF-II)) that accelerate the time frame of expres- (i.e., leukemia inhibitory factor, murine leukemia inhibi-
sion for tissue-specific markers of phenotypic differentia- tory factor (ESGRO), a fibroblast feeder layer, antidiffer-
tion. For example, skeletal muscle myoblasts grown in the entiation factor, etc.).
presence of IGF-I will demonstrate myogenic phenotypic- Examples of lineage-committed progenitor cells include
specific expression markers characteristic of skeletal mus- the unipotent myosatellite myoblasts of muscle (Mauro,
cle (i.e., sarcomeric myosin, myosin heavy chain, myosin 1961; Campion, 1984; Grounds et al., 1992; Young et al.,
fast chain, etc.) in about half the time taken by myoblasts 1993, 1995, 1998b); the unipotent adipoblast cells of adi-
15524892, 2004, 1, Downloaded from https://anatomypubs.onlinelibrary.wiley.com/doi/10.1002/ar.a.10134 by Cochrane Romania, Wiley Online Library on [31/10/2022]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
84 YOUNG AND BLACK

pose tissue (Ailhaud et al., 1992; Young et al., 1993, 1995); bryonic antigen, or carcinoembryonic antigen-cell adhe-
the unipotent chondrogenic cells and osteogenic cells of sion molecule-1 (Tables 1–3). They exist in a quiescent
the perichondrium and periosteum, respectively (Cruess, state of stasis in serum-free defined medium lacking pro-
1982; Young et al., 1993, 1995); the unipotent basal epi- liferation factors, inductive factors, progression factors, or
thelial cells of gastrointestinal mucosa (Bjerknes and inhibitory factors. In this state they display no cell prolif-
Cheng, 2002); and neuronal unipotent progenitor cells eration, no spontaneous differentiation, and no cell death
(Gage et al., 1995). Bipotent progenitor cells include the (Fig. 3A). These cells display contact inhibition at conflu-
adipofibroblasts of adipose tissue (Vierck et al., 1996) and ence, ceasing cell proliferation once a single layer of cells
the bipotent chondrogenic-osteogenic stem cells of marrow covers the surface of a flask (Fig. 3B), even in the contin-
(Owen, 1988; Owen and Friedenstein, 1988; Beresford, ued presence of proliferation agents such as PDGFs.
1989; Caplan et al., 1997; Prockop, 1997; Young, 2000). These precursor cells are responsive to both general and
Tripotent progenitor cells include the chondrogenic-osteo- specific inductive factors, but only with respect to cells of
genic-adipogenic stem cells of marrow (Pittenger et al., the mesodermal lineage. They can be induced to form 18 or
1999; Young, 2000). Multipotent hematopoietic cells of more phenotypes limited to the mesodermal lineage (Ta-
marrow (McGuire, 1998; Palis and Segel, 1998; Ratajczak ble 3, Fig. 3C–T) from a single cell, derived from either
et al., 1998) are an example of multipotent progenitor repetitive limiting serial dilution clonogenic analysis or
cells. See Table 1 for additional examples of lineage-com- cell sorting. These cells will only form progenitor cells
mitted progenitor cells. committed to the mesodermal lineage, being unresponsive
During the studies that led to the characterization of to general or specific inductive factors for cells of the
lineage-committed progenitor cells (Young et al., 1991, ectodermal or endodermal lineages. Once committed to a
1992a, 1992b, 1993, 1995, 1998a, 1998b, 1999, 2001a, particular mesodermal tissue lineage (i.e., myogenic,
2001b, 2004a; Rogers et al., 1995; Young and Lucas, 1998; chondrogenic, etc.), the newly induced lineage-specific pro-
Young, 2000, 2004), we noted the presence of another genitor cells are unresponsive to inductive factors outside
general category of precursor cells within the tissues, the that respective lineage. For example, newly induced pro-
lineage-uncommitted pluripotent stem cells. Further ex- genitor cells committed to the myogenic lineage are unre-
periments revealed the presence of unique subcategories sponsive to BMP-2, a factor inducing commitment to the
of these pluripotent stem cells. To date we have been bipotent chondrogenic-osteogenic lineage. Similarly,
concerned with the definitive characterization of two of newly induced progenitor cells committed to the chondro-
these subcategories of pluripotent stem cells. Both subcat- genic lineage are unresponsive to skeletal muscle morpho-
egories of pluripotent stem cells share a few similarities genetic protein (Sk-MMP), a factor inducing commitment
with progenitor cells, including quiescence in an environ- to the skeletal muscle myogenic lineage. However, these
ment that lacks serum, proliferation agents, inductive newly induced progenitor cells respond to progression fac-
agents, progression agents, and inhibitory agents or their tors by accelerating the expression of their differentiated
influences. They are also similar in their response to pro- phenotype. For example, myogenic progenitor cells ex-
liferation agents. However, these two pluripotent stem press differentiated muscle markers in about two weeks in
cell subcategories differ in many respects from lineage- the presence of IGF-I rather than the usual four weeks in
committed progenitor cells (Tables 1 and 2). They also the absence of a progression agent. Similarly, in the pres-
demonstrate similarities and differences with respect to ence of IGF-I, osteogenic progenitor cells express markers
each other (Tables 1–3). for differentiated bone in about four weeks rather than six
Both pluripotent stem cell subcategories are telomerase weeks in the absence of a progression agent. These newly
positive (Fig. 2A). Both exhibit extended capabilities for induced progenitor cells assume Hayflick’s limit of 50 –70
self-renewal without loss of differentiative capabilities, far population doublings before programmed senescence and
surpassing Hayflick’s limit of 50 –70 population doublings. cellular death occur. We designated these lineage-uncom-
Both pluripotent stem cell subcategories are unresponsive mitted precursor cells as pluripotent mesodermal stem
to progression factors such as insulin, IGF-I, or IGF-II cells (PPMSCs) due to their extended capabilities for self-
that normally accelerate phenotypic expression in lineage- renewal that far surpass Hayflick’s limit and their ability
committed progenitor cells. to form any cell type belonging to the mesodermal lineage.
The first subcategory of lineage-uncommitted pluripo- The second subcategory of lineage-uncommitted precur-
tent stem cells characterized by Young and colleagues sor cells undergoing characterization (Young et al., 2004a;
(Young and Lucas, 1998; Young et al., 1998a, 1998b, 1999, Young, 2004) expresses the following cell surface profile:
2001a, 2001b, 2004a; Young, 2000, 2004) express the fol- CD10⫹, CD66e⫹, CD1a–, CD2–, CD3–, CD4–, CD5–, CD7–,
lowing cell surface profile: CD10⫹, CD13⫹, CD34⫹, CD8–, CD9–, CD11b–, CD11c–, CD13–, CD14–, CD15–,
CD56⫹, CD90⫹, MHC-I⫹, CD1a–, CD2–, CD3–, CD4–, CD16–, CD18–, CD19–, CD20–, CD22–, CD23–, CD24–,
CD5–, CD7–, CD8–, CD9–, CD11b–, CD11c–, CD14–, CD25–, CD31–, CD33–, CD34–, CD36–, CD38–, CD41–,
CD15–, CD16–, CD18–, CD19–, CD20–, CD22–, CD23–, CD42b–, CD45–, CD49d–, CD55–, CD56–, CD57–, CD59–,
CD24–, CD25–, CD31–, CD33–, CD36–, CD38–, CD41–, CD61–, CD62E–, CD65–, CD68–, CD69–, CD71–, CD79–,
CD42b–, CD45–, CD49d–, CD55–, CD57–, CD59–, CD61–, CD83–, CD90–, CD95–, CD105–, CD117–, CD123–,
CD62E–, CD65–, CD66e–, CD68–, CD69–, CD71–, CD79–, CD135–, CD166–, Glycophorin-A–, MHC-I–, HLA-DRII–,
CD83–, CD95–, CD105–, CD117–, CD123–, CD135–, CD166–, FMC-7–, Annexin-V–, and LIN–. A clone with similar at-
Glycophorin-A–, HLA-DRII–, FMC-7–, Annexin-V–, and tributes to the CD10⫹, CD66e⫹ human precursor cell sub-
LIN–. These precursor cells are 10 –20 ␮m in size as de- category was derived from postnatal rats by limiting serial
termined by flow cytometric analysis of unfixed cells. dilution clonogenic analysis. Both the sorted adult human
These precursor cells do not express embryonic stem cell cells and the cloned postnatal rat cells show similarities
markers such as alkaline phosphatase, stage-specific em- and differences with respect to the PPMSCs characterized
bryonic antigen-1 (SSEA-1), SSEA-3, SSEA-4, carcinoem- previously. The similarities include the presence of telom-
15524892, 2004, 1, Downloaded from https://anatomypubs.onlinelibrary.wiley.com/doi/10.1002/ar.a.10134 by Cochrane Romania, Wiley Online Library on [31/10/2022]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
ADULT STEM CELLS 85
TABLE 3. Identification of phenotypic expression markers in postnatal pluripotent stem cells
Phenotypic Markers PPELSCs1 PPEctoSCs2 PPMSCs3 PPEndoSCs4
Embryonic
Alkaline Phosphatase ⫹ ⫺ ⫺ ⫺
SSEA-15 ⫹ ⫺ ⫺ ⫺
SSEA-36 ⫹ ⫺ ⫺ ⫺
SSEA-47 ⫹ ⫺ ⫺ ⫺
CEA8 ⫹ ⫺ ⫺ ⫺
HCEA9 ⫹ ⫺ ⫺ ⫺
CD66e10 ⫹ ⫺ ⫺ ⫺
CEA-CAM111 ⫹ ⫺ ⫺ ⫺
Oct-412 ⫹ nd13 nd nd
Ectoderm
Neurogenic Progenitor Cells14 ⫹ ⫹ ⫺ ⫺
Neurons15 ⫹ ⫹ ⫺ ⫺
Ganglia16 ⫹ ⫹ ⫺ ⫺
Oligodendrocytes17 ⫹ ⫹ ⫺ ⫺
Astrocytes18 ⫹ ⫹ ⫺ ⫺
Radial Glial Cells19 ⫹ ⫹ ⫺ ⫺
Keratinocytes20 ⫹ ⫹ ⫺ ⫺
Mesoderm
Skeletal Muscle21 ⫹ ⫺ ⫹ ⫺
Smooth Muscle22 ⫹ ⫺ ⫹ ⫺
Cardiac Muscle23 ⫹ ⫺ ⫹ ⫺
White Fat24 ⫹ ⫺ ⫹ ⫺
Brown Fat25 ⫹ ⫺ ⫹ ⫺
Hyaline Cartilage26 ⫹ ⫺ ⫹ ⫺
Articular Cartilage27 ⫹ ⫺ ⫹ ⫺
Elastic Cartilage28 ⫹ ⫺ ⫹ ⫺
Growth Plate Cartilage29 ⫹ ⫺ ⫹ ⫺
Fibrocartilage30 ⫹ ⫺ ⫹ ⫺
Endochondral Bone31 ⫹ ⫺ ⫹ ⫺
Intramembranous Bone32 ⫹ ⫺ ⫹ ⫺
Tendon/Ligament33 ⫹ ⫺ ⫹ ⫺
Dermis34 ⫹ ⫺ ⫹ ⫺
Scar Tissue35 ⫹ ⫺ ⫹ ⫺
Endothelial Cells36 ⫹ ⫺ ⫹ ⫺
Hematopoietic Cells37 ⫹ ⫺ ⫹ ⫺
Endoderm
Endodermal Progenitor Cells38 ⫹ ⫺ ⫺ ⫹
GI Epithelium39 ⫹ ⫺ ⫺ ⫹
Liver Oval Cells40 ⫹ ⫺ ⫺ ⫹
Liver Hepatocytes41 ⫹ ⫺ ⫺ ⫹
Liver Biliary Cells42 ⫹ ⫺ ⫺ ⫹
Liver Canalicular Cells43 ⫹ ⫺ ⫺ ⫹
Pancreatic Progenitor Cells44 ⫹ ⫺ ⫺ ⫹
Pancreatic Ductal Cells45 ⫹ ⫺ ⫺ ⫹
Pancreatic ␤-Cells46 ⫹ ⫺ ⫺ ⫹
Pancreatic ␣-Cells47 ⫹ ⫺ ⫺ ⫹
Pancreatic ␦-Cells48 ⫹ ⫺ ⫺ ⫹
1
PPELSCs, pluripotent epiblastic-like stem cells (isolated and cloned) (Young et al., 2004a, 2004b; Young, 2004).
2
PPEctoSCs, pluripotent ectodermal stem cells (induced) (Romero-Ramos et al., 2002; Young, 2004; Young et al., 2004a).
3
PPMSCs, pluripotent mesodermal stem cells (isolated and cloned) (Young et al., 1999, 2001a,2001b; Young, 2000, 2004).
4
PPEndoSCs, pluripotent endodermal stem cells (induced) (Young, 2004; Young et al., 2004a).
Embryonic cells were identified as follows.
5
SSEA-1, stage-specific embryonic antigen-1, MC480, Developmental Studies Hybridoma Bank (DSHB), Iowa City, IA (Solter
and Knowles, 1978).
6
SSEA-3, stage-specific embryonic antigen-3, antibody MC631 (DSHB) (Damjanov et al., 1982).
7
SSEA-4, stage-specific embryonic antigen-4, antibody MC-813-70 (DHSB) (Lannagi et al., 1983).
8
CEA, carcinoembryonic antigen, (Hixson) (Estrera et al., 1999).
9
HCEA, human carcinoembryonic antigen (Sigma) (Young et al., 2004a).
10
CD66e, carcinoembryonic antigen (Vector) (Kishimoto et al., 1997).
11
CEA-CAM1, carcinoembryonic antigen-cell adhesion molecule (Hixson) (Estrera et al., 1999).
12
Oct-4, a gene directly involved in the capacity for self-renewal and totipotency of mammalian embryonic stem cells (Pesce and
Scholer, 2001; Young et al., 2004a).
13
nd, not as yet determined.
Ectodermal lineage cells were identified as follows.
14
Neurogenic Progenitor Cells, were identified using FORSE-1 (DSHB) for neural precursor cells (Tole et al., 1995; Tole and
Patterson, 1995), RAT-401 (DSHB) for nestin (Hockfield and McKay, 1985), HNES (Chemicon, Temecula, CA) for nestin
(Young et al., 2004a,b), and MAB353 (Chemicon) for nestin (Gritti et al., 1996).
15524892, 2004, 1, Downloaded from https://anatomypubs.onlinelibrary.wiley.com/doi/10.1002/ar.a.10134 by Cochrane Romania, Wiley Online Library on [31/10/2022]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
86 YOUNG AND BLACK

erase activity (Fig. 2A); extended capabilities for self-re- limit of 50 –70 population doublings before programmed
newal far surpassing Hayflick’s limit; quiescence and sta- senescence and cellular death occur.
sis in serum-free defined medium lacking proliferation, However, distinct differences also exist between the plu-
induction, and inhibitory factors; unresponsiveness to pro- ripotent mesenchymal stem cells and the cells exhibiting
gression factors; responsiveness to inductive factors and CD10⫹, CD66e⫹ cell surface markers or their equivalent
inhibitory factors; and assumption of progenitor cell sta- clone. The adult-derived CD10⫹, CD66e⫹ precursor cells
tus once committed to a particular tissue lineage. Once are highly mobile and small (Fig. 4A and B), averaging
committed to a particular tissue lineage, the progenitor 6 – 8 mm in size by flow cytometric analysis of unfixed
cells derived from the CD10⫹, CD66e⫹ lineage exhibit cells. They do not exhibit contact inhibition at confluence,
contact inhibition at confluence, lack of responsiveness to but rather form multiple confluent layers of cells in the
induction factors outside their respective tissue lineage, presence of proliferation agents (Fig. 4C and D). Although
responsiveness to progression factors accelerating their derived from adults, these precursor cells express embry-
phenotypic expression, and conformance to Hayflick’s onic stem cell markers, i.e., alkaline phosphatase,

15
Neurons, were identified using 8A2 (DSHB) for neurons (Drazba et al., 1991), S-100 (Sigma) for neurons (Baudier et al.,
1986), T8660 (Sigma) for beta-tubulin III (Banerjee et al., 1988, 1990; Joshi and Cleveland, 1990), RT-97 (DSHB) for
neurofilaments (Wood and Anderton, 1981), N-200 (Sigma) for neurofilament-200 (Debus et al., 1983a; Franke, et al., 1991),
and SV2 (DSHB) for synaptic vesicles (Feany et al., 1992).
16
Ganglia, were identified using TuAg1 (Hixson) for ganglion cells (Faris et al., 1990; Hixson et al., 1990).
17
Oligodendrocytes, were identified using Rip (DSHB) for oligodendrocytes (Friedman et al., 1989) and CNPase (Sigma) for
oligodendrocytes and astroglia (Sprinkle et al., 1987; Sprinkle, 1989; Reynolds et al., 1989).
18
Astrocytes, were identified using CNPase (Sigma) for astroglia and oligodendrocytes (Sprinkle et al., 1987; Sprinkle, 1989;
Reynolds et al., 1989).
19
Radial Glial Cells, were identified using 40E-C (DSHB) for radial glial cells (Alvarez-Buylla et al., 1987).
20
Keratinocytes, were identified using VM-1 (DSHB) to keratinocyte cell surface protein (Morhenn, 2002).
Mesodermal lineage cells were identified as follows.
21
Skeletal Muscle, was identified as mononucleated myoblasts staining with OP137 (Calbiochem, San Diego, CA) for MyoD
(Thulasi et al., 1996), F5D (DSHB) for myogenin (Wright et al., 1991), and DEU-10 (Sigma) for desmin (Debus et al., 1983b),
and as multinucleated spontaneously contracting structures staining with MF-20 (DSHB) for sarcomeric myosin (Bader et al.,
1982), MY-32 (Sigma) for skeletal muscle fast myosin (Naumann and Pette, 1994), ALD-58 (DSHB) for myosin heavy chain
(Shafiq et al., 1984), and A4.74 (DSHB) for myosin fast chain (Webster et al., 1988).
22
Smooth Muscle, was identified as mononucleated cells staining with antibodies IA4 (Sigma) for smooth muscle alpha-actin
(Skalli et al., 1986) and Calp (Sigma) for calponin (Frid et al., 1992; Lazard et al., 1993).
23
Cardiac Muscle, was identified as binucleated cells co-staining with MF-20 (DSHB) ⫹ IA4 (Sigma) for sarcomeric myosin and
smooth muscle alpha actin (Eisenberg and Markwald, 1997; Eisenberg et al., 1997), MAB3252 (Chemicon) for cardiotin
(Schaart et al., 1997) and MAB1548 for cardiac muscle (Chemicon).
24
White Fat, also denoted as unilocular adipose tissue, was identified as a mononucleated cell with a peripherally-located
nucleus and containing a large central intracellular vacuole filled with refractile lipid and stained histochemically for
saturated neutral lipid using Oil Red-O (Sigma) and Sudan Black-B (Chroma-Gesellschaft, Roboz Surgical Co, Washington,
DC) (Young et al., 2001a).
25
Brown Fat, also denoted as multi-locular adipose tissue was identified as a mononucleated cell with a centrally-located
nucleus containing multiple small intracellular vacuoles filled with refractile lipid and stained histochemically for saturated
neutral lipid using Oil Red-O (Sigma) and Sudan Black-B (Chroma-Gesellschaft) (Young, 2000; Young et al., 2001b).
26 –30
Cartilage: structures thought to be cartilage nodules were tentatively identified as aggregates of rounded cells containing
pericellular matrix halos. Cartilage nodules were confirmed both by both histochemical and immunochemical staining.
Histochemically, cartilage nodules were visualized by staining the pericellular matrix halos for proteoglycans containing
glycosaminoglycan side chains with chondroitin sulfate and keratan sulfate moieties. This was accomplished using Alcian Blue
(Alcian Blue 8GS, Chroma-Gesellschaft), Safranin-O (Chroma-Gesellschaft) at pH 1.0, and Perfix/Alcec Blue. Verification of
glycosaminoglycans specific for cartilage was confirmed by loss of extracellular matrix staining following digestion of the
material with chondroitinase-AC (ICN Biomedicals, Cleveland, OH) and keratinase (ICN Biomedicals) (Young et al., 1989a,b,
2001a,b) prior to staining (negative staining control). Immunochemically, the chondrogenic phenotype was confirmed by initial
intracellular staining followed by subsequent staining of the pericellular and extracellular matrices with CIIC1 (DSHB) for
type-II collagen (Holmdahl et al., 1986), HC-II (ICN Biomedicals, Aurora, OH) for type-II collagen (Burgeson and Hollister,
1979; Kumagai et al., 1994), D1-9 (DSHB) for type-IX collagen (Ye et al., 1991), 9/30/8A4 (DSHB) for link protein (Caterson
et al., 1985), 12/21/1C6 (DSHB) for proteoglycan-hyaluronate binding region (Caterson, 2001), and 12C5 (DSHB) for versican
(Asher et al., 1995). Types of cartilage were segregated based on additional attributes.
26
Hyaline Cartilage, was identified by a perichondrial-like connective tissue surrounding the above stained cartilage nodule
and histochemical co-staining for type-I collagen (Young et al., 1989c).
27
Articular Cartilage, was identified as the above stained cartilage nodule without a perichondrial-like connective tissue
covering (Young et al., 1993).
28
Elastic Cartilage, was identified by nodular staining for elastin fibers and a perichondrial-like connective tissue surrounding
the above stained cartilage nodule and histochemical co-staining for type-I collagen (Young et al., 1989c).
29
Growth Plate Cartilage, was identified by nodular staining for cartilage phenotypic markers (see above) and co-staining for
calcium phosphate using the von Kossa procedure (Young et al., 1999, 2001a,b).
30
Fibrocartilage, was identified as three-dimensional nodules demonstrating extracellular histochemical staining for type-I
collagen (Young et al., 1989c) and co-staining for pericellular matrices rich in chondroitin sulfates A and C. The latter were
assessed by Alcian Blue pH 1.0 staining. Negative staining controls were digested prior to staining with chondroitinase-ABC
or chondroitinase-AC (Young et al., 1989a,b, 2001a,b).
15524892, 2004, 1, Downloaded from https://anatomypubs.onlinelibrary.wiley.com/doi/10.1002/ar.a.10134 by Cochrane Romania, Wiley Online Library on [31/10/2022]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
ADULT STEM CELLS 87
SSEA-1, SSEA-3, SSEA-4, carcinoembryonic antigen-cell tive tissue, endothelial cells, hematopoietic cells) (Table 3,
adhesion molecule-1, carcinoembryonic antigens (Table 1, Fig. 6A–P), and 11 or more endodermal cell types (i.e.,
Fig. 4E–L), and Oct-4 gene expression (Fig. 2B and C), in endodermal progenitor cells, gastrointestinal epithelial
a quiescent undifferentiated state. To date, these precur- cells, pancreatic progenitor cells, insulin-secreting ␤-cells,
sor cells have formed over 36 distinct cell types derived glucagon-secreting ␣-cells, somatostatin-secreting ␦-cells,
from all three primary germ layer lineages: 7 or more pancreatic ductal cells, liver oval cells, liver hepatocytes,
ectodermal cell types (i.e., neuronal progenitor cells, neu- liver biliary cells, and liver canalicular cells) (Table 3, Fig.
rons, ganglia, astrocytes, oligodendrocytes, radial glial 7A–P). We propose that the second subcategory of lineage-
cells, keratinocytes) (Table 3, Fig. 5A–S), 18 or more me- uncommitted precursor cells is a sequestered embryonic-
sodermal cell types (i.e., skeletal muscle, cardiac muscle, like stem cell existing within an adult. We have desig-
smooth muscle, white fat, brown fat, hyaline cartilage, nated this population as a pluripotent epiblastic-like stem
elastic cartilage, growth plate cartilage, articular carti- cell, due to its capacity for extended self-renewal well past
lage, fibrocartilage, cortical bone, trabecular bone, loose Hayflick’s limit and its ability to form cell types from all
fibrous connective tissues, tendon, ligament, scar-connec- three primary germ layer lineages.

31
Endochondral Bone, was identified as the formation of a three-dimensional structure with progressional staining from one
displaying chondrogenic phenotypic markers, i.e., pericellular type-II collagen, type-IX collagen, chondroitin sulfate/keratan
sulfate glycosaminoglycans (see above) to three-dimensional nodules displaying osteogenic phenotypic markers, i.e., WV1D1
(9C5) (DSHB) for bone sialoprotein II (Kasugai et al., 1992), MPIII (DSHB) for osteopontine (Gorski et al., 1990), and the von
Kossa procedure (Silber Protein, Chroma-Gesellschaft) for calcium phosphate. In the von Kossa procedure, negative staining
controls were pre-incubated in EGTA, a specific chelator for calcium (Sigma) (Young et al., 1993, 1999, 2001a,b).
32
Intramembranous Bone, was identified as a direct transition from stellate-shaped stem cells to three-dimensional nodules
displaying only osteogenic phenotypic markers, i.e., WV1D1 (9C5) (DSHB) for bone sialoprotein II (Kasugai et al., 1992), MPIII
(DSHB) for osteopontine (Gorski et al., 1990), and the von Kossa procedure (Silber Protein, Chroma-Gesellschaft) for calcium
phosphate. In the von Kossa procedure, negative staining controls were pre-incubated in EGTA, a specific chelator for calcium
(Sigma) (Young et al., 1993, 1999, 2001a,b).
33
Tendon/Ligament, was identified as linear structures with cellular staining for fibroblast specific protein IB10 (Sigma)
(Ronnov-Jessen et al., 1992) and displaying extracellular histochemical staining for type-I collagen (Young et al., 1989c).
34
Dermis, was identified by the presence of interwoven type-I collagen fibers (Young et al., 1989c) interspersed with
spindle-shaped cells staining for fibroblast specific protein IB10 (Sigma) (Ronnov-Jessen et al., 1992) with an extracellular
matrix rich in chondroitin sulfate and dermatan sulfate glycosaminoglycans as assessed by Alcian Blue pH 1.0 staining. In the
latter procedure negative staining controls were digested with chondroitinase-ABC or chondroitinase-AC prior to staining
(Young et al., 1989a,b, 2001a,b).
35
Scar Tissue, was identified as interwoven type-I collagen fibers (Young et al., 1989c) interspersed with spindle-shaped cells
staining for fibroblast specific protein IB10 (Sigma) (Ronnov-Jessen et al., 1992) with an extracellular matrix rich in
chondroitin sulfate glycosaminoglycans as assessed by Alcian Blue pH 1.0 staining. In the latter procedure negative staining
controls were digested with chondroitinase-ABC or chondroitinase-AC prior to staining (Young et al., 1989a,b, 2001a,b).
36
Endothelial Cells, were identified by staining with antibodies P2B1 (DSHB) for CD31-PECAM (Young et al., 2001b), H-Endo
(Chemicon) for CD146 (Gui et al., 1992; Solovey et al., 1997; St. Croix et al., 2000), P8B1 (DSHB) for VCAM (Dittel et al., 1993;
Young et al., 2001b), and P2H3 (DSHB) for CD62e selectin-E (Young et al., 2001b).
37
Hematopoietic Cells, were identified using H-CD34 (Vector) for sialomucin-containing hematopoietic cells (Kishimoto et al.,
1997; Young et al., 2001b), H5A4 (DSHB) for CD11b- granulocytes, monocytes, and Natural Killer cells (August and Hildreth,
2002), H5H5 (DSHB) for CD43 - leukocytes (August and Hildreth, 2002), H4C4 (DSHB) for CD44 - hyaluronate receptor
(August and Hildreth, 2002), Hermes-1 (DSHB) for CD44 - hyaluronate receptor (Butcher, 2002), H5A5 (DSHB) for CD45 - all
leukocytes (August and Hildreth, 2002), and H5C6 (DSHB) for CD63 - macrophages, monocytes, and platelets (August and
Hildreth, 2002).
Endodermal lineage cells were identified as follows.
38
Endodermal Progenitor Cells, were identified with H-AFP (Vector) and R-AFP (NORDIC) for alpha-fetoprotein (Mujoo et al.,
1983).
39
GI Epithelium, was identified with HESA (Sigma) for GI-epithelium (Young, 2004; Young et al., 2004a,b).
40
Liver Oval Cells, were identified with OC2 and OV6 (Hixson) for oval cells, liver progenitor cells, and biliary epithelial cells
(Faris et al., 1991; Gordon et al., 2000).
41
Liver Hepatocytes, were identified with H-1 and H-4 (Hixson) for hepatocyte cell surface marker and hepatocyte cytoplasm,
respectively (Walborg et al., 1985; Faris et al., 1991).
42
Liver Biliary Cells, were identified with OC2, OC3, OC4, OC5, OC10, DPP-IV, and OV6 (Hixson) for biliary epithelial cells,
liver progenitor cells, oval cells, and canalicular cells (Hixson et al., 1984, 1990, 2000; Walborg et al., 1985; Faris et al., 1991;
Gordon et al., 2000).
43
Liver Canalicular Cells, were identified with antibodies H4Ac19 (DSHB), DPP-IV, OV6, and LAP (Hixson) for bile canalic-
ular cells, liver progenitor cells, biliary epithelial cells, and canalicular cell surface protein (Hixson et al., 1984, 1990, 2000;
Hubbard et al., 1985; Walborg et al., 1985; Faris et al., 1991; Gordon et al., 2000).
44
Pancreatic Progenitor Cells, were tentatively identified as three-dimensional structures void of chondrogenic or osteogenic
phenotypic markers. This identity was confirmed by the presence phenotypic markers for pancreatic ductal cells, ␤-Cells,
␣-Cells, and ␦-Cells (Young, 2004; Young et al., 2004a).
45
Pancreatic Ductal Cells, were identified with cytokeratin-19 (Chemicon) to pancreatic ductal cells (Young, 2004; Young et al.,
2004a).
46
Pancreatic ␤-Cells, were identified with YM-PS5088 (Accurate) an antibody to insulin (Young, 2004; Young et al., 2004a).
47
Pancreatic ␣-Cells, were identified with YM-PS087 (Accurate) an antibody to glucagon (Young, 2004; Young et al., 2004a).
48
Pancreatic ␦-Cells, were identified with 11180 (ICN) an antibody to somatostatin (Young, 2004; Young et al., 2004a).
15524892, 2004, 1, Downloaded from https://anatomypubs.onlinelibrary.wiley.com/doi/10.1002/ar.a.10134 by Cochrane Romania, Wiley Online Library on [31/10/2022]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
88 YOUNG AND BLACK

We propose that while a majority of cells progress Cornelius et al., 1997; Eglitis and Mezey, 1997; Taylor et
through the normal embryogenetic developmental se- al., 1997; Ferrari et al., 1998; Young and Lucas, 1998;
quence to form an intact individual, a few cells leave this Gussoni et al., 1999; Petersen et al., 1999; Pittenger et al.,
genomically preprogrammed pathway to become seques- 1999; Alison et al., 2000; Bonner-Weir et al., 2000; Bosch
tered as reserve precursor cells. For example, a few cells et al., 2000; Brazelton et al., 2000; Clarke et al., 2000;
within the epiblast layer remain as epiblastic-like stem Galli et al., 2000; Lagasse et al., 2000; Lee et al., 2000;
cells and retain close contacts with the remaining cells as Mezey et al., 2000; Ramiya et al., 2000; Sanchez-Ramos et
they differentiate into the three primary germ layer lin- al., 2000; Woodbury et al., 2000; Deasy et al., 2001;
eages, ectoderm, mesoderm, and endoderm. Similarly, a Jankowski et al., 2001; Reyes and Verfaillie, 2001; Reyes
few cells from each germ layer remain, respectively, as et al., 2001; Toma et al., 2001; Jiang et al., 2002; McKin-
ectodermal stem cells, mesodermal stem cells, and ney-Freeman et al., 2002; Romero-Ramos et al., 2002) and
endodermal stem cells, and retain close contacts as the Table 1, we would thus propose the model and nomencla-
ectoderm differentiates into surface ectoderm and neural ture for adult tissue-resident precursor cells shown in
ectoderm, the mesoderm differentiates into lateral plate Figure 8. This model and nomenclature are based on the
mesoderm, intermediate mesoderm, and paraxial meso- normal developmental sequence shown in Figure 1.
derm, and the endoderm furthers its differentiation cas- Previous reports on the identification of adult precursor
cade. This process of precursor cell sequestration with cells based their nomenclature on the site of harvest for
subsequent further differentiation of the majority of the the tissue-resident precursor cells, i.e., bone marrow de-
cells continues to the unipotent progenitor cell and asso- rived, brain derived, skeletal muscle derived, etc. (Eglitis
ciated fully differentiated tissue state. Therefore, every and Mezey, 1997; Prockop, 1997; Bjornson et al., 1999;
tissue within the body retains a complement of precursor Jackson et al., 1999; Petersen et al., 1999; Brazelton et al.,
cells denoted by its embryological origin. Based on previ- 2000; Galli et al., 2000; Lagasse et al., 2000; Woodbury et
ous and current studies by ourselves and others (Young,
1977a, 1977b, 1977c, 1983, 2000, 2004; Young et al.,
1983a, 1983b, 1983c, 1983d, 1985, 1989a, 1989b, 1991,
1992a, 1992b, 1993, 1995, 1998a, 1998b, 1999, 2001a,
2001b, 2004a; Lucas et al., 1986, 1988, 1992, 1994a, Fig. 2. Molecular analysis of telomerase activity and Oct-4 expression
1994b, 1995; Lucas and Caplan, 1988; Lucas, 1989; Bow- in a ␤-Galactosidase-transfected postnatal rat pluripotent epiblastic-like
erman et al., 1991; Caplan, 1991; Shoptaw et al., 1991; stem cell clone, designated Rat-A2B2-scl-40 (␤-PPELSC) and a ␤-Galac-
Gage et al., 1995; Grande et al., 1995; Saito et al., 1995; tosidase-transfected postnatal rat pluripotent mesodermal stem cell clone,
designated Rat-A2A2-scl-2PG (␤-PPMSC) (Young et al., 2004a). A: Poly-
acrylamide gel electrophoresis of telomerase activity in pluripotent stem
cells. ␤-PPELSC clone, at 56 passages and 254 population doublings, and
␤-PPMSC clone, at 31 passages and 151 population doublings, were
utilized. Cells were propagated, harvested by trypsin release (Young et al.,
1999) and processed for telomerase activity as described by the manufac-
turer (TRAPeze Assay, Intergen). Lane 1 ⫹, extract of telomerase positive
cells (control), 1 ⫺, extraction buffer, Lane 2 ⫹, test extract of ␤-PPELSC
clone, 2 ⫺, heat inactivated extract of ␤-PPELSC clone; Lane 3 ⫹, test
extract of ␤-PPMSC clone, 3 ⫺, heat inactivated extract of ␤-PPMSC clone.
Note the presence of ladders of bands denoting the presence of telomerase
activity, compare ⫹ lanes 1–3. B: Presence of Oct-4. Oct-4 was detected
by the electrophoretic mobility shift assay using the oligonucleotide 5⬘-
TGTCGAATGCAAATCACTAGA-3⬘ containing the Oct-1 consensus binding
site. A ␤-PPELSC clone at 56 passages and 254 population doublings was
utilized. Cells were thawed, plated at 500 ⫻ 103 cells per 1% gelatinized
T-25 flask in stem cell propagation medium (SCPM), and grown past
confluence. SCPM consisted of 89% (v/v) Opti-MEM, 0.01 mM mercapto-
ethanol (␤ME), 1% antibiotics-antimycotic (ab-am), 10% SS3, at pH 7.4
(Young et al., 2004a). 10% SS3 contained platelet-derived growth factor
(PDGF)-like proliferative and anti-differentiation factor (ADF)-like inhibitory
activities (Young, 2000, 2004; Young et al., 2004a). Cells were harvested by
trypsin release (Young et al., 1999), processed for whole cell extracts as
described (Detn and Latchman, 1993), and aliquoted to 5,000 cell equiva-
lents. Cell aliquots were incubated for 30 min at room temperature with
Lane 1, no competitor; Lane 2, 100-fold excess of unlabelled Oct-1 oligo-
nucleotide; and Lane 3, Oct-4 specific antibody in 20mM Tris, pH 7.5, 4%
glycerol 0.5 mM dithiothreitol, 2␮g poly dIdC. 32P- labeled Oct-1 oligonu-
cleotide (1ng) was added and the mixture incubated for 30 min at room
temperature before electrophoresis through a 5% polyacrylamide gel. After
drying, bands were visualized with a phosphorimager and quantified using
the accompanying software. Two bands that represent binding by mem-
bers of the Oct family of transcription factors were obtained, as shown by
the competition for binding by unlabelled Oct oligonucleotide. C: Densito-
metric analysis of the area contained in the sidebar of the electrophoretic
mobility shift assay in Fig. 2B. Lane 1, solid line; Lane 2, long dashes; and
Lane 3, short dashes. Incubation with Oct-4 specific-antibody substantially
decreased the formation of the upper band, and slightly decreased the
formation of the lower band, indicating the presence of Oct-4.
15524892, 2004, 1, Downloaded from https://anatomypubs.onlinelibrary.wiley.com/doi/10.1002/ar.a.10134 by Cochrane Romania, Wiley Online Library on [31/10/2022]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
ADULT STEM CELLS 89
al., 2000; Deng et al., 2001; Vescovi et al., 2001, Yang et of the precursor cells were lineage-committed unipotent,
al., 2002). In contrast, the nomenclature for the precursor bipotent, tripotent, and multipotent progenitor cells for
cells shown in Figure 8 is based on the functionality of the other ectodermal, mesodermal, and endodermal tissue lin-
cells, i.e., their proliferation potential with respect to Hay- eages. Approximately 9% of the precursor cells were plu-
flick’s limit and their ability to commit to various tissue ripotent stem cells belonging to the ectodermal, mesoder-
lineages (Fig. 1), rather than their original site of harvest. mal, or endodermal cell lineages. And approximately 1% of
This model proposes that two general categories of pre- the precursor cells were pluripotent epiblastic-like stem
cursor cells exist within postnatal animals, including hu- cells, capable of forming any somatic cell of the body.
mans. The proposed general categories are lineage-un- Using the precursor cells derived from skeletal muscle as
committed pluripotent stem cells and lineage-committed an example, approximately 50% of the reserve precursor
progenitor cells. We would also propose that subcategories cells located in skeletal muscle consist of progenitor cells
exist within each general category of precursor cell and committed to the myogenic lineage. Approximately 40%
that each subcategory parallels the function of its respec- are progenitor cells committed to other lineages (i.e., adi-
tive counterpart in embryonic development (Fig. 1). The pogenic, fibrogenic, osteogenic, chondrogenic, endothelio-
proposed subcategories for pluripotent stem cells at the genic, hematopoietic, neurogenic, gliogenic, hepatogenic,
present time are epiblastic-like stem cells, ectodermal etc.). Approximately 9% are pluripotent germ layer stem
stem cells, surface ectodermal stem cells, neuroectodermal cells (i.e., pluripotent ectodermal stem cells, PPMSCs, and
stem cells, neural tube stem cells, neural crest stem cells, pluripotent endodermal stem cells). And approximately
mesodermal (mesenchymal) stem cells, paraxial mesoder- 1% are pluripotent epiblastic-like stem cells. Precursor
mal stem cells, intermediate mesodermal stem cells, lat- cells from the connective tissue compartments of such
eral plate mesodermal stem cells, and endodermal stem diverse tissues as dermis of the skin, bone marrow, brain,
cells. With time, additional subcategories of pluripotent adipose tissue (fat), perichondrium, periosteum, and vis-
stem cells may be identified. The proposed subcategories cera appear to follow the same pattern of progenitor cell
for progenitor cells are multipotent cells, tripotent cells, and pluripotent stem cell distribution as precursor cells
bipotent cells, and unipotent cells for each respective germ derived from the connective tissue compartments of skel-
layer lineage (Fig. 8). Thus, the presence within the tis- etal muscle (Young et al., 1993, 1995, 1998a, 2001a,
sues of both lineage-uncommitted pluripotent stem cells 2001b, 2004a; Young, 2000, 2004). Others have noted the
and lineage-committed progenitor cells provides for the presence of precursor cells within the tissues of animals,
continual maintenance and repair of the organism after including humans, with similar capabilities as those de-
birth. scribed above (Table 1).
To ascertain the validity or our proposed model and
nomenclature, we have been examining the precursor cell DIFFERENTATION OF PLURIPOTENT STEM
composition in the connective tissue compartments of CELLS VS. TRANSDIFFERENTIATION OF
many organs and tissues in nine different species, includ-
PROGENITOR CELLS
ing humans (review, Young, 2004; Young et al., 2004a).
We utilized our in vitro insulin-dexamethasone-ELICA Researchers have recently reported that precursor cells
phenotypic bioassay for the majority of these studies to derived from one organ are being reprogrammed to form
assess the identity of the isolated cells. Cellular identity tissues of another organ. For example, precursor cells
was based on their proliferation and differentiation poten- derived from skeletal muscle were initially reported to
tials. After an initial cell isolate, cells were assayed in form blood (Jackson et al., 1999). Precursor cells derived
96-well plates in defined testing medium (TM) only, TM from bone marrow have been reported to form neurons
plus 2–5 ␮g/ml insulin, and TM plus 10–10 to 10– 6 M and neural supportive tissues (Eglitis and Mezey, 1997;
dexamethasone. The cultures were grown for up to eight Brazelton et al., 2000; Woodbury et al., 2000), hepatic oval
weeks with medium changes three times per week. Cells cells (Petersen et al., 1999; Lagasse et al., 2000; Theise et
were assessed routinely for changes in morphology. After al., 2000), and muscle cells (Ferrari et al., 1998; Gussoni et
48 hr to eight weeks the cells were fixed and processed for al., 1999). Precursor cells derived from neuronal tissues
qualitative and quantitative ELICAs using our full library have been reported to form blood elements (Bjornson et
of probes for phenotypic expression markers of embryonic, al., 1999; Vescovi et al., 2001) and muscle cells (Clarke et
ectodermal, mesodermal, and endodermal lineage cells al., 2000; Galli et al., 2000; Tsai and McKay, 2000). And
(see legend to Table 3). Embryonic-like cells and differen- precursor cells derived from the liver have been reported
tiated cells expressed their respective phenotypic markers to form pancreatic islet cells (Yang et al., 2002). These
after incubation in TM only. Lineage-committed progeni- investigators proposed the theory of transdifferentiation,
tor cells expressed their respective phenotypic differenti- which involves the genetic reprogramming of progenitor
ation markers after incubation in TM plus insulin. Lin- cells from one cell lineage to form differentiated cells be-
eage-uncommitted pluripotent stem cells and lineage- longing to another lineage. However, none of these studies
committed progenitor cells expressed phenotypic markers addressed either the identity or differentiative capability
of their differentiation potential after incubation in TM of their precursor cells prior to experimentation. There-
plus dexamethasone. Therefore, to obtain a true measure fore, the theory of transdifferentiation amounts to more of
of pluripotent stem cell content, it was necessary to sub- an assumption than a proven theory.
tract the expressed differentiation markers in TM plus We believe that it is necessary to understand the histo-
insulin from those generated by TM plus dexamethasone. logical makeup of any given tissue and its inherent em-
These studies demonstrated that approximately 50% of bryological origin (Fig. 1) to realize the potential popula-
the precursor cells residing in a tissue-specific connective tions of precursor cells that may reside in that tissue. For
tissue compartment were lineage-committed progenitor example, the organ of skeletal muscle is composed of skel-
cells specific for that particular tissue. Approximately 40% etal muscle fibers of myotomal somitic or lateral plate
15524892, 2004, 1, Downloaded from https://anatomypubs.onlinelibrary.wiley.com/doi/10.1002/ar.a.10134 by Cochrane Romania, Wiley Online Library on [31/10/2022]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
YOUNG AND BLACK

Figure 3.
90
15524892, 2004, 1, Downloaded from https://anatomypubs.onlinelibrary.wiley.com/doi/10.1002/ar.a.10134 by Cochrane Romania, Wiley Online Library on [31/10/2022]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
ADULT STEM CELLS 91
somatic mesodermal origin, depending on the location of Dixson et al., 1996; Warejcka et al., 1996; Young and
the muscle within the body. The endomysial connective Lucas, 1998; Romero-Ramos et al., 2002) and others
tissue surrounding individual myofibers, the perimysial (Reyes and Verfaillie, 2001; Jiang et al., 2002; Zhao et al.,
connective tissue surrounding muscle fascicles, and the 2003) (see also Table 1) have observed that many tissues
epimysial connective tissues surrounding the entire mus- and organs contain a wide variety of precursor cells. The
cle are of sclerotomal somitic or lateral plate somatic me- precursor cells observed include pluripotent epiblastic-
sodermal origin, depending on the location of the muscle. like stem cells, pluripotent ectodermal stem cells, PPM-
The motor end plates and innervations to the muscle are SCs, and pluripotent endodermal stem cells; and multipo-
of neural crest origin. And the vasculature, blood vessels tent, tripotent, bipotent, and unipotent progenitor cells.
and lymphatic vessels, arise from mesoderm and are thus These precursor cells can be isolated from normal healthy
of somitic or lateral plate somatic mesodermal origin, de- tissues using numerous techniques, i.e., mechanical dis-
pending on location of the muscle. The blood vessels also ruption of the tissues, outgrowth, enzymatic release, dif-
contain circulating differentiated blood cells and blood ferential plating, etc. Then as a cell isolate the precursor
stem cells derived from lateral plate splanchnic meso- cells can be further segregated from differentiated cells by
derm. Based on the histology and embryological origin of cryopreservation, cell sorting, differential centrifugation
the tissue, we would expect to find the following precursor with and without percoll/ficoll gradients, log-phase expan-
cells within the organ of skeletal muscle: pluripotent epi- sion, etc.
blastic-like stem cells (representing all tissues within the In an ongoing series of studies (Young et al., 1993,
organ of skeletal muscle), PPMSCs (skeletal muscle, con- 1998b; Young, 2000, 2004; unpublished observations) we
nective tissue, and vasculature), pluripotent ectodermal have been examining the potential for transdifferentiation
stem cells (motor end plates, peripheral nerves and their in lineage-committed progenitor cell populations. We pos-
supportive glial cells), pluripotent neural crest cells (mo- tulated that if the theory of transdifferentiation were a
tor end plates, peripheral nerves and their supportive reality, then we could induce a unipotent lineage-commit-
glial cells), and multipotent, tripotent, bipotent, and ted progenitor cell clone to exhibit phenotypic expression
unipotent progenitor cells for each of the above-mentioned markers for an alternate tissue lineage. In our first series
tissue types, i.e., skeletal muscle myofibers, connective of experiments we utilized five separate unipotent progen-
tissue coverings, vessels, blood, blood stem cells, and ner- itor cell clones for the fibrogenic, myogenic, adipogenic,
vous tissues. chondrogenic, and osteogenic lineages, respectively. Next,
We (Young, 1977a, 1977b, 1977c, 1983, 2000, 2004; we incubated these clones individually for seven days with
Young et al., 1983a, 1983b, 1983c, 1983d, 1985, 1989a, recombinant or novel inductive factors outside their re-
1989b, 1991, 1992a, 1992b, 1993, 1995, 1998a, 1998b, spective tissue lineages, removed the inductive factor, and
1999, 2001a, 2001b, 2004a, 2004b; Lucas et al., 1993, then incubated the cells with insulin to accelerate pheno-
1995, 1996a, 1996b; Pate et al., 1993; Rogers et al., 1995; typic expression. The inductive factors utilized were fibro-

Fig. 3. Rat clone (A2A2) of postnatal pluripotent mesodermal stem nucleated cells with large ratios of nucleus to cytoplasm. B: Single layer
cells (PPMSC) (A, B, D, G, I, N) and human pluripotent mesodermal stem of nondescript contact-inhibited cells. C: Two linear structures (arrows)
cell lines (CD10⫹, CD13⫹, CD34⫹⫹, CD56⫹, CD90⫹, MHC-I⫹), i.e., hu- containing multiple nuclei. D: Dark structures stained with antibody to
man NHDF2 cells, derived from a dermal biopsy specimen taken from a sarcomeric myosin (MF-20) (arrow). Majority of unstained cells in back-
36 year-old human female, at 80 cell doublings (C, E, F, H, K-M, P-T) and ground are adipocytes (fat cells) (asterisks). E: Mononucleated cells
PAL3 cells, derived from 67 year old human male skeletal muscle biopsy staining intracellularly for smooth muscle alpha-actin (IA4). F: Binucle-
specimen, at 150 cell doublings (J, O) demonstrating mesodermal mor- ated cell co-staining intracellularly for sarcomeric myosin (MF-20) and
phologies. Cells were grown in stem cell medium (SCM), testing medi- smooth muscle alpha-actin (IA4). G: Cells with multiple Oil Red-O
um-6 (TM-6), or TM-6 with 10⫺6 M Dexamethasone (Dex) for 1 to 56 stained intracellular vesicles indicative of saturated neutral lipid-contain-
days. SCM consisted of 89% (v/v) Eagle’s MEM with Earle’s salts, 1% ing adipocytes (arrow). H: Mononucleated cell staining intracellularly for
antibiotics (ab), 10% SS3, pH 7.4. Selected serum-3 (SS3) contained type-II collagen (CIIC1). I: Aggregating nodule of cells (single arrow) with
PDGF-like proliferative and ADF-like inhibitory activities (Young, 2000). pericellular matrix halos staining with antibody to type-IX collagen (D1-
TM-6 consisted of 89, 94, or 98% (v/v) Opti-MEM with 0.01 mM ME; 10, 9). Also note aggregation of unstained adipocytes (double arrows) and
5, or 1% SS9; 1% ab-am, 2 ␮g/ml insulin, at pH 7.4. SS9 contained
individual cells (asterisk) stained with antibody to smooth muscle alpha-
inductive activities resembling those of skeletal muscle morphogenetic
actin (1A4). J: Nodule stained for chondroitin sulfate and keratan sulfate
protein (Sk-MMP), adipocyte morphogenetic protein (AMP), and bone
glycosaminoglycans (Alcian Blue, pH 1.0). K: Mononucleated cells stain-
morphogenetic protein-2 (BMP-2) (Young, 2000). Cultures were grown in
ing intracellularly for osteopontine (MP111). L: Mononucleated cells
SCM (A), TM-6 only (B), TM-6 with 10⫺6 M dexamethasone (Dex) (D, G,
staining intracellularly for bone sialoprotein II (WV1D1). M: Aggregation
I, N), TM-6 with 1%-SS9 (C, E, F, H, K-M, P-T), TM-6 with 10%-SS9 (J),
TM-6 with 1%-SS3 (O) for 1 day (A), 14 days (E), 28 days (M), 42 days (D, of cells with pericellular matrix halos. N: Aggregating nodule of cells
G, J, I, N, O), and 56 days (C, F, H, K, L, P-T). Morphologies and overlaid with horse shoe-shaped extracellular matrix staining with anti-
histochemical and immunochemical staining as noted (see also legend body to bone sialoprotein (WV1D1) (white asterisk). Unstained refractile
to Table 3). Photographed with phase contrast (A-C) and brightfield (D-T) intercellular vesicles belong to adipocytes (single arrows). A diagonally
microscopy, original magnifications ⫻ 200 (A, B), ⫻ 100 (C-L, N, P-R, T), oriented smooth muscle cell stained with an antibody to smooth muscle
⫻ 50 (O), ⫻ 40 (S), ⫻ 25 (M). (From Young et al., 2001a, Clonogenic alpha-actin (1A4) (double arrows) is located in the upper left-hand corner
analysis reveals reserve stem cells in postnatal mammals. I. Pluripotent of photograph. O: Nodules stained for calcium phosphate (von Kossa).
mesenchymal stem cells. Anatomical Record 263:350-360. Reprinted P: Mononucleated cells stained for fibroblast specific protein (1B10).
with permission from Wiley-Liss, Inc. (A, B, D, G, I, N).; Young et al., Q: Mononucleated cells stained for human endothelial cell surface
2001b, Human reserve pluripotent mesenchymal stem cells are present marker (P1H12). R: Mononucleated cells stained for peripheral cell ad-
in the connective tissues of skeletal muscle and dermis derived from hesion molecule (P2B1). S: Mononucleated cells stained for vascular cell
fetal, adult, and geriatric donors. Anat. Rec. 264:51-62, Reprinted with adhesion molecule (P8B1). T: Mononucleated cells stained for E-selectin
permission from Wiley-Liss, Inc. (C, E, F, H, J-M, O-T)). A: Small mono- (P2H3).
15524892, 2004, 1, Downloaded from https://anatomypubs.onlinelibrary.wiley.com/doi/10.1002/ar.a.10134 by Cochrane Romania, Wiley Online Library on [31/10/2022]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
92 YOUNG AND BLACK

blast morphogenetic protein (FMP) to induce fibrogenesis, ment utilizing these same specific inductive factors with a
adipocyte morphogenetic protein (AMP) to induce adipo- seven-day incubation schedule, removal of the inductive
genesis, BMP-2 to induce chondrogenesis and osteogene- factor, and then incubation of the cells with insulin to
sis, and Sk-MMP to induce skeletal myogenesis. In all accelerate phenotypic expression. In all experiments the
cases examined, the tissue-specific progenitor clones induced unipotent progenitor cell lines maintained their
maintained their original lineage commitment program- originally induced lineage commitment programming.
ming and exhibited tissue-specific phenotypic expression Again, these results suggested that once a cell is commit-
markers, even in the presence of inductive factors outside ted to a particular tissue lineage, it would not change its
their respective lineages, i.e., FMP, AMP, BMP-2, or Sk- genetic programming.
MMP. These results suggested that once a cell is commit- In a third series of experiments we mixed a clone of
ted to a particular tissue lineage, it would not change its lineage-uncommitted pluripotent stem cells with one or
genetic programming, i.e., revert to a more primitive state more clones of lineage-committed progenitor cells. We
and express markers for a different tissue lineage. then incubated these mixed cultures with one or more
In a second series of experiments we induced the forma- tissue-specific induction factors (same as above) with and
tion of unipotent lineage-committed progenitor cells from without insulin. The results demonstrated that the pro-
a lineage-uncommitted PPMSC clone using the same tis- genitor cells were responsive to insulin and would express
sue-specific lineage induction factors as above (i.e., FMP, their lineage-committed phenotype in the presence of in-
AMP, BMP-2, Sk-MMP). After establishment of the unipo- sulin or their respective lineage induction factor. In con-
tent progenitor cell lines, we repeated the above experi- trast, the lineage-uncommitted pluripotent stem cell clone
responded to the lineage-specific induction factor, but not
to insulin. For example, a myogenic clone was mixed with
a chondrogenic clone and a pluripotent stem cell clone.
The mixed culture was then incubated with FMP and
insulin, FMP alone, and insulin alone. The mixed culture
incubated with FMP and insulin demonstrated phenotypic
expression markers for myogenesis, chondrogenesis, and
fibrogenesis. The mixed culture incubated with FMP alone
demonstrated only phenotypic expression markers for fi-
brogenesis. The mixed culture incubated with insulin
alone demonstrated phenotypic expression markers for
myogenesis and chondrogenesis. These results suggested
that the myogenic and chondrogenic clones within the
mixed culture were responding to insulin, while the plu-
ripotent stem cell clone in the mixed culture was respond-
ing to FMP.

Fig. 4. Human (CD10⫹, CD66e⫹) (A, C, E- G, I-K) and Rat clone


(A2B2-scl-40) (B, D, H, L) postnatal pluripotent epiblastic-like stem cells
(PPELSC) demonstrating embryonic markers. PPELSC lines were grown
for 24 hours to 8 weeks in serum-free testing medium (TM) only, TM with
10% SS3, or TM with 2 ␮g/ml insulin. TM consisted of 99% (v/v)
Opti-MEM, 0.01 mM ␤ME, 1% ab-am, pH 7.4. 10% SS3 contains
proliferative activity resembling that of PDGF and inhibitory activity
resembling that of ADF (Young, 2000, 2004; Young et al., 2004a). In this
particular series a human (CD10⫹, CD66e⫹) PPELSC line and a rat clone
(A2B2-scl-40) PPELSC line were incubated in TM only (A, B), TM with
10% SS3 (C, D), or TM with insulin (E-L) for 24 hours (A, B) or seven days
(C-L). Morphologies and immunochemical staining as noted (see also
legend to Table 3). Photographed with phase contrast (A-D) and bright-
field microscopy (E-L). Original magnifications, ⫻ 400 (A, E, F), ⫻ 200 (B,
D, G-K) and ⫻ 100 (C, L). A: Very small cells with high ratios of nucleus
to cytoplasm. Stellate shape denotes an attached cell, linear shape is
indicative of mobile cell. B: Very small cells with high ratios of nucleus to
cytoplasm. Linear shape is indicative of mobile cell. C: Multilayered
confluent cells maintaining stellate morphology. D: Multilayered conflu-
ent cells maintaining stellate morphology. E: Moderate to heavy staining
for stage specific embryonic antigen-1 (SSEA-1). F: Moderate staining
for stage specific embryonic antigen-3 (SSEA-3). G: Moderate to heavy
staining for stage specific embryonic antigen-4 (SSEA-4). H: Moderate
to heavy staining for stage specific embryonic antigen-4 (SSEA-4).
I: Moderate to heavy staining for human carcinoembryonic antigen
(HCEA). J: Moderate to heavy staining for human carcinoembryonic
antigen (CD66e). K: Moderate staining for carcinoembryonic antigen cell
adhesion molecule-1 (CEA-CAM1). L: Moderate to heavy staining for
carcinoembryonic antigen cell adhesion molecule-1 (CEA-CAM1).
15524892, 2004, 1, Downloaded from https://anatomypubs.onlinelibrary.wiley.com/doi/10.1002/ar.a.10134 by Cochrane Romania, Wiley Online Library on [31/10/2022]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
ADULT STEM CELLS 93

Fig. 5. A human (CD10⫹, CD66e⫹) PPELSC line (A, C, E, G, H, J, L, F: Mononucleated cells showing moderate to heavy intracellular staining
M, O, S) and rat clone (A2B2-scl-40) PPELSC line (B, D, F, I, K, N, P-R) for neurons (8A2). G: Mononucleated cells showing moderate to heavy
were induced to express ectodermal (neuroectodermal and surface ec- intracellular staining for nestin (HNES). H: Mononucleated cell showing
todermal) phenotypic expression markers. The PPELSC lines were heavy intracellular staining for nestin (MAB353). I: Mononucleated cells
plated at 103 cells per well in 1% gelatin-coated 96-well plates and showing heavy intracellular staining for neuronal nestin (Rat-401).
grown for 24 hours to 8 weeks in testing medium (TM) only; TM with 10⫺6 J: Mononucleated cells showing low to moderate intracellular staining
to 10⫺10 M Dex; TM with 10, 5, or 1% SS12; or TM with 10⫺6 to 10⫺10 for b-tubulin-III (T8660). K: Mononucleated cells showing moderate to
M Dex and 10, 5, 3, or 1%-SS12. TM consisted of 89, 94, 96, or 98% heavy intracellular staining for b-tubulin-III (T8660). L: Mononucleated
(v/v) Opti-MEM with 0.01 mM ␤ME, 1% ab-am, 2 ␮g/ml insulin, at pH cells showing moderate to heavy intracellular staining for neuroglia (oli-
7.4. Five, 3, and 1% SS12 contains ectodermal inductive activities. In godendrocytes and astroglia) (CNPase). M: Mononucleated cell showing
this particular series human cells were grown for 7 days in TM with 10⫺6 heavy intracellular staining for oligodendrocytes (Rip). N: Mononucleated
M Dex and 1% SS12. Morphologies and immunochemical staining as cell showing heavy intracellular staining for oligodendrocytes (Rip).
noted (see also legend to Table 3). Photographed with brightfield mi- O: Mononucleated cells showing heavy intracellular staining for neuronal
croscopy, original magnifications, ⫻ 200 (A, C-H, J, L-O, S), ⫻ 100 (B, I, expression marker (S-100). P: Mononucleated cells showing moderate
P-R), ⫻ 40 (K). A: Mononucleated cells moderately to heavily stained for intracellular staining for neuronal expression marker (S-100). Q: Mono-
neural precursor cell expression marker (FORSE-1). B: Mononucleated nucleated cells showing moderate to heavy intracellular staining for
cells heavily stained for neural precursor cell expression marker (FORSE- neuronal vimentin for radial cells and radial glial cells (40E-C). R: Mono-
1). C: Mononucleated cells showing heavy intracellular staining for neu- nucleated cells showing moderate to heavy intracellular staining for
rofilaments (RT-97). D: Mononucleated cells showing moderate to heavy ganglion cells (TuAg1). S: Mononucleated cells showing low to moderate
intracellular staining for neurofilaments (RT-97). E: Mononucleated cells staining for keratinocytes (VM-1).
showing moderate to heavy intracellular staining for neurons (8A2).
15524892, 2004, 1, Downloaded from https://anatomypubs.onlinelibrary.wiley.com/doi/10.1002/ar.a.10134 by Cochrane Romania, Wiley Online Library on [31/10/2022]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
94 YOUNG AND BLACK

Fig. 6. Human postnatal pluripotent epiblastic-like stem cell line heavy intracellular staining for smooth muscle ␣-actin (IA4). D: Mono-
(CD10⫹, CD66e⫹) (B, C, D, H, M, N, O, P) and postnatal rat pluripotent nucleated and binucleated cells showing moderate intracellular staining
epiblastic-like stem cell clone (A2B2-scl-40) (A, E, F, G, I, J, K, L) induced for cardiotin (cardiac myocytes, MAB 3252). E: Mononucleated cells
to express mesodermal phenotypic expression markers. Multiple demonstrating heavy intracellular staining for bone sialoprotein II
PPELSC lines were plated at 103 cells per well in 1% gelatin-coated (WV1D1). F: Nodule of cells demonstrating extracellular staining for
96-well plates and grown for 24 hours to 8 weeks in testing medium (TM) osteopontine (MP111). G: Nodule of cells demonstrating extracellular
only; TM with 10⫺6 to 10⫺10 M Dex; TM with 10, 5, 3, or 1% SS9 or 1% staining for calcium phosphate using the von Kossa procedure (vK).
SS3; or TM with 10⫺6 to 10⫺10 M Dex and 10, 5, 3, or 1% SS9 or 1% H: Mononucleated cells with heavy intracellular staining for cartilage-
SS3. TM consisted of 89, 94, 96, or 98% (v/v) Opti-MEM with 0.01 mM specific collagen pro type-II (CIIC1). I: Single nodule of cells demonstrat-
␤ME with 1% ab-am, 2 ␮g/ml insulin, at pH 7.4. Ten, 5, 3, and 1% SS9 ing moderate to heavy extracellular staining for cartilage-specific colla-
contains inductive activities resembling that of Sk-MMP, AMP, BMP-2, gen type-II (HC-II). J: Three nodules demonstrating heavy extracellular
and endothelial inductive activity, while 1% SS3 contains inductive staining for cartilage-specific collagen type-IX (D1-9). K: Two nodules
activity resembling that of Sm-MMP (Young, 2004; Young et al., 2004a). demonstrating extracellular staining for sulfated glycosaminoglycan
In this particular series PPELSCs were grown for 1 week (A, B, C, D, L-P), chains of proteoglycans (Alcian Blue-O, pH 1.0). L: Mononucleated cells
2 weeks (E, H), or 4 weeks (F, G, I- K) in TM with 10⫺8 M Dex plus insulin with moderate to heavily stained intracellular vesicles demonstrating
plus 1% SS9 (A, B, L-P), 10⫺8 M Dex plus insulin plus 1% SS3 (C), or saturated neutral lipids (Oil Red-O), indicative of adipocytes. M: Three-
10⫺7 M Dex plus insulin plus 3% SS9 (E- K). Morphologies and immu- dimensional tubular structure demonstrating moderate to heavy staining
nochemical staining as noted (see also legend to Table 3). Photographed for CD146, endothelial cells (H-endo). N: Mononucleated cell demon-
with brightfield microscopy; original magnifications, ⫻ 200 (C, L, N), ⫻ strating heavy staining for VCAM, vascular (endothelial) cell adhesion
160 (O), ⫻ 100 (A, B, D, E, F, G, H, I, K, M, P), ⫻ 40 (J). A: Mononucleated molecule (P8B1). O: Mononucleated cells demonstrating low to moder-
cells showing heavy intracellular staining for myogenin (F5D). B: Mono- ate staining for CD62e, (endothelial) selectin-E (P2H3). P: Mononucle-
nucleated and binucleated cells and multinucleated linear and branched ated cells demonstrating moderate staining for CD44, hyaluronate re-
structures showing moderate to heavy intracellular staining for anti- ceptor (H4C4).
skeletal muscle fast myosin (MY-32). C: Mononucleated cells showing

Throughout all our studies trying to prove the validity of of another organ (review, Forbes et al., 2002; Poulsom et
transdifferentiation we were never able to repeat trans- al., 2002; Tsai et al., 2002) are actually due to contamina-
differentiation results utilizing known populations of pre- tion of the tissue isolate by unrecognized progenitor cells
cursor cells with known differentiation potentials. There- and/or pluripotent stem cells that are stimulated to form
fore, based on the above experiments, we would propose new progenitor cells of a different tissue lineage. For ex-
that differentiated progenitor cells do not undergo trans- ample, we propose that pluripotent stem cells (and/or he-
differentiation or dedifferentiation. We now believe that matopoietic progenitor cells) present in skeletal muscle
the experimental results that appear to require transdif- could give rise to the blood cells initially reported by
ferentiation or dedifferentiation of progenitor cells to oc- Jackson et al. (1999). Indeed, a subsequent analysis by the
cur during adult tissue restoration when progenitor cells same research group (McKinney-Freeman et al., 2002), in
derived from one organ are reprogrammed to form tissues an elegant study demonstrated that CD45⫹ (hematopoi-
15524892, 2004, 1, Downloaded from https://anatomypubs.onlinelibrary.wiley.com/doi/10.1002/ar.a.10134 by Cochrane Romania, Wiley Online Library on [31/10/2022]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
ADULT STEM CELLS 95

Fig. 7. Human (CD10⫹, CD66e⫹) postnatal pluripotent epiblastic-like cellular staining for somatostatin in ␦-cells of endocrine pancreas
stem cell line (B, D, G, N, O) and rat postnatal pluripotent epiblastic-like (11180). F: Cellular aggregation showing moderate to intense intracellu-
stem cell clone, A2B2-scl-40 (A, C, E, F, H-M, P) (PPELSCS) induced to lar staining for ductal cells of exocrine pancreas (CK-19). G: Mononucle-
express endodermal phenotypic markers. Multiple PPELSC lines were ated cells showing light to moderate intracellular staining for bile cana-
plated at 103 cells per well in 1% gelatin-coated 96-well plates and licular cells of liver (HA4c19). H: Nodule showing heavy intracellular
grown for 24 hours to 8 weeks in testing medium (TM) only; TM with 10⫺6 staining for progenitor cells, biliary epithelial cells, and oval cells of liver
to 10⫺10 M Dex; TM with 15 or 10% SS12; or TM with 10⫺6 to 10⫺10 M (OC2). I: Cellular aggregation and individual diffuse mononucleated cells
Dex and 15 or 10% SS12. Testing medium consisted of 74 or 89% (v/v) showing moderate to heavy intracellular staining for progenitor cells and
Opti-MEM with 0.01 mM ␤ME with 1% ab-am, 2 ␮g/ml insulin, at pH 7.4. biliary epithelial cells of liver (OC3). J: Cellular aggregation and individual
15 and 10% SS12 contains endodermal inductive activities. In this diffuse mononucleated cells showing moderate to intense intracellular
particular series PPELSCs were incubated for 1 week (A, D, E, G, N, O), staining for progenitor cells and biliary epithelial cells of liver (OC4).
2 weeks (F, K, L, Q) 3 weeks (I, J, M), 4 weeks (B, P), or 5 weeks (C, H) K: Diffuse mononucleated cells showing moderate to heavy intracellular
in TM with 15% SS12 and 10⫺6 M Dex. Morphologies and immuno- staining for progenitor cells and biliary epithelial cells of liver (OC5).
chemical staining as noted (see also legend to Table 3). Photographed L: Diffuse mononucleated cells showing moderate to intense intracellular
with brightfield microscopy, original magnifications, ⫻ 200 (A), ⫻ 100 staining for progenitor cells and biliary epithelial cells of liver (OC10).
(C-P). A: Mononucleated and binucleated cells showing intense intra- M: Diffuse and aggregated cells showing moderate to intense intracel-
cellular staining for rat-specific alpha-fetoprotein (RAFP). B: Mononucle- lular staining for cytoplasm of liver hepatocytes (H.4). N: Mononucleated
ated cell showing heavy intracellular staining for gastro-intestinal epi- cells showing moderate to intense staining for liver hepatocyte cell
thelium (HESA). C: Nodular aggregation showing moderate intracellular surface marker (H.1). O: Mononucleated cells showing light to moderate
staining for pro-insulin in ␤-cells of endocrine pancreas (YM-PS5088). to intense staining for progenitor cells, canalicular cells, and biliary
D: Mononucleated cells showing moderate to intense intracellular stain- epithelial cells of liver (DPP-IV). P: Diffuse cells showing moderate to
ing for glucagon in ␣-cells of endocrine pancreas (YM-PS087). E: Indi- heavy intracellular staining for biliary epithelial cells, oval cells, hepato-
vidual diffuse mononucleated cells showing moderate to intense intra- cyte canalicular cells of liver (OV6).

etic) stem cells residing in skeletal muscle gave rise to all three types of precursor cell, since all of these types of
blood cells, whereas the CD45– (nonhematopoietic) stem cells are CD45– by flow cytometric analysis (Young, 2000,
cells residing in skeletal muscle formed muscle. However, 2004; Young et al., 2001b, 2004a).
these investigators did not examine the CD45– stem cells Along a similar vein as above, Petersen et al. (1999) and
of skeletal muscle to determine their full differentiation Yang et al. (2002) described an adult progenitor cell from
potential, i.e., whether they would form other cell/tissue bone marrow that would form liver hepatocytes and an
types in addition to skeletal muscle. Several possibilities adult hepatic oval (stem) cell from liver that would form
exist. The CD45– stem cells could be progenitor cells com- pancreatic islets (pancreatic endocrine hormone-produc-
mitted to the myogenic lineage. Alternatively, the CD45– ing cells). In both instances they failed to characterize
stem cells could be PPMSCs or pluripotent epiblastic-like their identified stem cells for pluripotency prior to their
stem cells. The CD45– cells could also be a combination of experimental studies. They then used the theory of trans-
15524892, 2004, 1, Downloaded from https://anatomypubs.onlinelibrary.wiley.com/doi/10.1002/ar.a.10134 by Cochrane Romania, Wiley Online Library on [31/10/2022]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
96 YOUNG AND BLACK

Fig. 8. Flow chart of adult stem cell hierarchy and nomenclature, (PPMSCs), pluripotent paraxial mesodermal stem cells (PPPMSCs), plu-
from most primitive (top) to most differentiated (bottom). Adult stem cells ripotent intermediate mesodermal stem cells (PPIMSCs), pluripotent
are divided into two major categories: pluripotent stem cells (PPSCs) lateral plate mesodermal stem cells (PPLPMSCs), and pluripotent
and progenitor cells (ProgCs). Characteristics of each category are as endodermal stem cells (PPEndoSCs). PPELSCs have the capacity to
follows. PPSCs are lineage-uncommitted, telomerase positive, and have form any somatic cell of the body and display embryonic phenotypic
extensive capabilities for self-renewal. They are composed of at least the markers in the lineage-uncommitted state. Pluripotent germ layer stem
following categories, i.e., pluripotent epiblastic-like stem cells cells have the capacity to form any cell type within their respective germ
(PPELSCs), pluripotent germ layer lineage stem cells and subcategories layer lineage and display lineage-specific phenotypic markers. ProgCs
of pluripotent germ layer lineage stem cells, such as pluripotent ecto- are lineage-committed, e.g., committed to forming cells exclusive to
dermal stem cells (PPEctoSCs), pluripotent surface ectodermal stem particular germ layer lineages, and conform to Hayflick’s Limit of 50-70
cells (PPSurfEctoSCs), pluripotent neuroectodermal stem cells (PPNeu- population doublings before cell senescence and cellular death. They
roEctoSCs), pluripotent neural tube stem cells (PPNT-SCs), pluripotent are composed of multipotential, tripotential, bipotential, and unipotential
neural crest stem cells (PPNC-SCs), pluripotent mesodermal stem cells precursor cells for their respective germ layer lineages.

differentiation to explain the formation of tissues other begin with the characterization of the composition and
than that from which the cells were isolated. Based on our complete differentiation potentials of the precursor cells
studies (above, Young et al., 1998a, 1998b, 2001b, 2004a; utilized. For those investigators that still maintain that
Young, 2000, 2004) and those of others (Reyes and Ver- transdifferentiation is a valid hypothesis, we would sug-
faillie, 2001; Jiang et al., 2002), we believe that the start- gest a thorough characterization of the differentiation po-
ing populations of tissue-specific precursor cells in the tential of their cells prior to the onset of experimentation.
studies by Goodell’s group (Jackson et al., 2000; McKin- Unless this information is made available, it is very diffi-
ney-Freeman et al., 2002) and Petersen’s group (Petersen cult to interpret the results of such experiments. Due to
et al., 1999; Yang et al., 2002) could also be explained by the ubiquitous locations of identified precursor cells, we
the presence of pluripotent stem cells (and/or lineage- would also suggest that future experiments addressing
committed progenitor cells for other tissue lineages) as the issue of precursor cell function begin with a complete
resident precursor cell populations within the respective characterization of the isolated precursor cells, including
differentiated tissues examined. Thus, it is not necessary their inherent proliferation potential with respect to Hay-
to invoke the concept of transdifferentiation to explain the flick’s limit and their inherent differentiation potential
results obtained by these investigative groups. with respect to the embryogenetic developmental se-
We suggest that future experiments addressing the is- quence. The designated nomenclature for the isolated pre-
sue of transdifferentiation of adult precursor cells should cursor cells should then be based on the functionality of
15524892, 2004, 1, Downloaded from https://anatomypubs.onlinelibrary.wiley.com/doi/10.1002/ar.a.10134 by Cochrane Romania, Wiley Online Library on [31/10/2022]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
ADULT STEM CELLS 97
the cells, i.e., proliferation and differentiation potentials, BMP-2 and rhBMP-4 were the generous gifts of I.K. Mout-
rather than their location within the tissue. satsos, Genetics Institute, Inc., Cambridge, MA. The
PDECGF, IGF-II, IGF-I, PDGF-AA, and PDGF-BB were
TISSUE ENGINEERING generous gifts from G.F. Pierce, Amgen, Thousand Oaks,
CA. Sk-MMP, Sm-MMP, AMP, FMP, SIF, and ADF were
With respect to the treatment of trauma and disease,
the generous gifts of L. Rifkin, MorphoGen Pharmaceuti-
adult (postnatal) precursor cells can be very useful for
cals, Inc., New York, NY. We thank D. Hixson for the
therapies involving autologous transplantation. Precursor
cells can be isolated from newborn to geriatric individuals, generous gift of his antibodies OC2, OC3, OC4, OC5,
including patients awaiting treatment. Use of autologous OC10, DPP-IV, OV6, H-1, H-4, and LAP for endodermal
precursor cells circumvents the inherent morbidity and lineage cells and CEA-CAM1 for embryonic cells. The
mortality associated with HLA mismatches that require following antibodies were obtained from the Developmen-
the use of immunosuppressant drugs. Adult pluripotent tal Studies Hybridoma Bank developed under the aus-
stem cells can be harvested from a small biopsy of easily pices of the NICHD and maintained by the University of
accessible tissues, such as skeletal muscle or dermis. Plu- Iowa, Department of Biological Sciences, Iowa City, IA:
ripotent stem cells are telomerase positive, indicating that MC480, MC631, and MC813–70 developed by D. Solter;
vast quantities of cells can be produced from a few har- FORSE-1 developed by P. Patterson; RAT-401 and Rip
vested cells. Pluripotent stem cells can be stored for long developed by S. Hockfield; RT-97 developed by J. Wood;
periods of time with minimal loss of cell viability, pluripo- 8A2 developed by V. Lemmon; SV2 developed by K.M.
tentiality, and function. Progenitor cells derived from plu- Buckley; VM-1 developed by V.B. Morhenn; 151-Ig devel-
ripotent stem cells mimic native progenitor cells with re- oped by A. Hubbard; 40E–C developed by A. Alvarez-
spect to loss of differentiative potential with increased Buylla; F5D developed by W.E. Wright; MF-20 and
lineage commitment and adherence to Hayflick’s limit of ALD-58 developed by D.A. Fischman; A4.74 developed by
50 –70 population doublings before programmed cell se- H.M. Blau; CIIC1 developed by R. Holmdahl and K. Ru-
nescence and cell death. Pluripotent ectodermal stem cells bin; D1–9 developed by X.-J. Ye and K. Terato; 9/30/8A4
have the potential to form neurons, neuroglia, peripheral and 12/21/1C6 developed by B. Caterson; 12C5 developed
nerves, neuroendocrine cells, epidermis, and other cells of by R.A. Asher; WV1D1 (9C5) and MP111B101 developed
the ectodermal germ layer lineage. PPMSCs have the by M. Solursh and A. Frazen; P2B1 and P2H3 developed
potential to form skeletal muscle, smooth muscle, cardiac by E.A. Wayner and G. Vercellotti; P8B1 developed by
muscle, fat, cartilage, bone, tendons, ligaments, dermis, E.A. Wayner and T. LeBien; HA4c19 developed by A.
connective tissues, blood vessels, hematopoietic cells, and Hubbard; Hermes-1 developed by E.C. Butcher; and
other cells of the mesodermal germ layer lineage. And H4C4, H5A5, H5C5, H5C6, and H5A4 developed by J.T.
pluripotent endodermal stem cells have the potential to August and J.E.K. Hildreth.
form hepatocytes, biliary cells, canalicular cells, pancre-
atic islet cells, exocrine pancreatic cells, gastrointestinal LITERATURE CITED
epithelial cells, nonpancreatic endocrine cells, and other Ailhaud G, Grimaldi P, Negrel R. 1992. Cellular and molecular as-
cells of the endodermal germ layer lineage. Further, adult pects of adipose tissue development. Annu Rev Nutr 12:207–233.
pluripotent epiblastic-like stem cells have the potential to Alison MR, Poulsom R, Jeffery R, Dhillon AP, Quaglia A, Jacob J,
form cells from all three primary germ layer lineages, i.e., Novelli M, Prentice G, Williamson J, Wright NA. 2000. Hepatocytes
ectoderm, mesoderm, and endoderm. Additional studies from non-hepatic adult stem cells. Nature 406:257.
are planned or currently under way (Young et al., 2004a) Alvarez-Buylla A, Buskirk DR, Nottebohm F. 1987. Monoclonal anti-
to determine whether adult reserve pluripotent stem cells body reveals radial glia in adult avian brain. J Comp Neurol 264:
can be used in therapeutic treatments for various condi- 159 –170.
Asher RA, Scheibe RJ, Keiser HD, Bignami A. 1995. On the existence
tions such as Parkinson’s disease, stroke, spinal cord re-
of a cartilage-like proteoglycan and link proteins in the central
pair, blood transfusions, bone marrow transplants, diabe- nervous system. Glia 13:294 –308.
tes mellitus and other endocrine diseases, renal failure, August JT, Hildreth JEK. 2002. In: Developmental studies hybridoma
arthritis, muscular dystrophy, vascular ischemia, and bank product catalog, year 2002. Iowa City, IA: University of Iowa.
myocardial infarctions. p 11.
Bader D, Masaki T, Fischman DA. 1982. Immunochemical analysis of
ACKNOWLEDGMENTS myosin heavy chain during avian myogenesis in vivo and in vitro.
J Cell Biol 95:763–770.
I especially thank Paul A. Lucas for the generous ex- Banerjee A, Roach MC, Trcka P, Luduena RF. 1990. Increased micro-
change of ideas, reagents, and unpublished observations. I tubule assembly in bovine brain tubulin lacking the type III isotype
thank my collaborators, co-authors, and their technical of beta-tubulin. J Biol Chem 265:1794 –1799.
assistants for their insight and work ethic. We thank Julie Banerjee A, Roach MC, Wall KA, Lopata MA, Cleveland DW, Lu-
A. Floyd and Nicholas L. Henson for cell culture assis- duena RF. 1988. A monoclonal antibody against the type II isotype
tance; Kris Detmer and Angela Thompson for assistance of beta-tubulin. Preparation of isotypically altered tubulin. J Biol
with the telomerase and Oct-4 analyses; Cecile Duplaa Chem 263:3029 –3034.
and Catherine Moreau for ␤-galactosidase transfections; Baudier J, Glasser N, Gerard D. 1986. Ions binding to S100 proteins.
I. Calcium- and zinc-binding properties of bovine brain S100 alpha
and Nicholas L. Henson and John Knight for photographic
alpha, S100a (alpha beta), and S100b (beta beta) protein: Zn2⫹
assistance. The MyoD1 clone, maintained in the expres- regulates Ca2⫹ binding on S100b protein. J Biol Chem 261:8192–
sion vector pVZCII-␣3 scribe, and the myogenin cDNA 8203.
probe, subcloned into Bluescribe pEMSV-␣2 M13, were Beauchamp JR, Heslop L, Yu DSW, Tajbakhsh S, Kelly RG, Wernig A,
generous gifts from Dr. W.E. Wright. Bovine nasal septum Buckingham ME, Partridge TA, Zammit PS. 2000. Expression of
A1D1 proteoglycan and the 5D4 anti-keratan sulfate an- CD34 and Myf5 defines the majority of quiescent adult skeletal
tibody were the kind gifts of Dr. J.H. Kimura. The rh- muscle satellite cells. J Cell Biol 151:1221–1233.
15524892, 2004, 1, Downloaded from https://anatomypubs.onlinelibrary.wiley.com/doi/10.1002/ar.a.10134 by Cochrane Romania, Wiley Online Library on [31/10/2022]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
98 YOUNG AND BLACK

Beresford JN. 1989. Osteogenic stem cells and the stromal system of Debus E, Weber K, Osborn M. 1983b. Monoclonal antibodies to
bone and marrow. Clin Orthop 240:270 –280. desmin, the muscle-specific intermediate filament protein. EMBO J
Bjerknes M, Cheng H. 2002. Multipotential stem cells in adult mouse 2:2305–2312.
gastric epithelium. Am J Physiol Gastrointest Liver Physiol 283: Dell’Agnola C, Rabascio C, Mancuso P, Capillo M, Pruneri G, Gobbi A,
G767–G777. Minucci S, Ronzoni S, Volorio S, Calabrese L, Tradati N, Martinelli
Bjornson CRR, Rietze RL, Reynolds BA, Magli MC, Vescovi AL. 1999. G, Shultz L, Bertolini F. 2002. In vitro and in vivo hematopoietic
Turning brain into blood: a hematopoietic fate adopted by adult potential of human stem cells residing in muscle tissue. Exp He-
neural stem cells in vivo. Science 283:534 –537. matol 30:905–914.
Bonner-Weir S, Taneja M, Weir GC, Tatarkiewicz K, Song K-H, Deng W, Obrocka M, Fischer I, Prockop DJ. 2001. In vitro differenti-
Sharma A, O’Neil JJ. 2000. In vitro cultivation of human islets from ation of human marrow stromal cells into early progenitors of
expanded ductal tissue. Proc Natl Acad Sci USA 97:7999 – 8004. neural cells by conditions that increase intracellular cyclic AMP.
Bosch P, Musgrave DS, Lee JY, Cummins J, Shuler T, Ghivizzani TC, Biochem Biophys Res Commun 282:148 –152.
Evans T, Robbins TD, Huard J. 2000. Osteoprogenitor cells within Detn CL, Latchman DS. 1993. In: Latchman DS, editor. Transcription
skeletal muscle. J Orthop Res 18:933–944. factors: a practical approach. New York: Oxford University Press. p
Bowerman SG, Taylor SS, Putnam L, Young HE, Lucas PA. 1991. 1–26.
Transforming growth factor-␤ (TGF-␤) stimulates chondrogenesis Dittel BN, McCarthy JB, Wayner EA, LeBien TW. 1993. Regulation of
in cultured embryonic mesenchymal cells. Surg Forum XLII:535– human B-cell precursor adhesion to bone marrow stromal cells by
536. cytokines that exert opposing effects on the expression of vascular
Brazelton TR, Rossi FM, Keshet GI, Blau HM. 2000. From marrow to cell adhesion molecule-1 (VCAM-1). Blood 81:2272–2282.
brain: expression of neuronal phenotypes in adult mice. Science Dixson K, Murphy RW, Southerland SS, Young HE, Dalton ML,
290:1775–1779. Lucas PA. 1996. Recombinant human bone morphogenetic pro-
Burgeson RE, Hollister DW. 1979. Collagen heterogeneity in human teins-2 and 4 (rhBMP-2 and rhBMP-4) induce several mesenchymal
cartilage: identification of several new collagen chains. Biochem phenotypes in culture. Wound Rep Reg 4:374 –380.
Biophys Res Commun 87:1124 –1131. Doetsch F, Caille I, Lim DA, Garcia-Verdugo JM, Alvarez-Buylla A.
Butcher EC. 2002. In: Developmental studies hybridoma bank prod- 1999. Subventricular zone astrocytes are neural stem cells in the
uct catalog, year 2002. Iowa City, IA: University of Iowa. p 12. adult mammalian brain. Cell 97:703–716.
Campion DR. 1984. The muscle satellite cell: a review. Int Rev Cytol Donovan PJ, Gearhart J. 2001. The end of the beginning for pluripo-
87:225–251. tent stem cells. Nature 414:92–927.
Drazba J, Pierce M, Lemmon V. 1991. Studies of the developing chick
Caplan AI. 1991. Mesenchymal stem cells. J Orthop Res 9:641– 650.
retina using monoclonal antibody 8A2 that recognizes a novel set of
Caplan AI, Elyaderani M, Mochizuki Y, Wakitani S, Goldberg V.
gangliosides. Dev Biol 145:154 –163.
1997. Principles of cartilage repair and regeneration. Clin Orthop
Eglitis MA, Mezey E. 1997. Hematopoietic cells differentiate into both
Rel Res 342:254 –269.
microglia and macroglia in the brains of adult mice. Proc Natl Acad
Carlson BM. 1999. In: Human embryology & developmental biology,
Sci USA 94:4080 – 4085.
2nd edition. St. Louis: Mosby Inc.
Eisenberg CA, Markwald RR. 1997. Mixed cultures of avian blasto-
Caterson B, Baker JR, Christner JE, Lee Y, Lentz M. 1985. Monoclo-
derm cells and the quail mesoderm cell line QCE-6 provide evidence
nal antibodies as probes for determining the microheterogeneity of
for the pluripotentiality of early mesoderm. Dev Biol 191:167–181.
the link proteins of cartilage proteoglycan. J Biol Chem 260:11348 –
Eisenberg CA, Gourdie RG, Eisenberg LM. 1997. Wnt-11 is expressed
11356.
in early avian mesoderm and required for the differentiation of the
Caterson B. 2001. In: Developmental studies hybridoma bank product
quail mesoderm cell line QCE-6. Development 124:525–536.
catalog, year 2001, NICHHD. Iowa City, IA: University of Iowa. p
Estrera VT, Luo W, Phan D, Earley K, Hixson DC. 1999. The cyto-
12.
plasmic domain of C-CAM1 tumor suppressor is necessary and
Clarke DL, Johansson CB, Wilbertz J, Veress B, Nilsson E, Karlstrom sufficient for suppressing the tumorigenicity of prostate cancer
H, Lendahl U, Frisen J. 2000. Generalized potential of adult neural cells. Biochem Biophys Res Commun 263:797– 803.
stem cells. Science 288:1660 –1663. Faris RA, McEntire KD, Thompson NL, Hixson DC. 1990. Identifica-
Colter DC, Class R, DiGirolamo CM, Prockop DJ. 2000. Rapid expan- tion and characterization of a rat hepatic oncofetal membrane gly-
sion of recycling stem cells in cultures of plastic-adherent cells from coprotein. Cancer Res 50:4755– 4763.
human bone marrow. Proc Natl Acad Sci USA 97:3213–3218. Faris RA, Monfils BA, Dunsford HA, Hixson DC. 1991. Antigenic
Colter DC, Sekiya I, Prockop DJ. 2001. Identification of a subpopula- relationship between oval cells and a subpopulation of hepatic foci,
tion of rapidly self-renewing and multipotential adult stem cells in nodules and carcinomas induced by the ‘Resistant Hepatocyte’
colonies of human marrow stromal cells. Proc Natl Acad Sci USA model system. Cancer Res 51:1308 –1317.
98:7841–7845. Feany MB, Lee S, Edwards RH, Buckley KM. 1992. The synaptic
Cornelius JG, Tchernev V, Kao KJ, Peck AB. 1997. In vitro-generation vesicle protein SV2 is a novel type of transmembrane transporter.
of islets in long-term cultures of pluripotent stem cells from adult Cell 70:861– 867.
mouse pancreas. Horm Metab Res 29:271–277. Ferrari G, Cusella-De Angelis G, Coletta M, Paolucci E, Stornaiuolo
Cruess RL. 1982. The musculoskeletal system: embryology, biochem- A, Cossu G, Mavilio F. 1998. Muscle regeneration by bone marrow-
istry, and physiology. New York: Churchill Livingston. p 1–33, derived myogenic progenitors. Science 279:1528 –1530.
109 –169, 255–287. Forbes SJ, Vig P, Poulsom R, Wright NA, Alison MR. 2002. Adult stem
Damjanov I, Fox N, Knowles BB, Solter D, Lange BH, Fraley EE. cell plasticity: new pathways of tissue regeneration become visible.
1982. Immunohistochemical localization of murine stage specific Clin Sci (Lond) 103:355–369.
embryonic antigens in human testicular germ cell tumors. Am J Franke FE, Schachenmayr W, Osborn M, Altmannsberger M. 1991.
Pathol 108:225–230. Unexpected immunoreactivities of intermediate filament antibodies
Deasy BM, Jankowski RJ, Huard J. 2001. Muscle-derived stem cells: in human brain and brain tumors. Am J Pathol 139:67–79.
characterization and potential for cell-mediated therapy. Blood Frid MG, Shekhonin BV, Koteliansky VE, Glukhova MA. 1992. Phe-
Cells Mol Dis 27:924 –933. notypic changes of human smooth muscle cells during development:
Deasy BM, Qu-Peterson Z, Greenberger JS, Huard J. 2002. Mecha- late expression of heavy caldesmon and calponin. Dev Biol 153:185–
nisms of muscle stem cell expansion with cytokines. Stem Cells 193.
20:50 – 60. Friedman B, Hockfield S, Black JA, Woodruff KA, Waxman SG. 1989.
Debus E, Weber K, Osborn M. 1983a. Monoclonal antibodies specific In situ demonstration of mature oligodendrocytes and their
for glial fibrillary acidic (GFA) protein and for each of the neuro- processes: an immunocytochemical study with a new monoclonal
filament triplet polypeptides. Differentiation 25:193–203. antibody, rip. Glia 2:380 –390.
15524892, 2004, 1, Downloaded from https://anatomypubs.onlinelibrary.wiley.com/doi/10.1002/ar.a.10134 by Cochrane Romania, Wiley Online Library on [31/10/2022]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
ADULT STEM CELLS 99
Gage FH, Coates PW, Palmer TD, Kuhn HG, Fisher LJ, Suhonen JO, the preplate technique: potential for rapid isolation of muscle-de-
Peterson DA, Suhr ST, Ray J. 1995. Survival and differentiation of rived stem cells. Hum Gene Ther 12:619 – 628.
adult neuronal progenitor cells transplanted to the adult brain. Javazon EH, Colter DC, Schwarz EJ, Prockop DJ. 2001. Rat marrow
Proc Natl Acad Sci USA 92:11879 –11883. stromal cells are more sensitive to plating density and expand more
Galli R, Borello U, Gritti A, Minasi MG, Bjornson C, Coletta M, Mora rapidly from single-cell-derived colonies than human marrow stro-
M, De Angelis MGC, Fiocco R, Cossu G, Vescovi AL. 2000. Skeletal mal cells. Stem Cells 19:219 –225.
myogenic potential of human and mouse neural stem cells. Nat Jiang Y, Vaessen B, Lenvik T, Blackstad M, Reyes M, Verfaillie CM.
Neurosci 3:986 –991. 2002. Multipotent progenitor cells can be isolated from bone mar-
Gordon GJ, Coleman WB, Hixson DC, Grisham JW. 2000. Liver row, muscle and brain. Exp Hematol 30:896 –904.
regeneration in rats with retrorsine-induced hepatocellular injury Johansson CB, Momma S, Clarke DL, Risling M, Lendahl U, Frisen J.
proceeds through a novel cellular response. Am J Pathol 156:607– 1999. Identification of a neural stem cell in the adult mammalian
619. central nervous system. Cell 96:25–34.
Gorski JP, Griffin D, Dudley G, Stanford C, Thomas R, Huang C, Lai Johe KK. 2003. Development of neural stem cells for treatment of
E, Karr B, Solursh M. 1990. Bone acidic glycoprotein-75 is a major neurological diseases. Arch Neurol 60:297.
synthetic product of osteoblastic cells and localized as 75- and/or Joshi HC, Cleveland DW. 1990. Diversity among tubulin subunits:
50-kDa forms in mineralized phases of bone and growth plate and
toward what functional end? Cell Motil Cytoskel 16:159.
in serum. J Biol Chem 265:14956 –14963.
Kacsoh B. 2000. Endocrine physiology. New York: McGraw-Hill.
Grande DA, Southerland SS, Manji R, Pate DW, Schwartz RE, Lucas
Kasugai S, Nagata T, Sodek J. 1992. Temporal studies on the tissue
PA. 1995. Repair of articular cartilage defect using mesenchymal
compartmentalization of bone sialoprotein (BSP), osteopontin
stem cells. Tissue Eng 1:345–353.
(OPN), and SPARC protein during bone formation in vitro. J Cell
Gritti A, Parati EA, Cova L, Frolichsthal P, Galli R, Wanke E, Fara-
Physiol 152:467– 477.
velli L, Morassutti DJ, Roisen F, Nickel DD, Vescovi AL. 1996.
Multipotential stem cells from the adult mouse brain proliferate Kishimoto T, Kikutani H, Borne AEGKrvd, Goyert SM, Mason D,
and self-renew in response to basic fibroblast growth factor. J Neu- Miyasaka M, Moretta L, Okumura K, Shaw S, Springer T, Sug-
rosci 16:1091–1100. amura K, Zola H. 1997. Leucocyte typing VI, white cell differenti-
Gritti A, Frolichsthal-Schoeller P, Galli R, Parati EA, Cova L, Pagano ation antigens. Hamden, CT: Garland Publishing.
SF, Bjornson CR, Vescovia AL. 1999. Epidermal and fibroblast Kopen GC, Prockop DJ, Phinney DG. 1999. Marrow stromal cells
growth factors behave as mitogenic regulators for a single multipo- migrate throughout forebrain and cerebellum, and they differenti-
tent stem cell-like population from the subventricular region of the ate into astrocytes after injection into neonatal mouse brains. Proc
adult mouse forebrain. J Neurosci 19:3287–3297. Natl Acad Sci USA 96:10711–10716.
Grounds MD, Garrett KL, Lai MC, Wright WE, Beilharz MW. 1992. Krause DS, Theise ND, Collector Mi, Henegariu O, Hwang S, Gardner
Identification of muscle precursor cells in vivo by use of MyoD1 and R, Neutzel S, Sharkis SJ. 2001. Multi-organ, multi-lineage engraft-
myogenin probes. Cell Tissue Res 267:99 –104. ment by a single bone marrow derived stem cell. Cell 105:369 –377.
Gui L, Chang A, Solovey M, Brown PV, Keller G, Hebbel RP. 1992. Krebsbach PH, Robey PG. 2002. Dental and skeletal stem cells: po-
P1H12 as an embryonic endothelial-endothelial adhesion molecule tential cellular therapeutics for craniofacial regeneration. J Dental
and possibly hemangioblast marker. Blood 92(Suppl 1):697. Educ 66:766 –773.
Gussoni E, Soneoka Y, Strickland CD, Buzney EA, Khan MK, Flint Kruger GM, Mosher JT, Bixby S, Joseph N, Iwashita T, Morrison SJ.
AF, Kunkel LM, Mulligan RC. 1999. Dystrophin expression in the 2002. Neural crest stem cells persist in the adult gut but undergo
mdx mouse restored by stem cell transplantation. Nature 401:390 – changes in self-renewal, neuronal subtype potential, and factor
394. responsiveness. Neuron 35:657– 669.
Hayflick L. 1965. The limited in vitro lifetime of human diploid cell Kumagai J, Sarkar K, Uhthoff HK, Okawara Y, Ooshima A. 1994.
strains. Exp Cell Res 37:614 – 636. Immunohistochemical distribution of type I, II and III collagens in
Hixson DC, Ponce MDL, Allison JP, Walborg Jr EF. 1984. Cell surface the rabbit supraspinatus tendon insertion. J Anat 185:279 –284.
expression by adult rat hepatocytes of a non-collagen glycoprotein Kuznetsov SA, Mankani MH, Gronthos S, Satomura K, Bianco P,
present in rat liver biomatrix. Exp Cell Res 152:402– 414. Robey PG. 2001. Circulating skeletal stem cells. J Cell Biol 153:
Hixson DC, Faris RA, Thompson NL. 1990. An antigenic portrait of 1133–1139.
the liver during carcinogenesis. Pathobiology 58:65–77. LaBarge MA, Blau HM. 2002. Biological progression from adult bone
Hixson DC, Brown J, McBride A, Affigne S. 2000. Differentiation marrow to mononucleated muscle stem cell to multinucleate muscle
status of rat ductal cells and ethionine-induced hepatic carcinomas fiber in response to injury. Cell 111:589 – 601.
defined with surface-reactive monoclonal antibodies. Exp Mol Lagasse E, Connors H, Al-Dhalimy M, Reitsma M, Dohse M, Osborne
Pathol 68:152–169.
L, Wang X, Finegold M, Weisman IL, Grompe M. 2000. Purified
Hockfield S, McKay RD. 1985. Identification of major cell classes in
hematopoietic stem cells can differentiate into hepatocytes in vivo.
the developing mammalian nervous system. J Neurosci 5:3310 –
Nat Med 6:1229 –1234.
3328.
Lannagi R, Cochran NA, Ishigami F, Hakomori S-I, Andrews PW,
Holmdahl R, Rubin K, Klareskog L, Larsson E, Wigzell H. 1986.
Knowles BB, Solter D. 1983. Stage specific embryonic antigens
Characterization of the antibody response in mice with type II
(SSEA-3 and -4) are epitopes of a unique globo-series ganglioside
collagen-induced arthritis, using monoclonal anti-type II collagen
antibodies. Arthritis Rheum 29:400 – 410. isolated from human treatocarcinoma cells. EMBO J 2:2355–2361.
Hubbard AL, Bartles JR, Braiterman LT. 1985. Identification of rat Lazard D, Sastre X, Frid MG, Glukhova MA, Thiery JP, Koteliansky
hepatocyte plasma membrane proteins using monoclonal antibod- VE. 1993. Expression of smooth muscle-specific proteins in myoepi-
ies. J Cell Biol 100:1115–1125. thelium and stromal myofibroblasts of normal and malignant hu-
Jackson KA, Mi T, Goodell MA. 1999. Hematopoietic potential of stem man breast tissue. Proc Natl Acad Sci USA 90:999 –1003.
cells isolated from murine skeletal muscle. Proc Natl Acad Sci USA Lee JY, Qu-Petersen Z, Cao B, Kimura S, Jankowski R, Cummins J,
96:14482–14486. Usas A, Gates C, Robbins P, Wernig A, Huard J. 2000. Clonal
Jackson KA, Majka SM, Wang H, Pocius J, Hartley CJ, Majesky MW, isolation of muscle-derived cells capable of enhancing muscle regen-
Entman ML, Michael LH, Hirschi KK, Goodell MA. 2001. Regener- eration and bone healing. J Cell Biol 150:1085–1099.
ation of ischemic cardiac muscle and vascular endothelium by adult Lie DC, Dziewczapolski G, Willhoite AR, Kaspar BK, Shults CW,
stem cells. J Clin Invest 107:1395–1402. Gage FH. 2002. The adult substantia nigra contains progenitor cells
Janes SM, Lowell S, Hutter C. 2002. Epidermal stem cells. J Pathol with neurogenic potential. J Neurosci 22:6639 – 6649.
197:479 – 491. Lucas PA, Syftestad GT, Caplan AI. 1986. Partial isolation and char-
Jankowski RJ, Haluszczak C, Trucco M, Huard J. 2001. Flow cyto- acterization of a chemotactic factor from adult bovine bone for
metric characterization of myogenic cell populations obtained via mesenchymal cells. Bone 7:365–371.
15524892, 2004, 1, Downloaded from https://anatomypubs.onlinelibrary.wiley.com/doi/10.1002/ar.a.10134 by Cochrane Romania, Wiley Online Library on [31/10/2022]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
100 YOUNG AND BLACK

Lucas PA, Caplan AI. 1988. Chemotactic response of embryonic limb Pate DW, Southerland SS, Grande DA, Young HE, Lucas PA. 1993.
bud mesenchymal cells and muscle-derived fibroblasts to trans- Isolation and differentiation of mesenchymal stem cells from rabbit
forming growth factor-beta. Connect Tissue Res 18:1–7. muscle. Surg Forum XLIV:587–589.
Lucas PA, Price PA, Caplan AI. 1988. Chemotactic response of mes- Petersen BE, Bowen WC, Patrene KD, Mars WM, Sullivan AK, Mu-
enchymal cells, fibroblasts and osteoblast-like cells to bone Gla rase N, Boggs SS, Greenberger JS, Goff JP. 1999. Bone marrow as
protein. Bone 5:19 –23. a potential source of hepatic oval cells. Science 284:1168 –1170.
Lucas PA. 1989. Chemotactic response of osteoblast-like cells to trans- Pittenger MF, Mackay AM, Beck SC, Jaiswal RK, Douglas R, Mosca
forming growth factor-beta. Bone 10:459 – 463. JD, Moorman MA, Simonetti DW, Craig S, Marshak DR. 1999.
Lucas PA, Calcutt AF, Mulvaney DJ, Young HE, Southerland SS. Multilineage potential of adult human mesenchymal stem cells.
1992. Isolation of putative mesenchymal stem cells from rat embry- Science 148:143–147.
onic and adult skeletal muscle. In Vitro Cell Dev Biol 28:154A. Poulsom R, Alison MR, Forbes SJ, Wright NA. 2002. Adult stem cell
Lucas PA, Calcutt AF, Ossi P, Young HE, Southerland SS. 1993. plasticity. J Pathol 197:441– 456.
Mesenchymal stem cells from granulation tissue. J Cell Biochem Prockop DJ. 1997. Marrow stromal cells as stem cells for nonhema-
17E:122. topoietic tissues. Science 276:71–74.
Lucas PA, Young HE, Laurencin CT. 1994a. Muscle morphogenetic Quirici N, Soligo D, Bossolasco P, Servida F, Sumini C, Deliliers GL.
2002. Isolation of bone marrow mesenchymal stem cells by anti-
protein and use thereof. U.S. Patent 5,328,695.
nerve growth factor receptor antibodies. Exp Hematol 30:783–791.
Lucas PA, Calcutt AF, Ossi P, Young HE, Southerland SS. 1994b.
Ramiya VK, Maraist M, Arfos KE, Schatz DA, Peck AB, Cornelius JG.
Granulation tissue contains cells capable of differentiating into
2000. Reversal of insulin-dependent diabetes using islets generated
multiple mesenchymal phenotypes. J Cell Biochem 18C:276.
in vitro from pancreatic stem cells. Nat Med 6:278 –282.
Lucas PA, Calcutt AF, Southerland SS, Wilson JA, Harvey RL, Ware-
Ratajczak MZ, Pletcher CH, Marlicz W, Machlinski B, Moore J, Wasik
jcka DJ, Young HE. 1995. A population of cells resident within
M, Ratajczak J, Gewirtz AM. 1998. CD34⫹, kit⫹, rhodamine 123
embryonic and newborn rat skeletal muscle is capable of differen- (low) phenotype identifies a marrow cell population highly enriched
tiating into multiple mesodermal phenotypes. Wound Rep Reg for human hematopoietic stem cells. Leukemia 12:942–950.
3:457– 468. Reyes M, Verfaillie CM. 2001. Characterization of multipotent adult
Majka SM, Jackson KA, Kienstra KA, Majesky MW, Goodell MA, progenitor cells, a subpopulation of mesenchymal stem cells. Ann
Hirschi KK. 2003. Distinct progenitor populations in skeletal mus- NY Acad Sci 938:231–233; discussion, 233–235.
cle are bone marrow derived and exhibit different cell fates during Reyes M, Lund T, Lenvik T, Aguiar D, Koodle L, Verfaillie CM. 2001.
vascular regeneration. J Clin Invest 111:71–79. Purification and ex vivo expansion of postnatal human marrow
Mauro A. 1961. Satellite cell of skeletal muscle fibers. J Biophys mesodermal progenitor cells. Blood 98:2615–2625.
Biochem Cytol 9:493– 498. Reynolds R, Carey EM, Herschkowitz N. 1989. Immunohistochemical
McGuire WP. 1998. High-dose chemotherapy and autologous bone localization of myelin basic protein and 2⬘,3⬘-cyclicnucleotide 3⬘-
marrow or stem cell reconstitution for solid tumors. Curr Probl phosphohydrolase in flattened membrane expansions produced by
Cancer 22:135–137. cultured oligodendrocytes. Neuroscience 28:181–188.
McKinney-Freeman SL, Jackson KA, Camargo FD, Ferrari G, Mavil- Rogers JJ, Young HE, Adkison LR, Lucas PA, Black Jr AC. 1995.
lio F, Goodell MA. 2002. Muscle-derived hematopoietic stem cells Differentiation factors induce expression of muscle, fat, cartilage,
are hematopoietic in origin. Proc Natl Acad Sci USA 99:1341–1346. and bone in a clone of mouse pluripotent mesenchymal stem cells.
Mezey E, Chandross KJ, Harta G, Maki RA, McKercher SR. 2000. Am Surg 61:231–236.
Turning blood into brain: cells bearing neuronal antigens generated Romero-Ramos M, Vourc’h P, Young HE, Lucas PA, Wu Y, Chi-
in vivo from bone marrow. Science 290:1779 –1782. vatakarn O, Zaman R, Dunkelman N, El-Kalay MA, Chesselet M-F.
Moore KL, Persaud TVN. 1998. The developing human: clinically 2002. Neuronal differentiation of stem cells isolated from adult
oriented embryology, 6th edition. Philadelphia: W.B. Saunders Co. muscle. J Neurosci Res 69:894 –907.
Morhenn VB. 2002. In: Developmental studies hybridoma bank prod- Ronnov-Jessen L, Celis JE, Van Deurs B, Petersen OW. 1992. A
uct catalog, year 2002. Iowa City, IA: University of Iowa. p 24. fibroblast-associated antigen: characterization in fibroblasts and
Mujoo K, Ali M, Sahib MK. 1983. Isolation, characterization, and immunoreactivity in smooth muscle differentiated stromal cells.
synthesis of alpha-fetoprotein from neonatal rat brain. J Neuro- J Histochem Cytochem 40:475– 486.
chem 41:1223–1228. Saito T, Dennis JE, Lennon DP, Young RG, Caplan AI. 1995. Myo-
Muraglia A, Cancedda R, Quarto R. 2000. Clonal mesenchymal pro- genic expression of mesenchymal stem cells within myotubes of
genitors from bone marrow differentiate in vitro according to a mdx mice in vitro and in vivo. Tissue Eng 1:327–343.
hierarchial model. J Cell Sci 113:1161–1166. Sanchez-Ramos J, Song S, Cardozo-Pelaez F, Hazzi C, Stedeford T,
Willing A, Freeman TB, Saporta S, Janssen W, Patel N, Cooper DR,
Murayama A, Matsuzaki Y, Kawaguchi A, Shimazaki T, Okano H.
Sanberg PR. 2000. Adult bone marrow stromal cells differentiate
2002. Flow cytometric analysis of neural stem cells in the develop-
into neural cells in vitro. Exp Neurol 164:247–256.
ing and adult mouse brain. J Neurosci Res 69:837– 847.
Schaart G, Moens L, Endert JM, Ramaekers FCS. 1997. Biochemical
Naumann K, Pette D. 1994. Effects of chronic stimulation with dif-
characterization of cardiotin, a sarcoplasmic reticulum associated
ferent impulse patterns on the expression of myosin isoforms in rat
protein. FEBS Lett 403:168 –172.
myotube cultures. Differentiation 55:203–211.
Shafiq SA, Shimizu T, Fischman DA. 1984. Heterogeneity of type I
Oben JA, Yang S, Lin H, Ono M, Diehl AM. 2003. Acetylcholine skeletal muscle fibers revealed by monoclonal antibody to slow
promotes the proliferation and collagen gene expression of myofi- myosin. Muscle Nerve 7:380 –387.
broblastic hepatic stellate cells. Biochem Biophys Res Commun Shoptaw JH, Bowerman S, Young HE, Lucas PA. 1991. Use of gelfoam
300:172–177. as a substrate for osteogenic cells of marrow. Surg Forum XLII:537–
Otani A, Kinder K, Ewalt K, Otero FJ, Schimmel P, Friedlander M. 538.
2002. Bone marrow-derived stem cells target retinal astrocytes and Simper D, Stalboerger PG, Panetta CJ, Wang S, Caplice NM. 2002.
can promote or inhibit retinal angiogenesis. Nat Med 8:1004 –1010. Smooth muscle progenitor cells in human blood. Circulation 106:
Owen M. 1988. Marrow stromal cells. J Cell Sci Suppl 10:63–76. 1199 –1204.
Owen M, Friedenstein AJ. 1988. Stromal stem cells: marrow-derived Skalli O, Ropraz P, Trzeciak A, Benzonana G, Gillessen D, Gabbiani
osteogenic precursors. Ciba Found Symp 136:42– 60. G A. 1986. A monoclonal antibody against alpha-smooth muscle
Palis J, Segel GB. 1998. Developmental biology of erythropoiesis. actin: a new probe for smooth muscle differentiation. J Cell Biol
Blood Rev 12:1061–1064. 103:2787–2796.
Palmer TD, Schwartz PH, Taupin P, Kaspar B, Stein SA, Gage FH. Solovey A, Lin Y, Browne P, Choong S, Wayner E, Hebbel RP. 1997.
2001. Progenitor cells from human brain after death. Nature 411: Circulating activated endothelial cells in sickle cell anemia. N Eng
42– 43. J Med 337:1584 –1590.
15524892, 2004, 1, Downloaded from https://anatomypubs.onlinelibrary.wiley.com/doi/10.1002/ar.a.10134 by Cochrane Romania, Wiley Online Library on [31/10/2022]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
ADULT STEM CELLS 101
Solter D, Knowles BB, 1978. Monoclonal antibody defining a stage- Woodbury D, Schwarz EJ, Prockop DJ, Black IB. 2000. Adult rat and
specific mouse embryonic antigen (SSEA-1). Proc Natl Acad Sci human bone marrow stromal cells differentiate into neurons.
USA 75:5565–5569. J Neurosci Res 61:364 –370.
Sprinkle TJ, Agee JF, Tippins RB, Chamberlain CR, Faguet GB, Woodbury D, Reynolds K, Black IB. 2002. Adult bone marrow stromal
DeVries GH. 1987. Monoclonal antibody production to human and stem cells express germline, ectodermal, endodermal, and mesoder-
bovine 2⬘,3⬘-cyclic nucleotide 3⬘-phosphodiesterase (CNPase): high- mal genes prior to neurogenesis. J Neurosci Res 96:908 –917.
specificity recognition in whole brain acetone powders and conser- Wright WE, Binder M, Funk W. 1991. Cyclic amplification and selec-
vation of sequence between CNP1 and CNP2. Brain Res 426:349 – tion of targets (CASTing) for the myogenin consensus binding site.
357. Mol Cell Biol 11:4104 – 4110.
Sprinkle T. 1989. 2⬘,3⬘-cyclic nucleotide 3⬘-phosphodiesterase, an oli- Yang L, Shiwu L, Hatch H, Ahrens K, Cornelius JG, Petersen BE,
godendrocyte-Schwann cell and myelin-associated enzyme of the Peck AB. 2002. In vitro trans-differentation of adult hepatic stem
nervous system. CRC Crit Rev Neurobiol 4:235–301. cells into pancreatic endocrine hormone-producing cells. Proc Natl
St. Croix B, Rago C, Velculescu V, Traverso G, Romans KE, Mont- Acad Sci USA 99:8078 – 8083.
gomery E, Lal A, Riggins GJ, Lengauer C, Vogelstein B, Kinzler Ye XJ, Terato K, Nakatani H, Cremer MA, Yoo TJ. 1991. Monoclonal
KW. 2000. Genes expressed in human tumor endothelium. Science antibodies against bovine type IX collagen (LMW fragment): pro-
289:1197–1202. duction, characterization, and use for immunohistochemical local-
Tanguchi H, Toyoshima T, Fukao K, Nakauchi H. 1996. Presence of ization studies. J Histochem Cytochem 39:265–271.
hematopoietic stem cells in the adult liver. Nat Med 2:198 –203. Young HE. 1977a. Epidermal ridge formation during limb regenera-
Taylor DA, Atkins BZ, Hungspreugs P, Jones TR, Reedy MC, Hutch- tion in the adult salamander, Ambystoma annulatum. Proc Ark
enson KA, Glower DD, Kraus WE. 1997. Delivery of primary autol- Acad Sci 31:107–109.
ogous skeletal myoblasts into rabbit heart by coronary infusion: a Young HE. 1977b. Limb regeneration in the adult salamander, Am-
potential approach to myocardial repair. Proc Assoc Am Physicians bystoma annulatum Cope 1889 (Amphibia: Ambystomatidae). Fay-
109:245–253. etteville, AR: University of Arkansas Library Press.
Thulasi R, Dias P, Houghton PJ, Houghton JA. 1996. Alpha 2a- Young HE. 1977c. Anomalies during limb regeneration in the adult
interferon-induced differentiation of human alveolar rhabdomyo- salamander, Ambystoma annulatum. Proc Ark Acad Sci 31:110 –
sarcoma cells: correlation with down-regulation of the insulin-like 111.
growth factor type I receptor. Cell Growth Differ 7:531–541. Young HE. 1983. A temporal examination of glycoconjugates during
Tole S, Kaprielian Z, Ou SK, Patterson PH. 1995. FORSE-1: a posi- the initiation phase of limb regeneration in adult Ambystoma.
tionally regulated epitope in the developing rat central nervous Lubbock, TX: Texas Tech University Library Press.
system. J Neurosci 15:957–969. Young HE, Bailey CF. Dalley BK. 1983a. Environmental conditions
Tole S, Patterson PH. 1995. Regionalization of the developing prerequisite for complete limb regeneration in the postmetamorphic
forebrain: a comparison of FORSE-1, Dlx-2, and BF-1. J Neurosci adult land-phase salamander, Ambystoma. Anat Rec 206:289 –294.
15:970 –980. Young HE, Bailey CF, Dalley BK. 1983b. Gross morphological anal-
Toma JG, Akhavan M, Fernandes KJ, Barnabe-Heider F, Sadikot A, ysis of limb regeneration in postmetamorphic adult Ambystoma.
Kaplan DR, Miller FD. 2001. Isolation of multipotent stem cells Anat Rec 206:295–306.
from the dermis of mammalian skin. Nat Cell Biol 3:778 –784. Young HE, Dalley BK, Markwald RR. 1983c. Identification of hyal-
Tomita S, Li RK, Weisel RD, Mickle DA, Kim EJ, Sakai T, Jia ZQ. uronate within peripheral nervous tissue matrices during limb re-
1999. Autologous transplantation of bone marrow cells improves generation. In: Coates PW, Markwald RR, Kenny AD, editors. De-
damaged heart function. Circulation 100(Suppl II):11247–11256. veloping and regenerating vertebrate nervous systems, neurology
Tsai RY, Kittappa R, McKay RGD. 2002. Plasticity, niches, and the and neurobiology. New York: Alan R. Liss, Inc. 6, p 175–183.
use of stem cells. Dev Cell 2:707–712. Young HE, Dalley BK, Markwald RR. 1983d. The interaction of gly-
Turgeman G, Pittman DD, Muller R, Kurkalli BG, Zhou S, Pelled G, cosaminoglycans (GAG) and nervous tissue regeneration in adult
Peyser A, Zilberman Y, Moutsatsos IK, Gazit D. 2001. Engineered Ambystoma. Anat Rec 205:202.
human mesenchymal stem cells: a novel platform for skeletal cell Young HE, Bailey CF, Markwald RR, Dalley BK. 1985. Histological
mediated gene therapy. J Gene Med 3:240 –251. analysis of limb regeneration in postmetamorphic adult Ambys-
Vacanti MP, Leonard JL, Dore B, Bonassar LJ, Cao Y, Stachelek SJ, toma. Anat Rec 212:183–194.
Vacaqnti JP, O’Connell F, Yu CS, Farwell AP, Vacanti CA. 2001a. Young HE, Dalley BK, Markwald RR. 1989a. Glycoconjugates in
Tissue-engineered spinal cord. Transplant Proc 33:592–598. normal wound tissue matrices during the initiation phase of limb
Vacanti MP, Roy A, Cortiella J, Bonassar L, Vacanti CA. 2001b. regeneration in adult Ambystoma. Anat Rec 223:231–241.
Identification and initial characterization of spore-like cells in adult Young HE, Dalley BK, Markwald RR. 1989b. Effect of selected den-
mammals. J Cell Biochem 80:455– 460. ervations on glycoconjugate composition and tissue morphology
Vescovi AL, Galli R, Gritti A. 2001. The neural stem cells and their during the initiation phase of limb regeneration in adult Ambys-
transdifferentiation capacity. Biomed Pharmacother 55:201–205. toma. Anat Rec 223:231–241.
Vierck JL, McNamara JP, Dodson MV. 1996. Proliferation and differ- Young HE, Carrino DA, Caplan AI. 1989c. Histochemical analysis of
entiation of progeny of ovine unilocular fat cells (adipofibroblasts). newly synthesized and resident sulfated glycosaminoglycans during
In Vitro Cell Dev Biol Anim 32:564 –572. musculogenesis in the embryonic chick leg. J Morph 201:85–103.
Walborg Jr EF, Tsuchida S, Weeden DS, Thomas MW, Barrick A, Young HE, Morrison DC, Martin JD, Lucas PA. 1991. Cryopreserva-
McEntire KD, Allison JP, Hixson DC. 1985. Identification of dipep- tion of embryonic chick myogenic lineage-committed stem cells. J
tidyl peptidase IV as a protein shared by the plasma membrane of Tissue Cult Methods 13:275–284.
hepatocytes and liver biomatrix. Exp Cell Res 158:509 –518. Young HE, Ceballos EM, Smith JC, Lucas PA, Morrison DC. 1992a.
Warejcka DJ, Harvey R, Taylor BJ, Young HE, Lucas PA. 1996. A Isolation of embryonic chick myosatellite and pluripotent mesen-
population of cells isolated from rat heart capable of differentiating chymal stem cells. J Tissue Cult Methods 14:85–92.
into several mesodermal phenotypes. J Surg Res 62:233–242. Young HE, Sippel J, Putnam LS, Lucas PA, Morrison DC. 1992b.
Webster C, Silberstein L, Hays AP, Blau HM. 1988. Fast muscle fibers Enzyme-linked immuno-culture assay. J Tissue Cult Methods 14:
are preferentially affected in Duchenne muscular dystrophy. Cell 31–36.
52:503–513. Young HE, Ceballos EM, Smith JC, Mancini ML, Wright RP, Ragan
Winter A, Breit S, Parsch D, Benz K, Steck E, Hauner H, Weber RM, BL, Bushell I, Lucas PA. 1993. Pluripotent mesenchymal stem cells
Ewerbeck V, Richter W. 2003. Cartilage-like gene expression in reside within avian connective tissue matrices. In Vitro Cell Dev
differentiated human stem cell spheroids. Arthritis Rheum 48:418 – Biol Anim 29A:723–736.
429. Young HE, Mancini ML, Wright RP, Smith JC, Black Jr AC, Reagan
Wood JN, Anderton BH. 1981. Monoclonal antibodies to mammalian CR, Lucas PA. 1995. Mesenchymal stem cells reside within the
neurofilaments. Biosci Rep 1:263–268. connective tissues of many organs. Dev Dyn 202:137–144.
15524892, 2004, 1, Downloaded from https://anatomypubs.onlinelibrary.wiley.com/doi/10.1002/ar.a.10134 by Cochrane Romania, Wiley Online Library on [31/10/2022]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
102 YOUNG AND BLACK

Young HE, Lucas PA. 1998. Pluripotent mesenchymal stem cells and Ninomiya S, Detmer K, Bray RA, Steele TA, Hixson D, El-Kalay M,
methods of use thereof. U.S. Patent 5,827,735. Tobin BW, Russ RD, Horst MN, Floyd JA, Henson NL, Hawkins
Young HE, Wright RP, Mancini ML, Lucas PA, Reagan CR. Black Jr KC, Groom J, Parikh A, Blake L, Bland LJ, Thompson AJ, Kirincich
AC. 1998a. Bioactive factors affect proliferation and phenotypic A, Moreau C, Hudson J, Bowyer III FP, Lin TJ, Black Jr AC. 2004a.
expression in pluripotent and progenitor mesenchymal stem cells. Adult reserve stem cells and their potential for tissue engineering.
Wound Rep Reg 6:65–75. J Cell Biochem Biophys 40(1):(in press).
Young HE, Rogers JJ, Adkison LR, Lucas PA, Black Jr AC. 1998b. Young HE, Duplaa C, Yost MJ, Henson NL, Floyd JA, Detmer K,
Muscle morphogenetic protein induces myogenic gene expression in Thompson AJ, Powell SW, Gamblin TC, Kizziah K, Holland BJ,
Swiss-3T3 cells. Wound Rep Reg 6:530 –541. Boev A, Van de Water JM, Godbee DC, Jackson S, Rimando M,
Young HE, Steele TA, Bray RA, Detmer K, Blake LW, Lucas PA, Edwards CR, Wu E, Cawley C, Edwards PD, Macgregor A, Bozof R,
Black Jr AC. 1999. Human pluripotent and progenitor cells display Thompson TM, Petro Jr GJ, Shelton HM, McCampbell BL, Mills
cell surface cluster differentiation markers CD10, CD13, CD56, and JC, Flynt FL, Steele TA, Kearney M, Kirincich-Greathead A, Hardy
MHC Class-I. Proc Soc Exp Biol Med 221:63–71. W, Young PR, Amin AV, Williams RS, Horton MM, McGuinn S,
Young HE. 2000. Stem cells and tissue engineering. In: Huard J, Fu
Hawkins KC, Davis JG, Parikh A, Ericson K, Terracio L, Moreau C,
FH, editors. Gene Therapy in orthopaedic and sports medicine. New
Hixson D, Tobin BW, Hudson J, Bowyer III FP, Black Jr AC. 2004b.
York: Springer-Verlag. p 143–173.
Clonogenic analysis reveals reserve stem cells in postnatal mam-
Young HE, Duplaa C, Young TM, Floyd JA, Reeves ML, Davis KH,
mals. II. Pluripotent epiblastic-like stem cells. Anat Rec (in press).
Mancini GJ, Eaton ME, Hill JD, Thomas K, Austin T, Edwards C,
Zhao Y, Glesne D, Huberman E. 2003. A human peripheral blood
Cuzzourt J, Parikh A, Groom J, Hudson J, Black Jr AC. 2001a.
Clonogenic analysis reveals reserve stem cells in postnatal mam- monocyte-derived subset acts as pluripotent stem cells. Proc Natl
mals. I. Pluripotent mesenchymal stem cells. Anat Rec 263:350 – Acad Sci USA 100:2426 –2431.
360. Zuk PA, Zhu M, Mizuno H, Huang J, Futrell JW, Katz AJ, Benhaim
Young HE, Steele T, Bray RA, Hudson J, Floyd JA, Hawkins KC, P, Lorenz P, Hedrick MH. 2001. Multilineage cells from human
Thomas K, Austin T, Edwards C, Cuzzourt J, Duenzl M, Lucas PA, adipose tissue: implications for cell-based therapies. Tissue Eng
Black Jr AC. 2001b. Human reserve pluripotent mesenchymal stem 7:211–228.
cells are present in the connective tissues of skeletal muscle and Zulewski H, Abraham EJ, Gerlach MJ, Daniel PB, Moritz W, Muller
dermis derived from fetal, adult, and geriatric donors. Anat Rec B, Vallejo M, Thomas MK, Habener JF. 2001. Multipotential nes-
264:51– 62. tin-positive stem cells isolated from adult pancreatic islets differ-
Young HE. 2004. Existence of reserve quiescent stem cells in adults, entiate ex vivo into pancreatic endocrine, exocrine, and hepatic
from amphibians to humans. Curr Top Microbiol Immunol 280:71– phenotypes. Diabetes 50:521–533.
109. Zvaifler NJ, Marinova-Mutafchieva L, Adams G, Edwards CJ, Moss J,
Young HE, Duplaa C, Romero-Ramos M, Chesselet M-F, Vourc’h P, Burger JA, Maini RN. 2000. Mesenchymal precursor cells in the
Yost MJ, Ericson K, Terracio L, Asahara T, Masuda H, Tamura- blood of normal individuals. Arthritis Res 2:477– 488.

You might also like