Title of Invention

”AN IN-VITRO METHOD FOR ISOLATING STEM/PROGENITOR CELLS FROM THE AMNIOTIC MEMBRANE OF UMBILICAL CORD”.

Abstract The present invention relates to a method for isolating stem/progenitor cells from the amniotic membrane of umbilical cord, wherein the method comprises separating the amniotic membrane from the other components of the umbilical cord in vitro, culturing the amniotic membrane tissue under conditions allowing cell proliferation, and isolating the stem/progenitor cells from the tissue cultures. The isolated stem cells can have embryonic stem cell-like properties and can be used for various therapeutic purposes. In one embodiment, the invention relates to the isolation and cultivation of stem cells having embryonic properties such as epithelial and/or mesenchymal stem/progenitor cells under conditions allowing the cells to undergo mitotic expansion. Furthermore, the invention is directed to a method for the differentiation of the isolated stem/progenitor cells into epithelial and/or mesenchymal cells.
Full Text EOLATION OF STEM/PROGENITOR CELLS FROM AMNIOTIC MEMBRANE OF UMBILICAL
CORD
[0001] The present invention relates to a method for isolating
stem/progenitor cells from the amniotic membrane of umbilical cord, wherein
the method comprises separating the amniotic membrane from the other
components of the umbilical cord in vitro, culturing the amniotic membrane
tissue under conditions allowing cell proliferation, and isolating the
stem/progenitor cells from the tissue cultures. In particular, the invention relates
to the isolation and cultivation of stem cells having embryonic properties such
as epithelial and/or mesenchymal stem/progenitor cells under conditions
allowing the cells to undergo mitotic expansion. Furthermore, the invention is
directed to a method for the differentiation of the isolated stem/progenitor cells
into epithelial and/or mesenchymal cells and therapeutic uses of these
stem/progenitor cells:
BACKGROUND OF THE INVENTION
[0002] Stem cells are a cell population possessing the capacities to selfrenew
indefinitely and to differentiate in multiple cell or tissue types. Embryonic
stem cells (from approximately days 3 to 5. after1 fertilisation) proliferate
indefinitely and can differentiate spontaneously into all tissue types: they are
"thus termed plurfpotent stem cells (reviewed, for example, in Smith, A.G. (2001)
Annu. Rev. Cell. Dev. BioL 17, 435-462). Adult stem cells, however, are more
tissue-specific and may have less replicative capacity: they are thus termed
muttipotent stem cells (reviewed, for example, in Paul, G. et al. (2002) Drug
Dlscov. Today 7, 295-302). The "plasticity" of embryonic and adult stem cells
relies on their ability to trans-differentiate into tissues different from their origin
and, perhaps, across embryonic germ layers.
[0003] The ability of stem cells to self-renew is critical to their function as
reservoir of primitive undifferentiated cells. In contrast, most somatic cells have
a limited capacity for self-renewal due to telomere shortening (reviewed, for
example, in Dice, J.F. (1993) Physiol. Rev. 73, 149-159). Stem cell-based
therapies thus have the potentfal to be useful for the treatment of a multitude of
human and animal diseases.
[0004] Stem cells as well as stem/progenitor cells can be derived from
different sources. The "multi-lineage" potential of embryonic and adult stem
cells has been extensively characterized. Even though the potential of
embryonic stem cells is enormous, their use implies many ethical problems.
Therefore, non-embryonic stem cells derived from the bone marrow stroma, fat
tissue, dermis and umbilical cord blood have been proposed as alternative
sources. These cells can differentiate inter alia into chondrocytes, adipocytes,
osteoblasts, myoblasts, cardiomyocytes, astrocytes, and tenocytes in vitro and
undergo differentiation in vivo, making these stem cells - in general referred to
as mesenchymai stem cells - promising candidates for mesodermal defect
repair and disease management.
[0005] In clinical use, however, harvesting of such mesenchymai stem
cells causes several problems. The collection of the cells is a mental and
physical burden to the patient as a surgical procedure is required to obtain the
cells (for example, the collection of bone marrow is an invasive technique
performed with a biopsy needle that requires local or even general anesthesia).
Furthermore, in many cases the number of stem cells extracted is rather low.
More importantly, no epithelial cells are derived or differentiated from these
cells. This prompted the search for other possible sources of stem cells.
[0006] Umbilical cord blood has been identified as a rich source of
haematopoetic stem/progenitor cells. However, the existence of mesenchymai
stem/progenitor cells is discussed controversially. On the one hand, such cells
could not be isolated or successfully cultured from term umbilical cord blood
(Mareschl, K. et al. (2001) Haematologlca 86, 1099-1100). At the same time,
results obtained by Campagnoli, C. et al. (Blood (2001) 98, 2396-2402) as well
as Erices, A. et al. (Br. J. Haematol. (2000) 109, 235-242) suggest that
mesenchymai stem cells are present in several fetal organs and circulate in the
blood of pre-term fetuses simultaneously with hematopoietic precursors.
Accordingly, International Patent Application WO 03/070922 discloses isolation
and culture-expansion methods of mesenchymai stem/progenitor cells from
umbilical cord blood and a differentiation method of such cells into various
mesenchymai tissues. Isolation efficiencies of about 60% have been reported
(Bieback, K. et al. (2004) Stem Cells 22, 625-634). In the same study, both the
time period from collection of the umbilical cord blood to isolation of the cells
and the volume of the blood sample used have been determined as crucial
parameters for achieving such a yield. However, it is still a matter of debate
whether these stem/progenitor cells are indeed derived of umbilical cord tissue.
[0007] Recently, mesenchymal stem/progenitor cells have been
successfully isolated from umbilical cord tissue, namely from Wharton's jelly, the
matrix of umbilical cord, (Mitchell, K.E. et al. (2003) Stem Cells 21, 50-60; U.S.
Patent 5,919,702; US Patent Application 2004/0136967). These cells have
been shown to have the capacity to differentiate, for example, into a neuronal
phenotype and into cartilage tissue, respectively. Furthermore, mesenchymal
stem/progenitor cells have also been isolated from the endothelium and the
subendothelial layer of the umbilical cord vein, one of the three vessels (two
arteries, one vein) found within the umbilical cord (Romanov, Y.A. et al. (2003)
Stem Cells 21, 105-110; Covas, D.T. et al. (2003) Braz. J. Med. Biol. Res. 36,
1179-1183).
[0008] However, none of these approaches employed thus far has
resulted in the isolation or cultivation of epithelial stem/progenitor cells as a
source for epithelial cell-based therapies such as skin resurfacing, liver repair,
bladder tissue engineering and other engineered surface tissues. Thus, there is
still a need for methods and reliable sources useful for the isolation and
cultivation of epithelial stem/progenitor cells. Furthermore, rapid and efficient
methods which are ethically acceptable and do not pose a biomedical burden
on the patient for the isolation of epithelial and mesenchymal stem/progenitor
cells are still required in order to provide such cells in a sufficient amount for
various applications in regenerative medicine and tissue engineering.
SUMMARY OF THE INVENTION
[0009] The invention provides a method for isolating stem/progenitor
cells from the amniotic membrane of umbilical cord, the method comprising:
(a) separating the amniotjc membrane from the other components of
the umbilical cord In vitro;
(b) culturing the amniotic membrane tissue obtained in step (a) under
conditions allowing cell proliferation; and
(c) Isolating the stem/progenitor cells.
[0010] In one embodiment, the invention provides a method, further
comprising:
(a") separating the cells from the amniotic membrane tissue before
cultivation by a technique selected from the group consisting of enzymatic
digestion and direct tissue explant.
[0011] In one preferred embodiment, the invention provides a method for
isolating stem/progenitor cells that have embryonic stem cell-like properties.
[0012] In another preferred embodiment, the invention provides a method
for isolating epithelial and/or mesenchymal stem/progenitor cells.
[0013] In another embodiment, the invention provides a method further
comprising:
(d) culturing the stem/progenitor cells under conditions allowing the
cells to undergo clonal expansion.
[0014] In yet another embodiment, the invention provides a method
further comprising:
(e) culturing the stern/progenitor cells under conditions allowing the
differentiation of said cells into epithelial ceils and/or mesenchymal cells; and
(f) isolating the differentiated cells.
[0015] In yet another embodiment, the invention provides a method,
further comprising:
(g) preserving the isolated stem/progenitor cells for further use.
[0016] In yet a further embodiment, the invention comprising a method of
cultivating stem/progenitors cells of the invention, comprising:
Obtaining a tissue explant from the amniotic membrane of umbilical cord;
Cultivating the tissue explant in suitable cultivation media and cultivation
conditions over a suitable period of time.
In yet other embodiments, the invention is directed to therapeutic uses of the
stem/progenitor cells or cellular extracts thereof. One of these embodiments
provide a method of treating a subject having a disorder comprising
administering to the subject an effective amount of a stem/progenitor cell
isolated by the inventive method of explained above. Another embodiment
5
provides a corresponding pharmaceutical composition.
BRIEF DESCRIPTION OF THE DRAWINGS
[0017] The invention will be better understood with reference to the
detailed description when considered in conjunction with the non-limiting
examples and the drawings, in which:
[0018]Figure 1 depicts'epithelial cell outgrowth from umbilical cord
amniotic membrane by the method of direct tissue explant (40 x magnification)
at day 2 (Fig. 1A) and day 5 (Fig. 1B, C) of tissue culture. Cell culture plastic
surfaces were coated with collagen 1/collagen 4 mixtures (1:2; Becton
Dickinson) before placing the amniotic membrane on the surface. The amniotic
membrane specimens were submerged in 5 ml EpiLife medium or Medium
(both from Cascade Biologies). Medium was changed every 2 or 3 days and cell
outgrowth by explant was monitored under light microscopy. Microphotographs
were taken at different time intervals as stated above. The observed polyhedral
cell morphology is typical of epithelial cells.
[0019] Figure 2 depicts enzymatic digestion of the umbilical cord
segments yielding similar epithelial (40 x magnification) cells at day 2 (Fig. A,
C) and day 5 (Fig. B, D). Umbilical cord amniotic membrane was divided into
small pieces of 0.5 cm x 0.5 cm and digested in 0.1% (w/v) collagenase type
solution (Roche Diagnostics) at 37°C for 8 hours. The samples were vortexed
every 30 min for 3 min. Cells were harvested by centrifugation at 4000 rpm for
30 min. Cell pellets were resuspended in EpiLife medium or Medium 171 (both
from Cascade Biologies) supplemented with 50 ug/ml insulin-like growth factor-
1 (IGF-1), 50 jig/ml platelet-derived growth factor-BB (PDGF-BB), 5 ug/ml
transforming growth factor-£1 (TGF-£1) and 5 jag/ml insulin (all obtained from
R&D Systems), counted and seeded on 10 cm tissue culture dishes pre-coated
with collagen 1/coIlagen 4 mixtures (1:2; Becton Dickinson) at density of 1 x 106
cells/dish. After 24 hours, attached cells were washed with warm .phosphate
buffered saline (PBS) and the culture medium was replaced with EpiLife
medium or Medium 171 (both from Cascade Biologies). The medium was
changed every 2 OF 3 days, and cell outgrowth was monitored under light
microscopy. Microphotographs were taken at different time intervals as stated
6
above. Once again the cells demonstrated typical epithelial ceil polyhedral
morphology.
[0020] Figure 3 depicts outgrowing mesenchyma! cells explanted from
umbilical cord amniotic membrane. Cellular outgrowth was observed as early
as 48 hours after placement in tissue culture dishes using DMEM supplemented
with 10% fetal calf serum (PCS) as culture medium (40 x magnification) (Fig.
3A, C). The explants were submerged in 5 ml DMEM (Invitrogen) supplemented
with 10% fetal bovine serum (Hyclone) (DMEM/10% FBS). Medium was
changed every 2 or 3 days. Cell outgrowth was monitored under light
microscopy. Microphotographs were taken at different time intervals. The cells
were characterized by their spindle shaped morphology, and migrated and
expanded both easily and quickly in vitro, closely resembling fibroblasts (Fig.
3B, D).
[0021] Figure 4 (40 x magnification) depicts mesenchymal cells from
umbilica! cord amniotic membrane cells isolated by collagenase enzymatic
digestion. Fig. 4A shows mesenchymal cells isolated from umbilical cord
amniotic membrane at day 2. Cell proliferation was observed at day 5 (Fig. 4B).
Umbilical cord amniotic membrane was divided into small pieces of 0.5cm x
0.5cm and digested in 0.1% (w/v) collagenase typel solution (Roche
Diagnostics) at 37°C for 6 hours. The samples were vortexed every 15 min for 2
min. Cells were harvested by centrifugation at 4000 rpm for 30 min. Cell pellets
were resuspended in DMEM/10% FBS, counted and seeded on 10cm tissue
culture dish at density of 1 x 106 cells/dish. Medium was changed every 2 or 3
days. Cell outgrowing was monitored under light microscopy. Microphotographs
were taken at different time intervals. Once again, cells demonstrated spindle
shaped morphology typical of mesenchymal cells as fibroblasts.
[0022] Figure 5 (40 x magnification) depicts the morphology in serumfree
culture condition (DMEM) and serum culture condition (DMEM/10%FCS) of
umbilical cord amniotic membrane mesenchymal cells (UCMC, Fig. 5E, F, G, H)
isolated according to the method of the invention, normal dermal fibroblasts
(NF109 cells, Fig. 5A, B) and adipose-derived mesenchymal cells (ADMC, Fig.
5C, D). Fig. 5 shows changes in cell morphology of NF and ADMC cultured in
serum starvation conditions (DMEM only) reflected by flatter cells and less
dense cytoplasm as compared with serum rich conditions (DMEM/10%FCS)
where cells are more rounded with a dense cytoplasm (Fig. 5A, B, C, D). No
change in morphology was observed in both UCMC groups cultured under
identical conditions of serum-free vs. serum rich media (Fig. 5E, F, G, H),
indicating a difference in behavior and physiology of these latter mesenchymal
cells.
[0023] Figure 6 (40 x magnification) depicts UCMC isolated according to
the invention cultured in DMEM/10%FCS at days 3 and 7 without a 3T3 feeder
layer. The cells are seen to be growing well, and are forming a colony (vertical
growth) Instead of exhibiting radial spread. Once again, this indicates a
difference in behavior of these mesenchymal cells as compared to their more
differentiated counterparts.
[0024] Figure 7 (40 x magnification) depicts colony formation of
umbilical cord epithelial cells (UCEC) cultured on a 3T3 feeder layer at days 3
and 7. This appearance is similar to that of normal skin derived epithelial
keratinocyte stem cells. In the latter, the 3T3 feeder layer maintains sternness of
the cells.
[0025] Figure 8 (40 x magnification) depicts obvious colony formation of
umbilical cord mesenchymal cells (UCMC) isolated according to the invention
cultured on a 3T3 feeder layer at days 3 and 7. The 3T3 feeder layer normally
suppresses the growth of differentiated mesenchymal cells as human dermal
fibroblasts. Once again, this indicates a difference in behavior of these
mesenchymal cells as compared to their more differentiated counterparts.
[0026] Figure 9-1 to Figure 9-27 show Western blot analysis by which
the expression of several embryonic stem cell markers in UCEC and UCMC
isolated according to the invention, was compared to the expression of these
markers in human dermal fibroblasts (IMF), in bone marrow mesenchymal cells
(BMSC) and adipose-derived mesenchymal cells (ADMC). Fig. 9 also shows
highly secreted ActivinA and Follistatin detected by ELISA assay in
supematants of umbilical cord mesenchymal and epithelial stem cell culture in
comparison with bone marrow, adipose derived stem cells, human dermal
fibroblasts and epidermal keratinocytes
[0027] Figure 10 shows indirect immune-fluorescent analysis of markers
of epithelial cells expressed in umbilical cord epithelial stem cells such as
cytokeratins (CK)-general, CK17, CK6, CK10, CK19, CK18, CK16, CK15
(Fig.10-1); Hemidesmosome components-integrin alphas, integrin beta4;
Desmosome- components (Fig.10-2); Basement membrane componentslarnininl,
lamininS, collagen IV, collagen VII (Fig.10-3) and other important
extracellular matrix components as integrin-betal and fibronectin (Fig.10-4).
[0028] Figure 11 shows cytokine array analysis of secreted cytokines
and growth factors by umbilical cord mesenchymal stem cells (UCMC) in
comparison with human bone-marrow mesenchymal stem cells.
[0029] Figure 12 shows cytokine array analysis of secreted cytokines
and growth factors by umbilical cord epithelial stem cells (UCEC) in comparison
with human epidermal keratinocytes.
[0030] Figure 13. shows UCMC cells cultured in DMEM supplemented
with 10 % fetal calf serum (PCS) (Fig.13-1), serum-free media PTT-1 (Fig.13-
2), in serum-free media PTT-2 (Fig.13-3, Fig.13-4) and in serum-free media
PTT-3 (Fig.13-5). Fig. 13 also shows the growth of adipose derived stromal
cells (Fig.13-6) and bone marrow derived stromal cells (Fig.13-7) in serum free
medium PTT-3.
[0031] Figure 14 shows global gene expression in umbilical cord
epithelial and mesenchymal stem cells analyzed by DNA microarray. UCEC
expressed a total of 28055 genes and UCMC expressed a total of 34407 genes.
There are 27308 overlapping genes expressing in both cell types. 747 genes
expressed were unique to UCEC and 7099 genes expressed were unique to
UCMC. The selected genes of interest are presented in this Figure. Both stem
cell types expressed 140 genes related to embryonic stem cells and embryonic
development.
[0032] Figure 15 shows a schematic illustration of expansion of
umbilical cord epithelial and mesenchymal stem cells using repetitive explants
of umbilical cord lining membrane tissues.
[0033] Figure 16 depicts a cross section of an umbilical cord
demonstrating the umbilical cord amniotic lining membrane (LM), the contained
Wharton's jelly (WJ), as well as two umbilical arteries (UA) and one umbilical
vein (UV) supported within this jelly.
DETAILED DESCRIPTION
[0034]The invention is based on the surprising finding that the amniotic
membrane of umbilical cord represents a source, from which stem/progenitor
cells such as mesenchymal and epithelial stem/progenitor cells can be
successfully isolated and expanded under in vitro conditions. Even more
surprising is the finding that these cells show embryonic stem cell-like
characteristics. The amniotic membrane (also called amniotic lining membrane),
i.e. thin innermost membranous sac enclosing the placenta and developing
embryo of mammals,' has recently been used as a natural substrate in ocular
surface reconstruction and as a biological substrate for expanding limbal
epithelial stem cells (cf., e.g., Anderson, D.F. et al. (2001) Br. J. Ophthalmol. 85,
567-575; Gruterich, M. et al. (2003) Surv. Ophthalmol. 48, 631-646). However,
no methods have be'en described thus far for the isolation of stem/progenitor
cells from the amniotic membrane, at least for humans, nor has the amniotic
membrane covering the umbilical cord been reported as a source for stem cells.
[0035] The invention provides a method for isolating stem/progenitor cells
from the amniotic membrane of umbilical cord, the method comprising:
(a) separating the amniotic membrane from the other components of
the umbilical cord in vitro;
(b) culturing the amniotic membrane tissue obtained in step (a) under
conditions allowing cell proliferation; and
(c) isolating the stem/progenitor cells.
[0036]The term "stem/progenitor cell" as used herein refers to any cell
derived of umbilical cord having the capacities to self-renew indefinitely and to
differentiate in multiple cell or tissue types such as endothelial cells, epithelial
cells, fibroblasts, myocytes or neurons. Furthermore, the cells may be derived
of any mammalian species, such as mouse, rat, guinea pig, rabbit, goat, dog,
cat, sheep, monkey or human, with cells of human origin being preferred in one
embodiment.
[0037] The term "embryonic stem cell-like properties" refers to the ability
of the cells derived of umbilical cord that they can - almost like or exactly like
embryonic stem cells - differentiate spontaneously into all tissue types, meaning
that they are pluripotent stem cells.
[0038] The term "amniotic membrane" as used herein refers to the thin
innermost membranous sac enclosing the developing embryo of mammals.
During pregnancy, the fetus is surrounded and cushioned by a liquid called
amniotic fluid. This fluid, along with the fetus and the placenta, is enclosed
within a sac called the amniotic membrane, which also covers the umbilical
cord. The amniotic fluid is important for several reasons. It cushions and
protects the fetus, allowing the fetus to move freely. The amniotic fluid also
allows the umbilical cord to float, preventing it from being compressed and
cutting off the fetus1 supply of oxygen and nutrients derived from the circulating
blood within the placental blood vessels. The amniotic sac contains the amniotic
fluid which maintains a homeostatic environment protecting the fetal
environment from the outside world. This barrier additionally protects the fetus
from organisms (like bacteria or viruses) that could travel up the vagina and
potentially cause infection.
[0039] Media and reagents for tissue culture are well known in the art
(cf., for example, Pollard, J.W. and Walker, J.M. (1997) Basic Cell Culture
Protocols, Second Edition, Humana Press, Totowa, NJ; Freshney, R.I. (2000)
Culture of Animal Cells, Fourth Edition, Wiley-Liss, Hoboken, NJ). Examples of
suitable media for incubating/transporting umbilical cord tissue samples include,
but are not limited to, Dulbecco's Modified Eagle Medium (DMEM), RPMI
media, Hanks' Balanced Salt Solution (HBSS) phosphate buffered saline (PBS),
and L-15 medium, with the latter one being preferred in some embodiments.
Examples of appropriate media for culturing stem/progenitor cells according to
the invention include, but are not limited to, Dulbecco's Modified Eagle Medium
(DMEM), DMEM-F12, RPMI media, EpiLlfe medium, and Medium 171, with the
latter being preferred in some embodiments. The media may be supplemented
with fetal calf serum (PCS) or fetal bovine serum (FBS) as well as antibiotics,
growth factors, amino acids, inhibitors or the like, which is well within the
general knowledge of the skilled artisan.
[0040]In one embodiment, the invention provides a method, further
comprising:
(a") separating these stem/progenitor cells from the amniotic
membrane tissue by a enzymatic digestion and/or direct tissue explant
technique before cultivation. The term "enzymatic digestion technique" as used
herein means that enzymes are added to cleave the cells from the main tissue
mass .(here the amniotic membrane of the umbilical cord). The separated cells
are subsequently collected. The term "direct tissue explant technique" as used
herein means that the tissue is first placed in media without enzymes. Then
under careful conditions the cells separate from the main tissue mass by itselfand
the cells are then harvested for collection.
[0041] Methods for separating cells of a particular tissue or organ by
treatment with enzymes or by direct tissue explant are well known in the art (cf.,
for example, Pollard, J.W. and Walker, J.M. (1997) Basic Cell Culture Protocols,
Second Edition, Humana Press, Totowa, NJ; Freshney, R.l. (2000) Culture of
Animal Cells, Fourth Edition, Wiley-Liss, Hoboken, NJ). Any enzyme catalyzing
tissue dissociation may be used for performing the methods of the present
invention. In preferred embodiments, collagenase is used for that purpose. The
enzyme may be used as a crude preparation or in purified form. It may be
purified from any prokaryotic or eukaryotic organism (with Clostridium
histolyticum being most preferred) or produced recombinantly by means of gene
technology. Any type of collagenase may be employed, i.e. type 1, type 2, type
3, type 4, or any combination thereof. In some embodiments the use of
collagenase type 1 is being preferred.
[0042] In one embodiment, the invention provides a method for isolating
stem/progenitor cells that have embryonic stem cell-like properties. These cells
can ultimately be differentiated into, but not limited to, by morphology, epithelial
or mesenchymal cells.
[0043]Accordingly, in another embodiment, the invention provides a
method for isolating epithelial and/or mesenchymal stem/progenitor cells,
wherein in accordance with the above disclosure these cells may have
embryonic stem cell-like properties.
[0044] Epithelial stem/progenitor cells include any cells exhibiting a
epithelial cell like morphology (i.e. a polyhedral shape) that can be differentiated
into any type of epithelial cell such as, but not limited to, skin epithelial cells,
hair follicular cells, cornea epithelial cells, conjunctiva! epithelial cells, retinal
epithelial cells, liver epithelial cells, kidney epithelial cells, pancreatic epithelial
cells, oesophageal epithelial cells, small intestinal epithelial cells, large intestinal
epithelial cells, lung and airway epithelial cells, bladder epithelial cells or uterine
epithelial cells.
[0045] Mesenchymal stem/progenitor cells include any cells exhibiting a
mesenchymal cell like morphology (i.e. a spindle-like shape) that can be
differentiated into any type of mesenchymal cell such as, but not limited to, skin
fibroblasts, chondrocytes, osteoblasts, tenocytes, ligament fibroblasts,
cardiomyocytes, smooth muscle cells, skeletal muscle cells, adipocytes, cells
derived from endocrine glands, and all varieties and derivatives of
neurectodermal cells.
[0046] In another embodiment, the invention provides a method further
comprising:
(d) culturing the stem/progenitor cells under conditions allowing the
cells to undergo clonal expansion.
[0047]The term "clonal expansion" (sometimes also referred to as
"mitotic clonal expansion") relates to a process that occurs early in the
differentiation program of a cell, by which stem/progenitor cells become
committed to a particular lineage and then undergo terminal differentiation. It is
well known in the art that the conditions to induce clonal expansion of progenitor
cells may vary significantly between different cell types. Without being limited to
a particular method, the induction of clonal expansion is generally achieved by
cultivating the stem/progenitor cells in a medium that has been optimized for
cell proliferation. Such media are commercially available from many providers.
Non-limiting examples of such media are KGM®-Keratinocyte Medium
(Cambrex), MEGM-Mammary Epithelial Cell Medium (Cambrex), EpiLife
medium (Cascade Biologies) or Medium 171 (Cascade Biologies). Alternatively,
a culture medium may be supplemented with reagents inducing cell proliferation
such as growth factors. Such reagents may be admixed in a single solution
such as the Human Keratinocyte Growth Supplement Kit (Cascade Biologies),
to name one example, or may be supplemented individually. Such reagents
include, but are not limited to, growth factors (such as epidermal growth factor,
insulin-like growth factor-1, platelet-derived growth factor-BB, transforming
growth factor-^1, insulin, for example), hormones (such as a bovine pituitary
extract), hydrocortisone, transferrin and the like in any suitable combination to
induce clonal expansion of a given cell type. The term "clonal expansion" also
includes cultivation of the cell in vivo, for example, by injection of the cells into
mammals such as humans, mice, rats, monkeys, apes to name only a few.
[0048]In yet another embodiment, the invention provides a method
further comprising:
(e) culturing the stem/progenitor cells under conditions allowing the
differentiation of said cells into epithelial cells and/or mesenchymal cells; and
(f) isolating the differentiated cells.
[0049] In yet another embodiment, the invention provides a method,
further comprising:
(g) preserving the isolated stem/progenitor cells for further use.
[0050] Methods and protocols for preserving and storing of eukaryotic
cells, and in particular mammalian cells, are well known in the art (cf., for
example, Pollard, J.W. and Walker, J.M. (1997) Basic Cell Culture Protocols,
Second Edition, Humana Press, Totowa, NJ; Freshney, R.I. (2000) Culture of
Animal Cells, Fourth Edition, Wiley-Liss, Hoboken, NJ). Any method maintaining
the biological activity of the isolated epithelial or mesenchymal stem/progenitor
cells may be utilized in connection with the present invention. In one preferred
embodiment, the stem/progenitor cells are maintained and stored by using cryopreservation.
[0051] Accordingly, the invention is also directed to a progenitor/stem
cell derived from the amniotic membrane of umbilical cord by means of the
above methods. In addition, the invention is also directed to a cell bank
comprising or consisting of one or more progenitor/stem cells that have been
isolated as described here. This cell bank of progenitor/ stem cells may be
autologous to an individual or pooled, and subsequently can be employed by
further differentiation for regenerative medicine, tissue repair and regeneration,
for example.
[0052] In accordance with the above, the invention is also directed to a
pharmaceutical composition comprising a stem/progenitor cell isolated from the
amniotic membrane of umbilical cord by the above inventive method. The
pharmaceutical composition can be of any kind, and usually comprises the
stem/progenitor cells or a cellular extract thereof together with a suitable
therapeutically acceptable carrier/excipient. In some embodiments, the
pharmaceutical composition is adapted for systemic or topical application.
[0053] A pharmaceutical composition adapted for topical application may
be in liquid or viscous form. Examples thereof include an ointment, a cream,
and a lotion and the like. Examples for pharmaceutical compositions that are
suitable for systemic use are liquid compositions, wherein the stem/progenitor
cells or the cellular extract are dissolved in a buffer that is acceptable for
injection or infusion, for example.
[0054] Accordingly, the invention also relates to a method of treating a
subject having a disorder. This method comprises administering to the subject
an effective amount either of a stem/progenitor cell isolated as explained herein
or of a cellular extract derived from such a cell.
[0055] In principle, any condition which is suitable for being treated by
means of stem cells/progenitor cells can be treated with a cell or a cellular
extract of present invention. In some embodiments, the disorder is selected
from the group consisting of neoplastic disease, accelerated skin aging and skin
disorders, tissue disorders, visceral endocrine deficiencies, and neural
disorders.
[0056] The tissue disorder to be treated can be a congenital or an
acquired tissue deficiency. Examples of visceral endocrine deficiency that can
be treated with a cell of the invention include, but are not limited to, Diabetes
mellitus associated with insulin deficiency, testosterone deficiency, anemia,
hypoglycemia, hyperglycemia, pancreatic deficiency, adrenal deficiency, and
thyroid deficiencies.
[0057] Examples of neural disorders that can be treated include, but are
not limited to, Alzheimer's disease, Parkinson's, disease, Jacob Kreutzfeld's
disease, Lou Gehrig's disease, Huntington's disease and neural neoplastic
conditions.
[0058] An example of a skin disease is a wound or a damaged part of
the skin, for example, sun burned skin. Also aging of the skin is considered to
be a skin disease herein. Topical or similar delivery of stem/progenitor cells of
the invention or cellular extracts thereof, for example, as a constituent in lotions
or creams or any other suitable vehicle may thus be used for repair of sun
damaged skin and in addition may slow also down the aging process of skin
(anti-aging properties) by replenishing, and thus fortifying, deficient growth
factors and related peptide elements, without which skin aging would be
accelerated. The stem/progenitor cells may also migrate to injured regions of
the body such as surface wounds to form the necessary required cellular
elements necessary for the local reparative processes (cf. The Journal of
Immunology, 2001, 166: 7556-7562; or International Journal of Biochemical
and Cell Biology 2004; 36: 598-606.
[0059] The neoplastic disease may be cancer, in particular as recent
studies have demonstrated that stem cells may selectively target neoplastic
tumor tissue (Journal of the National Cancer Institute 2004; 96 (21): 1593-1603)
allowing for directed delivery of antineoplastic agents such as interferon to
neoplastic foci. The cancer can be any kind of cancer, including those cancers
that are able to form solid tumors, ranging from skin cancer to cancer of the
internal organs. Examples of cancers to be treaded include, squamous cell
carcinoma, breast ductal and lobular carcinoma, hepatocellular carcinoma,
nasopharyngeal carcinoma, lung cancer, bone cancer, pancreatic cancer, skin
cancer, cancer of the head or neck, cutaneous or intraocular malignant
melanoma, uterine cancer, ovarian cancer, rectal cancer, cancer of the anal
region, stomach cancer, colon cancer, breast cancer, testicular cancer, uterine
cancer, carcinoma of the fallopian tubes, carcinoma of the endometrium,
carcinoma of the cervix, carcinoma of the vagina, carcinoma of the vulva,
Hodgkin's Disease, non-Hodgkin's lymphoma, cancer of the esophagus, cancer
of the small intestine, cancer of the endocrine system, cancer of the thyroid
gland, cancer of the parathyroid gland, cancer of the adrenal gland, sarcoma of
soft tissue, cancer of the urethra, cancer of the penis, prostate cancer, chronic
or acute leukemias, solid tumors of childhood, lymphocytic lymphoma, cancer of
the bladder, cancer of the kidney or ureter, renal cell carcinoma, carcinoma of
the renal pelvis, neoplasm of the central nervous system (CMS), primary CNS
lymphoma, tumor angiogenesis, spinal axis tumor, brain stem glioma, pituitary
adenoma, Kaposi's sarcoma, epidermoid cancer or any combination of such
cancers, including disseminated (metastasising) forms thereof. In case of
treatment of a neoplastic disease the umbilical cord amnion derived stem cells
and/or their cellular extracts disclosed herein can be administered systemically
both as a direct treatment and/or as a carrier vehicle. In the latter case of antineoplastic
tumor therapy, the cells comprise an anti-neoplastic agent.
[0060] In another pharmaceutical use, stem/progenitor cells of the
present invention can be used for gene therapy. For this purpose, the cells can
be transformed with a nucleic acid encoding the protein that is to be produced in
the cells. The nucleic acid can be introduced into a cells of the invention using
any of the various methods that are well known to the skilled person, for
example, using a viral vector and/or a lipid containing transfection composition
such as as IBAfect (IBA GmbH, Gottingen, Germany), Fugene (Roche),
GenePorter (Gene Therapy Systems), Lipofectamine (Invitrogen), Superfect
(Qiagen), Metafecten (Biontex) or those ones described in the PCT application
WO 01/015755). In a related embodiment, the cells of the invention, after being
transformed with a nucleic acid encoding a polypeptide of choice, can be used
of recombinantly producing this polypeptide.
[0061] As mentioned above, stem cell extracts are rich in a variety of
growth factors and peptides that are relevant for normal tissue physiology. Such
growth factors and/or peptides may be deficient in exposed parts of the body,
such as the skin, which is the surface layer of all human beings protecting the
body from external elements for the maintenance of internal homeostasis.
Therefore in a further embodiment, stem/progenitor cells of the invention or
cellular extracts thereof are suitable for the treatment and/or maintenance of
internal homeostasis.
[0062] In a further embodiment and in line with the above disclosure, the
stem/progenitor cells of the invention can be used for the production of any
biological molecule. The biological molecule can be, for instance, any molecule
that is naturally produced in the cells or a molecule the coding nucleic acid of
which has been introduced into the cells via recombinant DMA technology.
Examples of molecules that can be produced by the cells of the invention
include, but are not limited to, a protein such as a cytokine, a growth factor such
as insulin-like growth factor (IGF), epidermal growth factor (EGF), transforming
growth factor beta (TGF-beta), Activin A, a bone morphogenetic protein (BMP),
PDGF or a hormone as insulin or erythropoietin or a transporter protein such
transferrin, a peptide such a growth factor or hormone (e.g. luteinic hormone
(LSH), follicle stimulating hormone (FSH)), a small organic molecule such as a
steroid hormone, an oligo- or polysaccharide, for example, heparin or heparan
suifate (cf., example WO 96/23003, or WO 96/02259 in this regard), a
proteoglycan, a glycoprotein such as collagen or laminin, or a lipid, to name
only a few.
[0063] In a further aspect and in accordance with recent approaches
(see, for example, Amit, M et al., Human feeder layers for human embryonic
stell cells, Biol Reprod 2003; 68: 2150-2156), the stem/progenitor cells
described here can be used as feeder layer for the cultivation of other
embryonic stem cells, in particular human embryonic stem cells. In one of these
embodiments the cells of the present invention are preferably of human origin,
since using human cells as feeder layer minimizes the risk of contaminating the
cell culture with animal-derived components such as animal pathogens or
immunogens. In this respect, it is to be noted that the cells of the invention can
be cultivated under serum free conditions. Accordingly, employing the cells as
feeder layer and cultivating the cell culture under with serum free media as the
one described herein later, or in Draper et al. (Culture and characterization of
human embryonic stem cell lines, Stem Cells Dev 2004, 13:325-336) or in the
International patent application WO 98/30679, for example.
[0064] In this connection, it is noted that in transplantation surgery and
cell-based therapy high quantities of low passage cells with a minimal
proportion of senescent cells (i.e., large proportion of high quality cells) are
crucial and are required to be derived within the shortest possible time during
cell expansion. For example, mesenchymal stem cells from bone marrow and
cord blood are low in quantity and therefore require expansion over many
passages for a long period of time in order to achieve the sufficient number of
cells required for cell transplant. The high passage cells however tend
to deteriorate in quality and may lead to cell senescence or cancerous
transformation.. It has been found here that high quantities of cells of the
present invention can be obtained by low passage numbers using a repetitive
explanation technique. The present invention thus also relates to a method of
cultivating stem/progenitors cells of the invention, wherein this method
comprises:
Obtaining a tissue explant from the amniotic membrane of umbilical cord;
Cultivating the tissue explant in suitable cultivation media and cultivation
conditions over a suitable period of time,
Optionally exposing the tissue explant to fresh cultivation media and
continuing the cultivation under suitable conditions over a suitable period of time
(cf., Fig. 15).
[0065] The cultivation can be carried out in for as many cycles
(passages) as wanted and be stopped once the desired number of cells has
been obtained. Exposing the tissue explant to fresh cultivation can be carried
out by removing the used cell cultivation medium from the vessel used for
growing the cells and adding fresh media to that vessel.' Instead of replacing the
media in the used vessel, exposing to fresh cultivation media can be achieved
by transferring the tissue explant to a new vessel which is filled with cultivation
media. The tissue explant used for cultivation/propagation of the cells can be
obtained by any suitable method, for example by the "direct tissue explant
technique" as explained above (in which the tissue is first placed in media
without enzymes, and then under careful conditions the cells separate from the
main tissue mass by itself- and the cells are then harvested for collection)..
[0066] The cultivation of the tissue explants can be carried out in any
media that is suitable for cultivation of mammal cells. Examples include the
conventional and commercially available media that are given above with
respect to the cultivation or the clonal expansion of the cells of the invention
such as, but not limited to, KGM®-Keratinocyte Medium (Cambrex), MEGMMammary
Epithelial Cell Medium (Cambrex) EpiLife medium (Cascade
Biologies), Medium 171 (Cascade Biologies), DMEM, DMEM-F12 or RPMI
media. The cultivation is typically carried out at conditions (temperature,
atmosphere) that are normally used for cultivation of cells of the species of
which the cells are derived, for example, at 37°C in air atmosphere with 5%CC>2.
In one embodiment, the cultivation is carried out using serum free, in particular
bovine serum free media. The cultivation (in one passage) is performed for any
suitable time the cells need for growth, typically, but by no means limited to, for
about 1 to several days, for example to about 7 or about 8 days.
[0067]The inventions illustratively described herein may suitably be
practiced in the absence of any element or elements, limitation or limitations,
not specifically disclosed herein. Thus, for example, the terms "comprising",
"including", "containing", etc. shall be read expansively and without limitation.
Additionally, the terms and expressions employed herein have been used as
terms of description and not of limitation, and there is no intention in the use of
such terms and expressions of excluding any equivalents of the features shown
and described or portions thereof, but it is recognized that various modifications
are possible within the scope of the invention claimed. Thus, it should be
understood that although the present invention has been specifically disclosed
by preferred embodiments and optional features, modification and variation of
the inventions embodied therein herein disclosed may be resorted to by those
skilled in the art, and that such modifications and variations are considered to
be within the scope of this invention.
[0068]The invention has been described broadly and generically herein.
Each .of the narrower species and subgeneric groupings falling within the
generic disclosure also form part of the invention. This includes the generic
description of the invention with a proviso or negative limitation removing any
subject matter from the genus, regardless of whether or not the excised material
is specifically recited herein.
[0069] Other embodiments are within the following claims and nonlimiting
examples. In addition, where features or aspects of the invention are
described in terms of Markush groups, those skilled in the art will recognize that
the invention is also thereby described in terms of any individual member or
subgroup of members of the Markush group.
EXAMPLES
Example 1: Collection of umbilical cord tissue
[0070] Umbilical cord tissue is collected immediately after delivery of the
child. The specimen is rinsed clean and immediately transferred into a 500ml
sterile glass bottle containing culture transport medium (L-15 medium
supplemented with 50 lU/ml penicillin, 50 ng/ml streptomycin, 250 pg/ml
fungizone, 50 jj,g/ml gentamicin; all reagents purchased from Invitrogen) prior to
transport to the laboratory. In the laboratory, stem cell extraction is conducted in
a laminar flow hood under sterile conditions. The specimen is first transferred to
a sterile stainless steel tray. All remaining blood in the cord vessels is removed
by multiple- syringing washes using warm phosphate-buffered saline (PBS)
supplemented with 5 lU/ml heparin (from Sigma). Plain PBS without heparin is
used in the final washes. The umbilical cord tissue specimen is then cut into
pieces 2cm in length and transferred into 10 cm diameter cell culture dishes,
where further washing and disinfection is performed with 70% ethanol followed
by multiple washes using PBS containing an antibiotic mixture (50 lU/ml
penicillin, 50 ug/ml streptomycin, 250 pg/ml fungizone, 50 ug/ml gentamicin; all
purchased from Invitrogen) until the solution becomes clear.
Example 2: Cell separation/cultivation
[0071] Dissection of umbilical cord tissue is first performed to separate
the umbilical cord amniotic membrane from Wharton's jelly (i.e. the matrix of
umbilical cord) and other internal components. The isolated amniotic membrane
is then cut into small pieces (0.5 cm x 0.5 cm) for cell isolation. Explant is
performed by placing the pieces of umbilical cord amniotic membrane on tissue
culture dishes at different cell culture conditions for isolation of either epithelial
or mesenchymal stem cells.
[0072] For mesenchymal cell separation/cultivation, the explants were
submerged in 5 ml DMEM (Invitrogen) supplemented with 10% fetal bovine
serum (Hyclone) (DMEM/10% FBS) and maintained in a CO2 cell culture
incubator at 37°C. The medium was changed every 2 or 3 days. Cell outgrowth
was monitored under light microscopy. Outgrowing cells were harvested by
trypsinization (0.125% trypsin/0.05% EDTA) for further expansion and cryopreservation
using DMEM/10% FBS.
[0073] For epithelial cell separation/cultivation, cell culture plastic
surfaces were coated with collagen 1/ collagen 4 mixtures (1:2) before placing
the tissue samples on the surface. The tissue samples were submerged in 5 ml
EpiLife medium or Medium 171 (both from Cascade Biologies). The medium
was changed every 2 or 3 days. Cell outgrowth from tissue culture explants was
monitored under light microscopy. Outgrowing cells were harvested by
trypsinization (0.125% trypsin/0.05% EDTA) using EpiLife medium or Medium
171.
[0074] For the enzymatic extraction method of cells, umbilical cord
amniotic membrane was divided into small pieces of 0.5 cm x 0.5 cm and
digested in 0.1% (w/v) collagenase typel solution (Roche Diagnostics) at 37°C
for 6 hours. The samples were vortexed every 15 min for 2 min. Cells were
harvested by centrifugation at 4000 rpm for 30 min. Two different approaches
were employed to isolate either epithelial or mesenchymal stem cells.
[0075] For isolation of epithelial stem cells, cell pellets were
resuspended in EpiLife medium or Medium 171 (both from Cascade Biologies)
supplemented with 50 jog/ml insulin-like growth factor-1 (IGF-1), 50 jog/ml
platelet-derived growth factor-BB (PDGF-BB), 5jag/ml transforming growth
factor-/?1 (TGF-£1), and 5 fig/ml insulin (all obtained from R&D Systems),
counted and seeded on 10 cm tissue culture dishes pre-coated with collagen
1/coIlagen 4 mixtures (1:2; Becton Dickinson) at density of 1 x 106 cells/dish.
After 24 hours, attached cells were washed with warm PBS and medium was
replaced with supplement-added EpiLife medium or Medium 171. The medium
was changed every 2 or 3 days. Cell growth and expanding clonal formation
was monitored under light microscopy. At a confluence of about 70%, cells were
sub-cultured by trypsinization (0.125% trypsin/0.05% EDTA) for further
expansion and cryo-preservation.
[0076] For isolation of mesenchymal stem cells, cell pellets were
resuspended in DMEM/10% FBS, counted and seeded on 10 cm tissue culture
dishes at density of 1 x 106 cells/dish. The culture medium was changed every 2
or 3 days. Cell growth and expansion was monitored under light microscopy. At
a confluence of about 90%, cells were sub-cultured as outlined above.
[0077] For cultivation of epithelial and mesenchymal stem cells on
feeder layer, umbilical cord lining membrane was digested by collagenase
treatment, counted and seeded on 10cm tissue culture dishes coated with
lethally irradiated or Mitomycin C treated 3T3 fibroblasts (feeder layer) in
Green's medium. The culture medium was changed every 2 or 3 days. Colony
formation was monitored under light microscopy and photographed.
Example 3: Identification of stem/progenitor cells
[OQ78]Epithelial cells: Fig. 1 shows pictures of outgrowing epithelial cells
from umbilical cord amniotic membrane prepared by the method using tissue
explant (40 x magnification). Pictures were taken at day 2 (Fig. 1A) and day 5
(Fig. 1B, C) of tissue culture. Cell morphology analysis demonstrated
polyhedral shaped epithelial-like cells. Enzymatic digestion of the umbilical cord
segments yielded similar (Fig. 2), epithelial cells at day 2 (Fig. A, C) and day 5
(Fig. B, D) (40 x magnification). Fig.7 shows pictures of colony formation of
epithelial stem cells from umbilical cord amniotic membrane cultured on feeder
layer using Green's method (40 x magnification). A colony of polyhedral shaped
epithelial-like cells expanded rapidly from day 3 to day 7.
[0079] Mesenchymal cells: Outgrowth of mesenchymal cells explanted
from umbilical cord amniotic membrane was observed as early as 48 hours
after placement in tissue culture dishes using DMEM supplemented with 10%
fetal calf serum (PCS) as culture medium (Fig. 3A, C) (40 x magnification). The
cells were characterized by their spindle shaped morphology, and migrated and
expanded both easily and quickly in vitro, closely resembling fibroblasts (Fig.
3B, D) (40 x magnification). Similar observations were noted in the cell group
isolated by collagenase enzymatic digestion (Fig. 4). Fig. 4A shows
mesenchymal cells isolated from umbilical cord amniotic membrane at day 2.
Cell proliferation was observed at day 5 (Fig. 4B) (40 x magnification). Figures
6 and 8 show pictures of colony formation of mesenchymal stem cells from
umbilical cord amniotic membrane cultured on non-feeder layer and feeder
layer condition in DMEM/10%FCS (40 x magnification). The colonies of
elongated shaped fibroblastic-like cells expanded rapidly from day 3 to day 7.
[0080] Western blot analysis (Fig.9) shows that mesenchymal stem cells
from umbilical cord amniotic membrane (UCMC) and umbilical cord epithelial
cells (UCEC) isolated in accordance with the invention expressed the POU5f1
gene which encodes the transcription factor Octamer-4 (Oct-4) a specific
marker of embryonic stem cells (cf. Niwa, H., Miyazaki, JM and Smith, A.G.
(2000). Nat. Genet. 24, 372-376), Thus, this analysis indicates the embryoniclike
properties of these stem cells. These cells also highly expressed the other
growth factors such as connective tissue growth factor (CTGF), vascular
endothelial growth factor (VEGF), placenta-like growth factor PLGF, STATS,
stem cell factor (SCF), Hepatoma-derived Growth Factor (HDGF), Fibroblast
Growth Factor-2 (FGF-2), Platelet-derived Growth Factor (PDGF), alpha-
Smooth Muscle Actin (u-SMA), Fibronectin, Decorin, Syndecan-1,2,3,4. In Fig.
9, the expression of these genes is compared to human dermal fibroblasts,
bone marrow mesenchymal cells (BMSC) and adipose-derived mesenchymal
cells (ADMC). Fig. 9 also shows highly secreted Activin A and Follistatin (both
of which proteins are well known to promote tissue repair and regeneration,
enhanced angiogenesis, and maintain embryonic stem cell culture, so that
expression of the respective genes is a sign for the embryonic properties and
ability of the cells to differentiate) detected by ELISA assay in supematants of
umbilical cord mesenchymal and epithelial stem cell culture in comparison with
bone marrow, adipose derived stem cells, human dermal fibroblasts and
epidermal keratinocytes. Also these results indicate 'that the cells of the
invention are promising candidates in therapeutic application of these cells
areas such as regenerative medicine, aging medicine, tissue repair and tissue
engineering.
[0081] Mesenchymal cells were further characterized by analysis of
secreted cytokines and growth factors in comparison with human bone-marrow
mesenchymal stem cells. The umbilical cord epithelial stem cells (UCEC) were
analysed in comparison with human epidermal keratinocytes. This analysis was
carried out as follows: Briefly, UMMC, UCEC, dermal fibroblasts, bone-marrow
mesenchymal cells, epidermal keratinocytes were cultured in growth media until
100% confluence (37°C, 5% CO2) and then synchronized in starvation medium
(serum-free DMEM) for 48 hours. The next day, the medium was replaced the
next against fresh serum-free DMEM and the cells then were cultivated for
another 48 hours. Conditioned media were collected, concentrated and
analyzed using a Cytokine Array (RayBiotech, Inc. GA, USA).
[0082]The results of this analysis show that UCMC secrete Interleukin-6
(lL-6); (MCP1); hepatocyte growth factor (HGF); lnterieukin-8 (IL8); sTNFRI;
GRO; TIMP1; TIMP2; TRAILR3; uPAR; ICAM1; IGFBP3; IGFBP6 (Fig.11),
whereas UCEC secrete IGFBP-4; PARC; EGF; IGFBP-2; IL-6; Angiogenin;
GCP-2; IL1Ra; MCP-1; RANTES; SCF; TNF# HGF; IL8; sTNFR; GRO; GRO-a;
Amphiregulin; IL-1R4/ST2; T1MP1; TIMP2; uPAR; VEGF (Fig.12).
[0083] Accordingly, this shows that both cells types secrete large
amounts of cytokines and growth factors that play important roles in
developmental biology, tissue homeostasis, tissue repair and regeneration and
angiogenesis. This further demonstrates the versatility of the cells of the
invention for use in the respective therapeutic applications.
[0084] In addition, the cells of the invention were further examined with
respect to their safety profile using mouse teratoma formation assay as an
indicator. Six SCID mice were used in these experiments. A suspension of more
than 2 million UCMG was injected with a sterile 25G needle into the thigh
muscle of each SCID mouse. Animals were kept up to 6 months and tumor
formation, was assessed. No tumor formation was observed in these mice (data
not shown). This indicates that the cells of the invention are safe and do not
have any capability to form tumors, benign or otherwise.
Example 4: Cultivation of stem/progenitor cells in serum free media
[0085] UCMC cells were cultured in DMEM containing 10 PCS and in
serum-free media, PTT-1, PTT-2 and PTT-3. The three media PTT-1, PTT-2
and PTT-3 were prepared by one of the present inventors, Dr Phan. in brief,
these 3 media do not contain fetal bovine or human serum, but contain different
cytokines and growth factors such as IGF, EGF, TGF-beta, Activin A, BMPs,
PDGF, transferrin, and insulin. The growth factor components vary between
media to assess differential growth characteristics. The cultivation was carried
out as follows: Different proportions of growth factors and cytokines were added
in basal media. UCMC were thawed and maintained in these media for 10 days.
Ceil proliferation was monitored under light microscopy.
[0086] Fig. 13 shows good UCMC growth in the 4 different media groups
(Fig. 13-1 to Fig.13-5), wherein the morphology of UCMC cells is different
depending on the ratio or proportion of cytokines or growth factors present in
the respective media. In contrast, bone marrow and adipose-derived
mesenchymal cells did not grow well in these serum-free media (Fig.13-6 and
Fig.13-7). Accordingly, the good growth of the UCMC demonstrates the
robustness of the cells of the invention and their high viability, indicating that
their growth characteristics are superior to conventional sources of
mesenchymal stem cells as bone marrow derived and adipose-derived
mesenchymal cells. In this respect, it is worth to note that (bovine) serum free
medium was used in these experiments and that the majority of human
mesenchymal cells do not grow well in serum-free medium systems. Thus,
using the cells of the invention in connection with defined serum-free media
technologies is a big advantage in cell therapy as the risks of using fetal bovine
serum for cell culture and expansion are removed. (Although use of bovine
serum has been practiced for a long time and typically optimizes cell growth,
concerns of its used have been raised as to the transmission of zoonoses as
Bovine Spongiform Encephalopathy (Mad Cow Disease)).
Example 5: Characterization of the gene expression profile of umbilical
cord epithelial and mesenchymal stem cells
[0087]The gene expression profile of umbilical cord epithelial and
mesenchymal stem cells was analyzed using a DNA microarray. For this
purpose, UCMC and UCEC were cultured in growth media at 37°C, 5% CO2
until 100% confluence. Cells were synchronized in basal media for 48 hours
then replaced with fresh basal media for another 48 hours. Total RNA was
harvested and sent to Silicon Genetics Microarray Service. Data analysis was
performed using GeneSpring 7.2). Fig. 14 summarizes the global gene
expression. UCEC expressed a total of 28055 genes and UCMC expressed a
total of 34407 genes. There are 27308 overlapping genes expressing in both
cell types. 747 genes expressed were unique to UCEC and 7099 genes
expressed were unique to UCMC. The selected genes of interest are presented
in Fig. 14.
[0088] Both stem cell types expressed 140 genes related to embryonic
stem cells and embryonic development, further supporting that the cells of the
invention have embryonic stem cell-like properties: Nanog; Alpha-fetal protein;
Pre-B-cell leukemia transcription factor 3; Laminin alpha 5; Carcinoembryonic
antigen-like 1; abhydrolase domain containing 2; Delta-like 3 (Drosophila);
Muscleblind-like (Drosophila); GNAS complex locus; Carcinoembryonic antigenrelated
cell adhesion molecule 3; Palmitoyl-protein thioesterase 2; Pregnancy
specific beta-1-glycoprotein 2; Carcinoembryonic antigen-like 1; Embryonic
ectoderm development;Maternal embryonic leucine zipper kinase; Chorionic
somatomammotropin hormone 2; Forkhead box D3;radical fringe homolog
(Drosophila); Kinesin family member 1B; Myosin, heavy polypeptide 3, skeletal
muscle, embryonic; Split hand/foot malformation (ectrodactyiy) type 3; TEA
domain family member 3; Laminin, alpha 1; Chorionic somatomammotropin
hormone 1; placental lactogen; Corticotropin releasing hormone receptor 1;
thyrotrophic embryonic factor; Aryl-hydrocarbon receptor nuclear translocator
Membrane frizzled-related protein; Neuregulin 1'Collagen, type XVI, alpha 1;
Neuregulin 1; Chorionic somatomammotropin hormone 1 (placental lactogen);
CUG triplet repeat, RNA binding protein 1; Chorionic somatomammotropin
hormone 1 (placental lactogen) Bystln-llke; MyoD family inhibitor; Retinoic acid
induced 2; GNAS complex locus; Pre-B-cell leukemia transcription factor 4;
Laminin, alpha 2 (merosin, congenital muscular dystrophy); SMAD, mothers
against DPP homolog 1 (Drosophila); Homo sapiens transcribed sequence with
moderate similarity to protein pir:D28928 (H.sapiens) D28928 pregnancy26
specific beta-1 glycoprotein IB, abortive - human (fragment); Kinesin family
member 1B; Bruno-like 4, RNA binding protein (Drosophila); Embryo brain
specific protein;Pregnancy-induced growth inhibitonSMAD, mothers against
DPP homolog 5 (Drosophila); Chorionic somatomammotropin hormone
2;AdenyIate cyclase activating polypeptide 1 (pituitary);Carcinoembryonic
antigen-related cell adhesion molecule; Laminin, alpha 3; Protein Ofucosyltransferase
1; Jagged 1 (Alagille syndrome); Twisted gastrulation
homolog 1 (Drosophila); ELAV (embryonic lethal, abnormal vision, Drosophila)-
like 3 (Hu antigen C); Thyrotrophic embryonic factor, Solute carrier family 43,
member 3; Inversin; nephronophthisis 2 (infantile); inversion of embryonic
turning; Homo sapiens inversin (INVS), transcript variant 2, mRNA;Homo
sapiens transcribed sequences; Homeo box D8; Embryonal Fyn-associated
substrate; ELAV (embryonic lethal, abnormal vision, Drosophila)-Iike 1 (Hu
antigen R); Basic helix-loop-helix domain containing, class B, 2; Oxytocin
receptor, Teratocarcinoma-derived growth factor 1; Fms-related tyrosine kinase
1 (vascular endothelial growth factor/vascular permeability factor receptor);
Adrenomedullin; Nuclear receptor coactivator6-CUG triplet repeat, RNA binding
protein 1; Twisted gastrulation homolog 1 (Drosophila); Carcinoembryonic
antigen-related celi adhesion molecule 4;Protein tyrosine phosphatase, receptor
type, R; Acrg embryonic lethality (mouse) minimal region ortholog; EPH
receptor A3;Delta-like 1 (Drosophila); Nasal embryonic LHRH factor;
Transcription factor CP2-like 1; Split hand/foot malformation (ectrodactyly) type
3; Jagged 2; Homo sapiens transcribed sequence; Neuregulin 1; Split hand/foot
malformation (ectrodactyly) type 1; Solute carrier family 43, member 3;
Hydroxyacyl-Coenzyme A dehydrogenase/3-ketoacyl-Coenzyme A
thiolase/enoyl-Coenzyme A hydratase (trifunctional protein), alpha
subunit;Fucosyitransferase 10 (alpha (1,3) fucosyltransferase); Acrg embryonic
lethality (mouse) minimal region ortholog; Carcinoembryonic antigen-related cell
adhesion molecule 7; Nucleophosmin/nucleoplasmin, 2; Fc fragment of IgG,
receptor, transporter, alpha; Twisted gastrulation homolog 1 (Drosophila); Homo
sapiens similar to vacuolar protein Bortlng 35; maternal-embryonic 3
(LOC146485), mRNA;abhydrolase domain containing 2; T, brachyury homolog
(mouse); A dlslntegrin and metalloproteinase domain 10; Ribosomal protein
L29; Endothelin converting enzyme 2; ELAV (embryonic lethal, abnormal vision,
Drosophila)-like 1 (Hu antigen R);Trophinin;Homeo box B6; Laminin, alpha
Homeo box B6; hypothetical protein FLJ13456; NACHT, leucine rich repeat and
PYD containing 5; ELAV (embryonic lethal, abnormal vision, Drosophila)-like 1
(Hu antigen R); Undifferentiated embryonic cell transcription factor 1;
Pregnancy-associated plasma protein A, pappalysin 1; Secretoglobln, family
1A, member 1 (uteroglobin); Parathyroid hormone-like hormone;
Carclnoembryonic antigen-related cell adhesion molecule 1 (biliary
glycoprotein); Laminin, alpha 1.
[0089] Both stem cell types also expressed thousands of genes related to
developmental biology, cell growth and differentiation, cell homeostasis, cell
and tissue repair and regeneration. Examples of such growth factors and their
receptors is as follows: (G-CSF, FGFs, IGFs, KGF, NGF, VEGFs, PIGF,
Angiopoietin, CTGF, PDGFs, HGF, EGF, HDGF, TGF-beta, Activins and
inhibins, Follistatin, BMPs, SCF/c-Kit, LIF, WNTs, SDFs, OncostatinM,
Interleukins, Chemokines and many others); MMPs, TIMPs extracellular
matrices (collagens, laminins, fibronectins, vitronectins, tenascins, intergrins,
syndecans, decorin, fibromoludin, proteoglycans, sparc/osteonectin, mucin,
netrin, glypican, cartilage associated protein, matrilin, hyaluronan, fibulln,
ADAMTS, biglycan, discoidin, desmosome components, ICAMs, cadherins,
catenins and many others); cytokeratins.
[0090]There are groups of genes present only in UCMC. These genes
are related to the following: Normal Physiological Processes (Insulin-like growth
factor 1 (somatomedin C); Insulin-like 4 (placenta); Relaxin 1; Plasminogen;
Insulin-like growth factor 1 (somatomedin C); Insulin-like 5; Insulin-like growth
factor 1 (somatomedin C); Insulin-like growth factor 2 (somatomedin ,
A),Homeostasis (Radial spokehead-Hke 1; Hemochromatosis; Chemoklne (OC
motif) ligand 5; Interleukin 31 receptor A; Chemokine (C-X-C motif) ligand 12
(stromal cell-derived factor 1); Nuclear receptor subfamily 3, group C, member
2; Hemochromatosis; Chemokine (C-C motif) ligand 23; Chemokine (C-C motif)
ligand 23; Ferritin mitochondrial; Peroxisome proliferative activated receptor,
gamma, coactivator 1, alpha; Surfactant, pulmonary-associated protein D;
Chemokine (C-C motif) ligand 11; Chemokine (C-C motif) ligand 3; Egl nine
homolog 2 (C. elegans); Peroxisome proliferative activated receptor, gamma,
coactivator 1, beta; Chemokine (C-C motif) ligand 1; Chemokine (C-X-C motif)
ligand 12 (stromal cell-derived factor 1); ATPase, Na+/K+ transporting, alpha 2
(+) polypeptide; Chemokine (C motif) iigand 2; Hemopexin; Ryanodine receptor
3), Morphogenesis (Spectrin, alpha, erythrocytic 1 (elliptocytosis 2); Homeo box
D3; Eyes absent homolog 1 (Drosophila); Ras homolog gene family, member J;
Leukocyte specific transcript 1; Ectodysplasin A2 receptor, Glypican 3; Paired
box gene 7; Corin, serins protease; Dishevelled, dsh homolog 1 (Drosophila);
Ras homolog gene family, member J; T-box 3 (ulnar mammary syndrome);
Chondroitin beta1,4 N-acetylgalactosaminyltransferase; Chondroitin beta"! ,4 Nacetylgalactosaminyltransferase;
SRY (sex determining region Y>box 10;
Myosin, heavy polypeptide 9, non-muscle; Luteinizing
hormone/choriogonadotropin receptor; radical fringe homolog (Drosophila);
Secreted frizzled-related protein 5; Wingless-type MMTV integration site family,
member 11; Eyes absent homolog 2 (Drosophiia); Musclebtind-like
(Drosophila); T-box 5; Mab-21-like 1 (C. elegans); Growth arrest-specific 2; Sex
comb on midleg homolog 1 (Drosophila); T-box 6; Filamin-binding LIM protein-
1; Melanoma cell adhesion molecule; Twist homolog 1 (acrocephalosyndactyly
3; Saethre-Chotzen syndrome) (Drosophila); Homeo box A11; Keratocan;
Fibroblast growth factor 1 (acidic); Carboxypeptidase M; CDC42 effector protein
(Rho GTPase binding) 4; LIM homeobox transcription factor 1, beta; Engrailed
homolog 1; Carboxypeptidase M; Fibroblast growth factor 8 (androgeninduced);
Fibroblast growth factor 18; Leukocyte specific transcript 1;
Endothelin 3; Paired-like homeodomain transcription factor 1), Embryonic
Development (Pregnancy specific beta-1-glycoprotein 3; ELAV (embryonic
lethal, abnormal vision, DrosophilaHike 4 (Hu antigen D); G protein-coupled
receptor 10; Ectodysplasin A2 receptor, ATP-binding cassette, sub-family B
(MDR/TAP), member 4; Pregnancy specific beta-1-glycoprotein 11; Nasal
embryonic LHRH factor, Relaxin 1; Notch homolog 4 (Drosophila); Pregnancy
specific beta-1-glycoprotein 6; pih-2P; Homo sapiens pregnancy-induced
hypertension syndrome-related protein (PIH2); Oviductal glycoprotein 1,
120kDa (mucin 9, oviductin); Progestagen-associated endometrial protein;
Myosin, light poiypeptide 4, alkali; atrial, embryonic; Prolactin; Notch homolog
(Drosophila); Pre-B-ce!l leukemia transcription factor 1; radical fringe homolog
(Drosophila); Corticotropin releasing hormone; Nuclear receptor subfamily 3,
group C, member 2; Neuregulin 2; Musclebllnd-like (Drosophila); Myosin, light
polypeptide 4, alkali; atrial, embryonic; Homo sapiens cDNA FLJ27401 fis,
clone WMC03071; Extraembryonic, spermatogenesis, homeobox 1-like; Insulinlike
4 (placenta); Human processed pseudo-pregnancy-specific glycoprotein
(PSG12) gene, exon B2C containing 3' untranslated regions of 2 alternative
splice sites C1 and C2; Fms-related tyrosine kinase 1 (vascular endothelial
growth factor/vascular permeability factor receptor); Pre-B-cell leukemia
transcription factor 1; Pregnancy specific beta-1 -glycoprotein 3;
carcinoembryonic antigen-related cell adhesion molecule 1 (biliary
glycoprotein); Steroid sulfatase (microsomal), arylsulfatase C, isozyme S;
Homeo box B6; Protein O-fucosyltransferase 1; LIM homeobox transcription
factor 1, beta; Carcinoembryonic antigen-related cell adhesion molecule 1
(biliary glycoprotein); Follicle stimulating hormone, beta polypeptide;
Angiotensinogen (serine (or cysteine) proteinase inhibitor, clade A (alpha-1
antiproteinase, antitrypsin), member 8); Carcinoembryonic antigen-related cell
adhesion molecule 6 (non-specific cross reacting antigen); Protein kinase C,
alpha binding protein; Collectin sub-family member 10 (C-type lectin); Laminin,
alpha 1), the Extracellular Space (Carboxylesterase 1 (monocyte/macrophage
serine esterase 1); Fibroblast growth factor 5; Progastricsin (pepsinogen C);
Sperm associated antigen 11; Proprotein convertase subtilisin/kexin type 2 ;
Hyaluronan binding protein 2; Sema domain, immunoglobulin domain (Ig), short
basic domain, secreted, (semaphorin) 3F; Interleukin 2; Chymotrypsin-like;
Nome disease (pseudoglioma); mucin 5, subtypes A and C,
tracheobronchial/gastric; Carboxypeptidase B2 (plasma, carboxypeptidase U);
radical fringe homolog (Drosophila); Pregnancy specific beta-1-glycoprotein 11;
Meprin A, alpha (PABA peptide hydrolase); Tachykinin, precursor 1 (substance
K, substance P, neurokinin 1, neurokinin 2, neuromedin L, neurokinin alpha,
neuropeptide K, neuropeptide gamma); Fibroblast growth factor 8 (androgeninduced);
Fibroblast growth factor 13; Hemopexin; Breast cancer 2, early onset;
Fibroblast growth factor 14; Retinoschisis (X-linked, juvenile) 1; Chitinase 3-Iike
1 (cartilage glycoprotein-39); Dystonin; Secretoglobin, family 1D, member 2;
Noggin; WAP four-disulfide core domain 2; CDS antigen-like (scavenger
receptor cysteine rich family); Scrapie responsive protein 1; Gremlin 1 homolog,
cysteine knot superfamily (Xenopus laevis); Interleukin 16 (lymphocyte
chemoattractant factor); Chemokine (C-C motif) ligand 26; Nucleobindin 1;
Fibroblast growth factor 18; Insulin-like growth factor binding protein 1;
Surfactant, pulmonary-associated protein A1; Delta-like 1 homolog (Drosophila);
Cocaine- and amphetamine-regulated transcript; Meprin A, beta; Interleukin
17F; Complement factor H; Cysteine-rich secretory protein 2; Dystonin; WAP
four-disulfide core domain 1; Prolactin; Surfactant, pulmonary-associated
protein B; Fibroblast growth factor 5; Dickkopf homolog 2 (Xenopus laevis);
Sperm associated antigen 11; Chemokine (C-C motif) ligand 11; Meprin A,
alpha (PABA peptide hydrolase); Chitinase 3-like 2; C-fos induced growth factor
(vascular endothelial growth factor D); Chemokine (C-C motif) ligand 4;
Poliovirus receptor; Hyaluronoglucosaminidase 1; Oviductal glycoprotein 1,
120kDa (mucin 9, oviductin); Chemokine (C-X-C motif) ligand 9; Secreted
frizzled-related protein 5; Amelogenin (amelogenesis imperfecta 1, X-linked);
Relaxin 1; Sparc/osteonectin, cwcv and kazal-like domains proteoglycan
(testican); Chemokine (C-C motif) iigand 26; Fibroblast growth factor 1 (acidic);
Angiopoiet'n-like 2; Fms-related tyrosine kinase 1 (vascular endothelial growth
factor/vascular permeability factor receptor); Dystonin; Insulin-like 4 (placenta);
Transcobalamin II; macrocytic anemia; Chemokine (C-C motif) ligand 1; Insulinlike
growth factor binding protein, acid labile subunit; Complement factor H;
Pregnancy specific beta-1-glycoprotein 6; Silver homolog (mouse);
Proteoglycan 4; Fibroblast growth factor 16; Cytokine-like protein C17;
Granulysin; Angiopoietin 2; Chromogranin B (secretogranin 1); Sema domain,
immunoglobulin domain (Ig), and GPI membrane anchor, (semaphorin) 7A;
Pleiotrophin (heparin binding growth factor 8, neurite growth-promoting factor
1); Chloride channel, calcium activated, family member 3; Secretoglobin, family
1D, member 1; Fibulin 1; Phospholipase A2 receptor 1, 180kDa), and the
Extracellular Matrix (ADAMTS-like 1; Periostin, osteoblast specific factor;
Glypican 5; Leucine rich repeat neuronal 3; Transglutaminase 2 (C polypeptide,
protein-glutamine-gamma-glutamyltransferase); A disintegrin-Iike and
metalloprotease (reprolysin type) with thrombospondin type 1 motif, 2;
Microfibrillar-associated protein 4; Glypican 3; Collagen, type V, alpha 3; Tissue
inhibitor of metalloprotemase 2; Keratocan; Cartilage oligomeric matrix protein;
Lumican; Hyaluronan and proteoglycan link protein 3; Statherin; A disintegrin-
like and metalloprotease (reprolysin type) with thrombospondin type 1 motif,
Spondin 1, extracellular matrix protein; Chitinase 3-like 1 (cartilage glycoprotein31
39); Collagen, type IV, alpha 3 (Goodpasture antigen); Wingless-type MMTV
integration site family, member 7B; Collagen, type VI, alpha 2; Lipocalin 7;
Hyaluronan and proteoglycan link protein 4; A disintegrin-like and
metalloprotease (reprolysin type) with thrombospondin type 1 motif, 5
(aggrecanase-2); Fibronectin 1; Matrilin 1, cartilage matrix protein; Hypothetical
protein FLJ13710; Chondroitin beta1,4 N-acetylgalactosaminyltransferase;
Matrix metalloproteinase 16 (membrane-inserted); Von Willebrand factor;
Collagen, type VI, alpha 2; Transmembrane protease, serine 6 ; Matrix
metalloproteinase 23B ; Matrix metalloproteinase 14 (membrane-inserted);
Leucine rich repeat neuronal 3; SPARC-like 1 (mastQ, hevin);
Sparc/osteonectin, cwcv and kazal-like domains proteoglycan (testican) 3;
Dermatopontin; collagen, type XIV, alpha 1 (undulin); Amelogenin, Y-linked;
Nidogen (enactin); ADAMTS-like 2; Hyaluronan and proteoglycan link protein
Collagen, type XV, alpha 1; Glypican 6; Matrix metalloproteinase 12
(macrophage elastase); Amelogenin (amelogenesis imperfecta 1, X-linked); A
disintegrin-like and metalloprotease (reprolysin type) with thrombospondin type
1 motif, 15; Transmembrane protease, serine 6; A disintegrin-like and
metalloprotease (reprolysin type) with thrombospondin type 1 motif, 16;
Sparc/osteonectin, cwcv and kazaHike domains proteoglycan (testican); A
disintegrin-like and metalloprotease (reprolysin type) with thrombospondin type
1 motif, 20; Collagen, type XI, alpha 1; Hyaluronan and proteoglycan link protein
1; Chondroitin beta1,4 N-acetylgalactosaminyltransferase ; Asporin (LRR class
1) ; Collagen, type III, alpha 1 (Ehlers-Danlos syndrome type IV, autosomal
dominant); Secreted phosphoprotein 1 (osteopontin, bone sialoprotein I, early
T-lymphocyte activation 1); Matrix Gla protein; Fibulin 5; collagen, type XIV,
alpha 1 (undulin); Tissue inhibitor of metalloproteinase 3 (Sorsby fundus
dystrophy, pseudoinflammatory); Collagen, type XXV, alpha 1; Cartilage
oligomeric matrix protein ; Collagen, type VI, alpha 1 ; Chondroadherin ;
Collagen, type XV, alpha 1 ; A disintegrin-like and metalloprotease (reprolysin
type) with thrombospondin type 1 motif, 16; Collagen, type IV, alpha 4 ; Dentin
matrix acidic phosphoprotein ; Collagen, type IV, alpha 1; Thrombospondin
repeat containing 1; Matrix metalloproteinase 16' (membrane-inserted);
Collagen, type I, alpha 2; Fibulin 1; Tectorin beta; Giycosylphosphatidylinositol
specific phospholipase D1; Upregulated in colorectal cancer gene 1).
Cytoskeleton: (Rlamin B, beta (actSn binding protein 278); Centrin, EF-hand
protein, 1; PERM domain containing 3; Bridging integrator 3; Parvin, gamma;
Rho guanine nucleotide exchange factor (GEF) 11; Tyrosine kinase 2; Kelchlike
4 (Drosophila); Spectrin, beta, erythrocytic (includes spherocytosis, clinical
type I); Arg/Abl-interacting protein ArgBP2; Advillin; Spectrin repeat containing,
nuclear envelope 1; Catenin (cadherin-associated protein), delta 1; Erythrocyte
membrane protein band 4.1 like 5; Catenin (cadherin-associated protein), alpha
2; Chemokine (C-C motif) ligand 3; Sarcoglycan, gamma (35kDa dystrophinassociated
glycoprotein); Nebulin; Thymosin, beta, identified in neuroblastoma
cells; 3-phosphoinositide dependent protein kinase-1; Wiskott-Aldrich syndrome
protein interacting protein; Dystonin; Huntingtin interacting protein 1; KIAA0316
gene product; Tropomodulin 4 (muscle); Deleted in liver cancer 1; Villin-like;
Syntrophin, beta 1 (dystrophin-associated protein A1, 59kDa, basic component
1); Protein kinase, cGMP-dependent, type I; Homo sapiens similar to keratin 8;
cytokeratin 8; keratin, type II cytoskeletal 8 (LOC345751), mRNA; Adducin 1
(alpha); Protein kinase C and casein kinase substrate in neurons 3; Dystonin;
Kell blood group; Filamin A interacting protein 1; Growth arrest-specific 2; ;
Chromosome 1 open reading frame 1; Stathrnin-like 2; Spectrin, alpha,
erythrocytic 1 (eiliptocytosis 2); FKSG44 gene; Kinesin family member 1C;
Tensin; Kaptin (actin binding protein); Neurofibromin 2 (bilateral acoustic
neuroma); Pleckstrin homology, Sec7 and coiled-coi! domains 2 (cytohesin-2);
Actin-related protein T1; Wiskott-Aldrich syndrome-like; Kelch-like 4
(Drosophila); Fascin homolog 1, actin-bundling protein (Strongylocentrotus
purpuratus); Amphiphysin (Stiff-Man syndrome with breast cancer 128kDa
autoantigen); Polycystic kidney disease 2-like 1; Ankyrin 2, neuronal; CDC42
binding protein kinase alpha (DMPK-like); Hypothetical protein FLJ36144;
Arg/Abl-interacting protein ArgBP2; Formin-like 3; Catenin (cadherin-associated
protein), beta 1, 88kDa; Profilin 2; Synaptopodin 2-like; Syntrophin, gamma 2;
Phospholipase D2; Engulfment and cell motility2 (ced-12 homolog, C. elegans);
Neurofilament, light polypeptide 68kDa; Dystonin; Actin-like 7B; Kinesin family
member 1C; PDZ and LIM domain 3; Adducin 2 (beta); obscurin, cytoskeletai
caimodulin and titin-interacting RhoGEF; Tubulin, beta polypeptide paralog;
Filamin A interacting protein 1; Talin 1; Homo sapiens similar to [Segment 1 of
2] Piccolo protein (Aczonin) (LOC375597); CDC42 effector protein (Rho
GTPase binding) 4; Syndecan 1; Filamin A, alpha (actin binding protein 280);
Profilin 2; Tensin like C1 domain containing phosphatase; Hypothetical protein
MGC33407; Rho family GTPase 1; Flavoprotein oxidoreductase MICAL2;
Ca2+-dependent secretion activator; Rabphilin 3A-like (without C2 domains);
Myosin XVA; Protein kinase, cGMP-dependent, type I; Myosin regulatory light
chain interacting protein; Kinesin family member 13B; Muscle RAS oncogene
homolog; Spectrin, beta, non-erythrocytic 1; TAO kinase 2; Filamin B, beta
(actin binding protein 278); Neurofibromin 2 (bilateral acoustic neuroma);
Catenin (cadherin-associated protein), alpha 3; obscurin, cytoskeletal
calmodulin and titin-interacting RhoGEF; Coronin, actin binding protein, 1A;
Erythrocyte membrane protein band 4.1-like 1; Spectrin, beta, non-erythrocytic
4; Thymosin, beta 4, Y-linked; Tektin 2 (testicular); Ras homolog gene family,
member J; Serine/threonine kinase with Dbl- and pleckstrin homology domains;
Dystrobrevin, beta; Actin, gamma 2, smooth muscle, enteric; Tara-like protein;
Caspase 8, apoptosis-related cysteine protease; Kelch repeat and BTB (POZ)
domain containing 10; Mucin 1, transmembrane; Microtubule-associated protein
tau; Tensin; Ras homolog gene family, member F (in filopodia); Adducin 1
(alpha); Actinin, alpha 4; Erythrocyte membrane protein band 4.1 (elliptocytosis
1, RH-linked); Bicaudal D homolog 2 (Drosophila); Ankyrin 3, node of Ranvier
(ankyrin G); Myosin VIIA (Usher syndrome 1B (autosomal recessive, severe));
Catenin (cadherin-associated protein), alpha 2; Homo sapiens similar to keratin
8, type II cytoskeletal - human (LOC285233); Fascin homolog 3, actin-bundling
protein, testicular; Ras homolog gene family, member J; Beaded filament
structural protein 2, phakinin; Desmin; Myosin X; Signal-induced proliferationassociated
gene 1; Scinderin; Coactosin-like 1 (Dictyostelium); Engulfment and
cell motility 2 (ced-12 homolog, C. elegans); Tubulin, beta 4; Ca2+-dependent
secretion activator; FERM domain containing 4A; Actin, alpha 1, skeletal
muscle; Talin 1; Caldesmon 1; Filamin-binding LIM protein-1; Microtubuleassociated
protein tau; Syntrophin, alpha 1 (dystrophln-associated protein A1,
59kDa, acidic component); Adducin 2 (beta); Filamin A interacting protein 1;
PDZ and LIM domain 3; Erythrocyte membrane protein band 4.1 like 4B; FYN
binding protein (FYB-120/130); Bridging integrator 3). Extracellular: (A
disintegrin-like and metalloprotease (reprolysin type) with thrombospondin type
1 motif, 20; SPARC-like 1 (mastQ, hevin); Serine {or cysteine) proteinase
inhibitor, clade G (C1 inhibitor), member 1, (angioedema, hereditary); Urocortin;
Chymotrypsin-Iike; Platelet-derived growth factor beta polypeptide (simian
sarcoma viral (v-sis) oncogene homolog); BMP-binding endothelial regulator
precursor protein; Complement factor H; Chorionic somatomammotropin
hormone-like 1; Chemokine (C-C motif) ligand 18 (pulmonary and activationregulated);
Fibronectin 1; Pregnancy specific beta-1-glycoprotein 3; A
disintegrin-like and metalloprotease (reprolysin type) with thrombospondin type
1 motif, 3; CocoaCrisp; Insulin-like 4 (placenta); Wingless-type MMTV
integration site family, member 11; Cartilage oligomeric matrix protein;
Transmembrane protease, serine 6; C-fos induced growth factor (vascular
endothelial growth factor D); Family with sequence similarity 12, member B
(epididymal); Protein phosphatase 1, regulatory subunit 9B, spinophilin;
Transcobalamin II; macrocytic anemia; Coagulation factor V (proaccelerin, labile
factor); Phosphoiipase A2, group IID; Tumor necrosis factor, alpha-induced
protein 6; Collagen, type XV, alpha 1; Hyaluronan and proteoglycan link protein
3; collagen, type XIV, alpha 1 (undulin) ; Interleukin 19; Protease Inhibitor 15;
Cholinergic receptor, nicotinic, beta polypeptide 1 (muscle); Lysyl oxidase-like
3; Insulin-like growth factor binding protein 5; Growth hormone 1; Casein beta;
NEL-like 2 (chicken); I factor (complement); Chemokine (C-C motif) ligand 23 ;
Interferon, alpha 2; Matrix metalloproteinase 16 (membrane-inserted); Matrix
metalloproteinase 12 (macrophage elastase); Glypican 5; Pregnancy specific
beta-1-glycoprotein 3; Fibroblast growth factor 6; Gremlin 1 homolog, cysteine
knot superfamily (Xenopus laevis); Protein S (alpha); Chondroitin beta1,4 Nacetylgalactosaminyltransferase;
Glycosylphosphatidylinositol specific
phospholipase D1; Fibroblast growth factor 1 (acidic); Spondin 1, extracellular
matrix protein; Bone morphogenetic protein 1; Surfactant, pulmonaryassociated
protein B; Dentin matrix acidic phosphoprotein ; Lipoprotein, Lp(a);
Mucin 1, transmembrane; Mannan-binding lectin serine protease 1 (C4/C2
activating component of Ra-reactive factor); Meprin A, beta; Secretoglobin,
family 1D, member 1; Asporin (LRR class 1); Chemokine (C-C motif) ligand
Cytoklne-like protein C17; Insulin-like 5; Meprin A, alpha (PABA peptide
hydrolase) ; Scrapie responsive protein 1; Fibroblast growth factor 18;
Chemokine (C-X-C motif) ligand 9; Inhibin, beta B (activin AB beta polypeptide);
Fibroblast growth factor 8 (androgen-induced); Granulysin; Cocaine- and
amphetamine-regulated transcript; Collagen, type I, alpha 2; Chemokine (C-C
motif) ligand 17; Chemokine (C-C motif) ligand 23 ; Sparc/osteonectin, cwcv
and kazal-like domains proteoglycan (testican) 3; Gamma-aminobutyric acid
(GABA) A receptor, beta 3; Defensin, alpha 4, corticostatin; Leucine rich repeat
neuronal 3; Glypican 6; Mitogen-activated protein kinase kinase 2; Coagulation
factor XI (plasma thromboplastin antecedent); Chemokine (C-C motif) ligand
Dystonin; Frizzled-related protein; Coagulation factor XIII, A1 polypeptide;
Insulin-like growth factor 1 (somatomedin C); Hypothetical protein MGC45438;
Sperm associated antigen 11; Insulin-like growth factor 1 (somatomedin C);
Periostin, osteoblast specific factor; Alpha-2-macroglobuIin; Gammaaminobutyric
acid (GABA) A receptor, alpha 5; Serine (or cysteine) proteinase
inhibitor, clade A (alpha-1 antiproteinase, antitrypsin), member 3; Silver
homolog (mouse); Frizzled-related protein; Chondroadherin; Chondroitin
betal ,4 N-acetylgalactosaminyltransferase; 5-hydroxytryptamine (serotonin)
receptor 3, family member C; Collagen, type VI, alpha 2; Toll-like receptor 9;
Amelpgenin, Y-linked; Vascular endothelial growth factor B; Radial spokeheadiike
1; Fms-related tyrosine kinase 1 (vascular endothelial growth
factor/vascular permeability factor receptor); Protease inhibitor 16; Interleukin
Clusterin (complement lysis inhibitor, SP-40,40, sulfated glycoprotein 2,
testosterone-repressed prostate message 2, apolipoprotein J); Follicle
stimulating hormone, beta polypeptide; A disintegrin-Iike and metalloprotease
(reprolysin type) with thrombospondin type 1 motif, 16; Lysozyme (renal
amyloidosis); radical fringe homolog (Drosophila); Insulin-like growth factor
binding protein 5; Taxilin; Apolipoprotein A-V; Platelet derived growth factor C;
Chemokine (C-C motif) ligand 3-like 1; Fibroblast growth factor 16; Collagen,
type VI, alpha 2; Serine (or cysteine) proteinase inhibitor, clade C
(antithrombin), member 1; Chemokine (C-C motif) ligand 11; Collagen, type IV,
alpha 4; Bruton agammaglobulinemia tyrosine kinase; Insulin-like growth factor
2 (somatomedin A); Kazal-type serine protease inhibitor domain 1; Fibrinogen,
A alpha polypeptide; Chemokine (C-C motif) ligand 1; Inhibin, beta E; Sex
hormone-binding globulin; Collagen, type IV, alpha 1; Lecithin-cholesterol
acyltransferase; Cysteine-rich secretory protein 2; Hyaluronan and proteoglycan
link protein 1; Natriuretic peptide precursor C; Rlbonuclease, RNase A family,
k6; Fibroblast growth factor 14; ADAMTS-like 2; Collagen, type IV, alpha 3
(Goodpasture antigen); Angiopoietin 2; Apolipoprotein L, 3; Chemokine (C-X-C
motif) ligand 12 (stromal cell-derived factor 1); Hyaluronan binding protein 2;
Coagulation factor VI! (serum prothrombin conversion accelerator); collagen,
type XiV, alpha 1 (undulin) ; Oviductal glycoprotein 1, 120kDa (rnucin 9,
oviductin); Matrilin 1, cartilage matrix protein; mucin 5, subtypes A and C,
tracheobronchial/gastric; Tumor necrosis factor receptor superfamily, member
11b (osteoprotegerin); Transglutaminase 2 (C polypeptide, protein-glutaminegamma-
glutamyltransferase); Keratocan; Collagen, type V, alpha 3; WAP fourdisulfide
core domain 2; Chemokine (C-X3-C motif) Hgand 1; Serine (or
cysteine) proteinase inhibitor, clade D (heparin cofactor), member 1; Secretory
protein LOC348174; Coagulation factor X; Interleukin 16 (lymphocyte
chemoattractant factor); Pancreatic Hpase-related protein 2; HtrA serine
peptidase 3; Glycine receptor, alpha 3; CDS antigen-like (scavenger receptor
cysteine rich family); Hypothetical protein MGC39497; Coagulation factor VIII,
procoagulant component (hemophilia A); Dermatopontin; Noggin; Secreted
LY6/PLAUR domain containing 1; ADAMTS-like 1; Alpha-1-B glycoprotein;
Chromosome 20 open reading frame 175; Wingless-type MMTV integration site
family, member 8B; Fibulin 1; Fibulin 5; Cathepsin S; Nidogen (enactin);
Chemokine (C-C motif) ligand 26; Endothelial cell-specific molecule 1; Chitinase
3-like 1 (cartilage glycoprotein-39); Gamma-aminobutyric acid (GABA) A
receptor, beta 1; Secretoglobin, family 1D, member 2; Mannan-binding lectin
serine protease 1 (C4/C2 activating component of Ra-reactive factor);
ADAMTS-like 1; Sema domain, immunoglobulin domain (Ig), and GPI
membrane anchor, (semaphorin) 7A; A disintegrin-like and metalloprotease
(reprolysin type) with thrombospondin type 1 motif, 15; Proprotein convertase
subtilisin/kexin type 2 ; Insulin-like growth factor 1 (somatomedin C);
Retinoschisis (X-linked, juvenile) 1; A dislntegrin-like and metalloprotease
(reprolysin type) with thrombospondin type 1 motif, 16; Chemokine (C motif)
ligand 2; Fibroblast growth factor 5; Sperm associated antigen 11; Microfibrillarassociated
protein 4; Poliovirus receptor; Extracellular signal-regulated kinase
8; Transmembrane protease, serine 6; Protein kinase C, alpha; Chitinase 3-like
2; Interleukin 9; Apolipoprotein L, 6; Surfactant, pulmonary-associated protein
A1; Collagen, type VI, alpha 1; Apolipoprotein L, 6; Hypothetical protein
FLJ13710; Carboxypeptidase B2 (plasma, carboxypeptidase U) ;
37
Bactericidal/permeability-increasing protein-like 2; Fibroblast growth factor 5;
Secreted phosphoprotein 1 (osteopontin, bone sialoprotein I, early Tlymphocyte
activation 1); HtrA serine peptidase 3; Deleted in liver cancer 1;
Endothelial cell-specific molecule 1; Von Willebrand factor; A disintegrin-Iike
and metalloprotease (reprolysin type) with thrombospondin type 1 motif, 5
(aggrecanase-2); Sema domain, immunoglobulin domain (Ig), short basic
domain, secreted, (semaphorin) 3A; Chemokine (OX-C motif) ligand 12
(stromal cell-derived factor 1); Statherin; Extracellular signal-regulated kinase 8;
Tissue inhibitor of metalloproteinase 3 (Sorsby fundus dystrophy,
pseudoinflammatory); Platelet factor 4 (chemokine (C-X-C motif) ligand 4);
Surfactant, pulmonary-associated protein D; Complement factor H; Delta-like 1
homolog (Drosophila); WAP four-disulfide core domain 1; Insulin-like growth
factor binding protein, acid labile subunit; Breast cancer 2, early onset; Pre-B
lymphocyte gene 1; Corticotropin releasing hormone; Hypothetical protein
DKFZp434B044; Prolactin-induced protein; RAS guanyl releasing protein 4;
Progastricsin (pepsinogen C); Sema domain, immunoglobulin domain (Ig), short
basic domain, secreted, (semaphorin) 3F; Upregulated in colorectal cancer
gene 1; Proteoglycan 4; Cholinergic receptor, nicotinic, delta polypeptide;
Cartilage oligomeric matrix protein; ABO blood group (transferase A, alpha
N-acetylgalactosaminyltransferase; transferase ~B, alpha 1-3-
galactosyltransferase); Interleukin 12A (natural killer cell stimulatory factor 1,
cytotoxic lymphocyte maturation factor 1, p35); Fibroblast growth factor 7
(keratinocyte growth factor); Kin of IRRE like 3 (Drosophila); Cholinergic
receptor, nicotinic, alpha polypeptide 2 (neuronal); Palate, lung and nasal
epithelium carcinoma associated; Collagen, type XV, alpha 1; Pleiotrophin
(heparin binding growth factor 8, neurite growth-promoting factor 1J;
Angiopoietin-like 2; Norrie disease (pseudoglioma); Chemokine (C-C motif)
ligand 3; Chitinase 3-like 1 (cartilage glycoprotein-39); Inter-alpha (globulin)
inhibitor H3; Amelogenin (amelogenesis imperfecta 1, X-linked); Epidermal
growth factor (beta-urogastrone); Fibroblast growth factor 13; Wingless-type
MMTV integration site family, member 7B; Cholinergic receptor, nicotinic,
gamma polypeptide; Pregnancy specific beta-1-glycoprotein 6; Matrix
metalloproteinase 14 (membrane-inserted); Chemokine (C-C motif) ligand 26;
Interferon, alpha 6; Tachykinin, precursor 1 (substance K, substance P,
38
neurokinin 1, neurokinin 2, neuromedin L, neurokinin alpha, neuropeptide K,
neuropeptide gamma); Secreted frizzled-related protein 5; Hyaluronan and
proteoglycan link protein 4; Complement component 4B; Matrix
metalloproteinase 16 (membrane-inserted); Fibroblast growth factor 7
(keratinocyte growth factor); Apolipoprotein Oil; Chloride channel, calcium
activated, family member 3; Tetranectin (plasminogen binding protein);
Collagen, type III, alpha 1 (Ehlers-Danlos syndrome type IV, autosomal
dominant); KIAA0556 protein; Chemokine (C-C motif) ligand 4; Hemopexin;
Inter-alpha (globulin) inhibitor H1; Relaxin 1; Matrix Gla protein; A disintegrinlike
and metalloprotease (reprolysin type) with thrombospondin type 1 motif, 2;
Interferon (alpha, beta and omega) receptor 2; Acid phosphatase, prostate;
Guanine nucleotide binding protein (G protein), gamma 8; Matrix
metalloproteinase 23B; Meprin A, alpha (PABA peptide hydrolase);
Hyaluronoglucosaminidase 1; Angiotensinogen (serine (or cysteine) proteinase
inhibitor, clade A (alpha-1 antiproteinase, antitrypsin), member 8); Cartilage
intermediate layer protein, nucleotide pyrophosphohydrolase; Purinergic
receptor P2X ligand-gated ion channel, 7; Glypican 3; Tectorin beta; Interferon,
alpha 5; Lipocalin 7; Platelet factor 4 variant 1; Nucleobindin 1; Collagen, type
XI, alpha 1; Gastric inhibitory polypeptide; Thrombospondin repeat containing 1;
5-hydroxytryptamine (serotonin) receptor 3 family member D; Collagen, type
XXV, alpha 1; Growth differentiation factor 9; Hypothetical protein
DKFZp434B044; Endothelin 3; Chemokine (C motif) ligand 2; Prokineticin 2;
Tumor necrosis factor receptor superfamily, member 11b (osteoprotegerin);
Tissue inhibitor of metalloproteinase 2; Dystonin; Chromogranin B
(secretogranin 1); Hyaluronan and proteoglycan link protein 2; Leucine rich
repeat neuronal 3; Lumican; Matrilin 1, cartilage matrix protein ; Phospholipase
A2, group IIA (platelets, synovial fluid); Carboxylesterase 1
(monocyte/macrophage serine esterase 1); Sparc/osteonectin, cwcvand kazallike
domains proteoglycan (testican); Dickkopf homolog 2 (Xenopus laevis);
Gamma-aminobutyric acid (GABA) A receptor, alpha 3; Pregnancy specific
beta-1-glycoprotein 11; Insulin-like growth factor binding protein 1; Defensin,
beta 106; Interleukin 17F; Ligand-gated ion channel subunit; Phospholipase A2
receptor 1,180kDa; I factor (complement); Dystonin; LAG1 longevity assurance
homolog 1 (S. cerevlsiae); Prolactin; Testis expressed sequence 264; Sema
domain, immunoglobulin domain (Ig), short basic domain, secreted,
(semaphorin) 3D; secreted frizzled-related protein 2; secreted frizzied-related
protein 4).
[0091]There are groups of genes present only in UCEC. These genes
are related to the following: Horneostasis (Albumin; Calcium-sensing receptor,
Aquaporin 9; Lactotransferrin. Morphogenesis: Homeo box HB9; Epithelial Vlike
antigen 1). Embryonic Development (RelaxSn 2; Carcinoembryonic antigenrelated
cell adhesion molecule 8; Indoleamine-pyrrole 2,3 dioxygenase; EPH
receptor A3; Thyrotrophic embryonic factor; Pregnancy specific beta-1-
glycoprotein 1; Laminin, alpha 3), the Extracellular Space (Surfactant,
pulmonary-associated protein A1; Pregnancy specific beta-1-glycoprotein 1;
Lactotransferrin; TGF-alpha; Albumin; FGF-23; S100 calcium binding protein A9
(calgranulin B)), the Extracellular Matrix (Laminin, beta 4; Laminin, alpha 3;
Zona pellucida glycoprotein 4. Structural Molecule Activity: Chromosome 21
open reading frame 29; Laminin, alpha 3; Microtubule-associated. protein 2;
Laminin, beta 4; Keratin 6B; Ladinin 1; Keratin 6A; Occludin; Loricrin;
Erythrocyte membrane protein band 4.1 (eiliptocytosis 1, RH-linked); Crystallin,
beta A2; eye lens structural protein; Contactin associated protein-like 4; Ciaudin
19; Hypothetical protein LOC144501; Keratin 6E; Keratin 6L; Lens intrinsic
membrane protein 2, 19kDa), the Cytoskeleton (Microtubule-associated protein
2; Erythrocyte membrane protein band 4.1 like 5; Homo sapiens trichohyalin
(THH); Keratin 6B; Keratin 6A; Epithelial V-like antigen 1; Hook homoiog 1
(Drosophila); Loricrin; Erythrocyte membrane protein band 4.1 (eiliptocytosis 1,
RH-linked); Tropomodulin 1; MAP/microtubule affinity-regulating kinase 1;
Keratin 6E; Actin binding L1M protein family, member 2), Cell Adhesion
Molecules (Cadherin 19, type 2; Myetoid/lymphoid or mixed-lineage leukemia;
Chromosome 21 open reading frame 29; Kin of IRRE like 2; Laminin, alpha 3;
Sialoadhesin; CD84 antigen (leukocyte antigen); Lectin, galactoside-binding,
soluble, 2 (galectin 2); Epithelial V-like antigen 1; CD96 antigen;
Tubulointerstitial nephritis antigen; Carcinoembryonic antigen-related cell
adhesion molecule 8; IL-18; Immunoglobulin superfamily, member 1; Integrin,
beta 8; Ornithine arbamoyitransferase; Integrin, beta 6; Contactin associated
protein-like 4; Collagen, type XVII, alpha 1; Cadherin-like 26; Mucin and
cadherin-Iike), Cell Differentiation proteins (Protein tyrosine phosphatase,
receptor-type, Z polypeptide 1; Laminin, alpha 3; CD84 antigen (leukocyte
antigen); EDRF2; Homo sapiens erythroid differentiation-related factor 2; Tumor
protein p73-like; NB4 apoptosis/differentiation related protein; Homo sapiens
PNAS-133; Similar to seven in absentia 2; Interleukin 24; Keratin 6B; Keratin
6A; Dehydrogenase/reductase (SDR family) member 9; Gap junction protein,
beta 5 (connexin 31.1); Iroquois homeobox protein 4; Ventral anterior
homeobox 2; Chemokine (C-X-C motif) ligand 10;Tumor necrosis factor
receptor superfamily, member 17; Calcium channel, voltage-dependent, beta 2
subunit; Parkinson disease (autosomal recessive, juvenile) 2, parkin; Kallikrein
7 (chymotryptic, stratum corneum); Glial cells missing homolog 2; AP-2
alpha;Protein tyrosine phosphatase, receptor-type, Z polypeptide 1; Troponln
T1; Sciellin; Glucosaminyl (N-acetyl) transferase 2, l-branching enzyme;
Collagen, type XVII, alpha 1; Suppressor of cytokine signaling 2; Distal-less
homeo box 1; Zygote arrest 1; Interleukin 20; Growth differentiation factor 3;
FGF-23; Wingless-type MMTV integration site family, member 8A. Extracellular.
Chromosome 21 open reading frame 29; Laminin, alpha 3; Laminin, beta 4;
Interleukin 24; Pregnancy specific beta-1-glycoprotein 1; Chemokine (C-X-C
motif) ligand 11; Surfactant, pulmonary-associated protein A1;
Prepronociceptin; 5-hydroxytryptamine (serotonin) receptor 3B;
Carcinoembryonic antigen-related cell adhesion molecule 8; Chemokine (C-X-C
motif) Hgand 10; IL-18 (interferon-gamma-inducing factor); Lactotransferrin;
Albumin; Fas ligand (TNF superfamily, member 6); Cholinergic receptor,
nicotinic, beta polypeptide 4; Cathelicidin antimicrobial peptide; Airway trypsinlike
protease;S100 calcium binding protein A9 (calgranulin B); TGF-alpha;
Kallikrein 10; Serine protease inhibitor, Kunitztype 1; WNT1 inducible signaling
pathway protein 3; Relaxin 2; Interferon, kappa; Defensin, beta 103A; IL-20;
Zona pellucida glycoprotein 4; Growth differentiation factor 3; FGF-23;
Wingless-type MMTV integration site family, member 8A; Complement factor Hrelated
5), Developmental proteins (EPH receptor A3; MIMA (never in mitosis
gene a)-related kinase 2; Zinc finger protein 282; TANK-binding kinase 1;
MRE11 meiotic recombination 11 homolog A; E2F transcription factor 2; Protein
tyrosine phosphatase, receptor-type, Z polypeptide 1; Homo sapiens clone
161455 breast expressed mRNA from chromosome X Laminin, alpha 3; v-myb
myeloblastosis viral oncogene homolog (avian)-like 1; Regulator of G-protein
signalling 11; Microtubule-associated protein 2; Transmembrane protein 16A;
Adenomatosis polyposis coli 2; Homeo box HB9; Centromere protein F,
350/400ka (mitosin); CD84 antigen (leukocyte antigen); EDRF2; Homo sapiens
erythroid differentiation-related factor 2; Tumor protein p73-Iike; NB4
apoptosis/differentiation related protein; Homo sapiens PNAS-133; Forkhead
box P2; Homo sapiens gastric-associated differentially-expressed protein
YA61P (YA61); Tenascin N; Chromosome 6 open reading frame 49; Zinc finger
protein 462; Zinc finger protein 71 (Cos26); SRY (sex determining region Y)-box
7; Triggering receptor expressed on myeloid cells-like 4; Interteukin 24;
Pregnancy specific beta-1-glycoprotein 1;Chondroitin sulfate proteoglycan 5
(neuroglycan C); Keratin 6B; Keratin 6A; Dehydrogenase/reductase (SDR
family) member 9; Epithelial V-like antigen 1; Gap junction protein, beta 5
(connexin 31.1); G protein-coupled receptor 51; 'Interferon regulatory factor 6;
Neurotrophin 5 (neurotrophin 4/5); CD96 antigen; Iroquois homeobox protein
Interieukin 1 receptor-like 1; G-2 and S-phase expressed 1; Nuclear receptor
subfamily 2, group E, member 3; Ventral anterior homeobox 2; Zinc finger
protein 215; DMA segment on chromosome 4 (unique) 234 expressed
sequence; Carcinoembryonic antigen-related cell adhesion molecule 8;
Chemokine (C-X-C motif) ligand 10; IL-18; Indoleamine-pyrrole 2,3
dioxygenase; Albumin; Calcium-sensing receptor (hypocalciuric hypercalcemia
1, severe neonatal hyperparathyroidism); Fas ligand (TNF superfamily, member
6); TNFR superfamily, member 17; Calcium channel, voltage-dependent, beta 2
subunit; Parkinson disease (autosomal recessive, juvenile) 2, parkin; Kallikrein
7 (chymotryptic, stratum corneum); Glial cells missing homolog 2; TGF-alpha;
Thyrotrophic embryonic factor; AP-2 .alpha (activating enhancer binding protein
2 alpha); Kallikrein 10; Regulator of G-protein signalling 7; Protein tyrosine
phosphatase, receptor-type, Z polypeptide 1; Serine protease inhibitor, Kunitz
type 1; WNT1 inducible signaling pathway protein 3; Zic family member 3
heterotaxy 1 (odd-paired homolog, Drosophila); TTK protein kinase; Troponin
T1, skeletal, slow; Sciellin; TGFB-fnduced factor 2-like, X-linked; Kallikrein 8
(neuropsin/ovasin); Glucosaminyl (N-acetyl) Iransferase 2, l-branching enzyme;
Ankyrin repeat domain 30A; Relaxin 2; Collagen, type XVII, alpha 1; Gene
differentially expressed in prostate; Phosphatase and actin regulator 3;
Suppressor of cytoklne signaling 2; Nuclear receptor subfamily 4, group A,
member 3; Angiotensin I converting enzyme (peptidyl-dipeptidase A) 1;
Hypothetical protein MGC17986; Distal-less homeo box 1; LAG1 longevity
assurance homolog 3 (S. cerevisiae); Zygote arrest 1; Interferon, kappa; IL-20;
ICEBERG caspase-1 inhibitor; Growth differentiation factor 3; FGF-23; Test's
expressed sequence 15; Wingless-type MMTV integration site family, member
8A; SRY (sex determining region Y)-box 7; Camitine deficiency-associated,
expressed in ventricle 1; Prokineticin 1; CAMP responsive element binding
protein 3-like 3; Caspase recruitment domain family, member 15; FLJ23311
protein).
Example 6: Direct differentiation of umbilical cord epithelial stem cells
(UCEC) into skin epidermal keratinocytes
[0092] For differentiation into skin epidermal keratinocytes, umbilical
cord epithelial stem cells, UCEC cells, were cultured according to a standard
protocol for the cultivation of keratinocytes. Cell isolation techniques were as
described above. UCEC were then cultured in serum-free keratinocyte growth
media, KGM, KGM-2 (Cambrex), EpiLife (Cascade Biologies) or in Green's
medium in the presence of irradiated or Mytomycin-C treated 3T3 mouse
embryonic feeder layer at 37°C, 5% COz). UCEC cell morphology thus
differentiated resembled human epidermal keratinocytes. Epithelial cells have
similar morphology under light microscope and can be easily turned into
fibroblasts using conventional and commercially available media (cf., Figure 2).
[0093]lmmunofluorescent analysis shows that the cultivated UCEC also
express epidermal keratinocyte molecular markers such as keratins,
desmosome, hemidesmosome and basement membrane components (see also
Fig. 10 that shows that UCEC are qualified to be epithelial cells in general by
expressing a variety of these epithelial cell markers). Accordingly, these results
show that umbilical cord epithelial progenitor/stem cells of the present invention
can be differentiated into skin cells such as epidermal keratinocytes which can
be used for wound healing and have great potential for the development of
cultured skin equivalents.
Example 7: Expansion of umbilical cord epithelial and mesenchymal stem
cells using repetitive tissue explants of umbilical cord lining membrane
tissues
[0094] Umbilical cord epithelial and mesenchymal stem cells of the
invention were expanded using repetitive explants of umbilical cord amniotic
membrane tissue as follows. Briefly, at day 1 of process, tissue explants were
plated onto tissue culture dishes in growth media (DMEM/10%FCS, EpiLife,
KGM, KGM-2 or M171) at 37°C, 5%CO2; media was changed every 2 or 3
days. Cell outgrowths started and continued migrating from the explants for 7
days. After that, tissue explants were transferred to other dishes to allow further
cell outgrowth. This process was continued until the explants had diminished in
size, preventing further explantation. In this connection it is noted that the
explants progressively shrink in size until they are too small for further tissue
explant since during the process of cells outgrowing and migrating from tissue
explants, the cells produce proteases to digest and break down tissue. Fig. 16
schematically illustrates the rapid and robust expansion process of umbilical
cord epithelial and mesenchymal stem cells achieved using this protocol. Thus,
this study demonstrates the high yield of UCMC and UMEC cells can be
obtained from this source, further reflecting the high viability and pro-growth
characteristics oft these cells in comparison with other sources of cells as bonemarrow
or adipose-derived stem cells. In addition, being a solid tissue, the
successful repetitive explant technique used herein demonstrates that the cells
of the invention can be uniformly extracted from the entire tissue instead of only
certain portions. This allows the maximum number of cells that can be derived
at a low passage instead of passing the cells through many generations causing
deterioration of cells.
Example 8: Direct differentiation of umbilical cord mesenchymal cells
(UCMC) into skin dermal fibroblasts
[0095J For differentiation into skin dermal fibroblasts, umbilical cord
mesenchymal stem cells, UCMC cells were cultured according to a standard
protocol for the cultivation of fibroblasts. Cell isolation techniques were as
described above in Example 6. UCMC were then cultured in DMEM or
commercially available fibroblast growth media (FGM). UCMC cell morphology
thus differentiated resembled human dermal fibroblasts. Mesenchymal cells
have similar morphology under light microscope and can be easily turned into
fibroblasts using conventional and commercially available media (cf.,



What is claimed is:
1. A method for isolating stem/progenitor cells from the amniotic membrane
of umbilical cord, the method comprising:
(a) separating the amniotic membrane from the other components of the
umbilical cord in vitro;
(b) culturing the amniotic membrane tissue obtained in step (a) under
conditions allowing cell proliferation; and
(c) isolating the stem/progenitor cells.
2. The method of claim 1, further comprising:
(a") separating the cells of the amniotic membrane tissue before
cultivation by a method selected from the group consisting of
enzymatic separation and direct tissue explant.
3. The method of claim 1 or 2, wherein the stem/progenitor cells have
embryonic stem cell-like properties.
4. The method of claim any of claims 1 to 3, wherein the stem/progenitor cells
are epithelial and/or mesenchymal stem/progenitor cells.
5. The method of any of claims 1 to 4, further comprising:
(d) culturing the stem/progenitor cells under conditions allowing the cells
to undergo clonal expansion.
6. The method of claim 5, further comprising:
(e) culturing the stem/progenitor cells under conditions allowing the
differentiation of said cells into epithelial cells and/or mesenchymal
cells; and
(f) isolating the differentiated cells.
7. The method of claim 6, wherein the epithelial cells are selected from the
group consisting of: skin epithelial cells, hair follicular cells, cornea epithelial
cells, conjunctiva! epithelial ceils, retinal epithelial cells, liver epithelial cells,
kidney epithelial cells, pancreatic epithelial cells, oesophageal epithelial
cells, small intestinal epithelial cells, large intestinal epithelial cells, lung
epithelial cells, airway epithelial cells, bladder epithelial cells and uterine
epithelial cells.
8. The method of claim 6, wherein the mesenchymal cells are selected from the
group consisting of: skin fibroblasts, chondrocytes, osteoblasts, tenocytes,
ligament fibroblasts, cardiomyocytes, smooth muscle cells, skeletal muscle
cells, adipocytes, cells derived from endocrine glands, neurectodermal cells,
and all varieties and derivatives of neurectodermal cells.
9. The method of any of claims 1 to 8, further comprising
(g) preserving the isolated stem/progenitor cells for further use.
10.The method of claim 9, wherein preserving is carried out by using cryopreservation.
11. A stem/progenitor cell isolated from the amniotic membrane of umbilical
cord by means of the method of any of claims 1 to 10.
12. A cell bank comprising a stem/progenitor cell isolated by means of the
method of any of claims 1 to 10.
13. A pharmaceutical composition comprising a stem/progenitor cell or a cellular
extract thereof isolated from the amniotic membrane of umbilical cord by
means of the method of any of claims 1 to 10.
14. The pharmaceutical composition of claim 13, wherein the pharmaceutical
composition is adapted for systemic or topical application.
15.The pharmaceutical composition of claim 13 or 14, wherein the
pharmaceutical composition adapted for topical application is selected from
the group consisting of an ointment, a cream, and a lotion.
16. A method of treating a subject having a disorder comprising administering to
the subject an effective amount of a stem/progenitor cell isolated by means
of the method of any of claims 1 to 10.
17. The method of treating of claim 16, wherein the disorder is selected from the
group consisting of neoplastic disease, skin disease, visceral endocrine
deficiency, and a neural disorder.
18. The method of claim 17, wherein the tissue disorder is a congenital or an
acquired tissue deficiency.
19. The method of claim 17, wherein the visceral endocrine deficiency is
selected from the group consisting of insulin deficiency, Diabetes mellitus
associated with insulin deficiency, testosterone deficiency, anemia,
hypoglycemia, hyperglycemia, pancreatic deficiency, adrenal deficiency, and
thyroid abnormality.
20.The method of claim 17, wherein the neural disorder is Alzheimer's disease,
Parkinson's disease, Jacob Kreutzfeld's disease, Lou Gehrig's disease, and
Huntington's disease.
21. The method of claim 17, wherein the skin disease is accelerated aging or a
' wound.
22. The method of claim 17, wherein the neoplastic disease is cancer.
23. The method of claim 22, wherein the cancer is a selected from the group
consisting of squamous cell carcinoma, breast ductal and lobular carcinoma,
hepatocellular carcinoma, nasopharyngeal carcinoma, lung cancer, bone
cancer, pancreatic cancer, skin cancer, cancer of the head or neck,
cutaneous or intraocular malignant melanoma, uterine cancer, ovarian
cancer, rectal cancer, cancer of the anal region, stomach cancer, colon
cancer, breast cancer, testicular cancer, uterine cancer, carcinoma of the
fallopian tubes, carcinoma of the endometrium, carcinoma of the cervix,
carcinoma of the vagina, carcinoma of the vulva, Hodgkin's Disease, non-
Hodgkin's lymphoma, cancer of the esophagus, cancer of the small intestine,
cancer of the endocrine system, cancer of the thyroid gland, cancer of the
parathyroid gland, cancer of the adrenal gland, sarcoma of soft tissue,
cancer of the urethra, cancer of the penis, prostate cancer, chronic or acute
leukemias, solid tumors of childhood, lymphocytic lymphoma, cancer of the
bladder, cancer of the kidney or ureter, renal cell carcinoma, carcinoma of
the renal pelvis, neoplasm of the central nervous system (CNS), primary
CMS lymphoma, tumor angiogenesis, spinal axis tumor, brain stem glioma,
pituitary adenoma, Kaposi's sarcoma, epidermoid cancer or any combination
of such cancers, including disseminated (metastasizing) forms thereof.
24. The use of a stem/progenitor cell as defined in claim 11 for the production of
a biological molecule.
25. The use of claim 24, wherein the biological molecule is selected from the
group consisting of a protein, a peptide, a small organic molecule, an
oligosaccharide, a polysaccharide, a proteoglycan and a lipid.
26. The use of a stem/progenitor cell as defined in claim 11 as feeder layer in
the cultivation of mammalian cells.
27. A method of cultivating stem/progenitors cells as defined in claim 11,
comprising:
Obtaining a tissue explantfrom the amniotic membrane of umbilical cord;
Cultivating the tissue explant in suitable cultivation media and cultivation
conditions over a suitable period of time.
28. The method of daim 27, further comprising exposing the tissue explant to
fresh cultivation media and continuing the cultivation under suitable
conditions over a suitable period of time.
29. The use of serum-free cell culture media to culture mesenchymal stem cells
obtained by a method of any of claims 1 to 10.

Documents:

1492-delnp-2007-1-Claims-(15-05-2013).pdf

1492-delnp-2007-1-Correspondence Others-(15-05-2013).pdf

1492-delnp-2007-Abstract-(23-12-2013).pdf

1492-delnp-2007-abstract.pdf

1492-delnp-2007-Claims-(03-12-2013).pdf

1492-delnp-2007-Claims-(21-11-2013).pdf

1492-delnp-2007-Claims-(23-12-2013).pdf

1492-delnp-2007-claims.pdf

1492-delnp-2007-Correspondence Others-(15-05-2013).pdf

1492-delnp-2007-Correspondence Others-(03-12-2013).pdf

1492-delnp-2007-Correspondence Others-(08-01-2014).pdf

1492-delnp-2007-Correspondence Others-(09-09-2013).pdf

1492-delnp-2007-Correspondence Others-(09-11-2012).pdf

1492-delnp-2007-Correspondence Others-(12-11-2013).pdf

1492-delnp-2007-Correspondence Others-(13-03-2013).pdf

1492-delnp-2007-Correspondence Others-(16-06-2011).pdf

1492-delnp-2007-Correspondence Others-(23-12-2013).pdf

1492-delnp-2007-Correspondence Others-(31-07-2013).pdf

1492-DELNP-2007-Correspondence-Others-(01-10-2012).pdf

1492-delnp-2007-Correspondence-Others-(04-07-2013).pdf

1492-delnp-2007-Correspondence-Others-(07-06-2013).pdf

1492-delnp-2007-Correspondence-Others-(07-08-2013).pdf

1492-delnp-2007-Correspondence-Others-(18-02-2013).pdf

1492-delnp-2007-Correspondence-Others-(21-11-2013).pdf

1492-delnp-2007-Correspondence-Others.pdf

1492-delnp-2007-description (complete).pdf

1492-delnp-2007-drawings.pdf

1492-DELNP-2007-Form-1.pdf

1492-delnp-2007-Form-2-(23-12-2013).pdf

1492-delnp-2007-form-2.pdf

1492-DELNP-2007-Form-26.pdf

1492-delnp-2007-Form-3-(08-01-2014).pdf

1492-delnp-2007-Form-3-(09-11-2012).pdf

1492-delnp-2007-Form-3-(12-11-2013).pdf

1492-delnp-2007-Form-3-(13-03-2013).pdf

1492-delnp-2007-Form-3-(16-06-2011).pdf

1492-delnp-2007-Form-3-(18-02-2013).pdf

1492-delnp-2007-Form-3-(31-07-2013).pdf

1492-DELNP-2007-Form-3.pdf

1492-delnp-2007-form-5.pdf

1492-delnp-2007-GPA-(03-12-2013).pdf

1492-delnp-2007-pct-101.pdf

1492-delnp-2007-pct-105.pdf

1492-delnp-2007-pct-202.pdf

1492-delnp-2007-pct-210.pdf

1492-delnp-2007-pct-220.pdf

1492-DELNP-2007-PCT-237.pdf

1492-delnp-2007-pct-301.pdf

1492-delnp-2007-pct-308.pdf

1492-DELNP-2007-PCT-326.pdf

1492-DELNP-2007-PCT-373.pdf

1492-delnp-2007-pct-notification.pdf

1492-delnp-2007-Petition-137-(15-05-2013).pdf


Patent Number 258562
Indian Patent Application Number 1492/DELNP/2007
PG Journal Number 04/2014
Publication Date 24-Jan-2014
Grant Date 21-Jan-2014
Date of Filing 23-Feb-2007
Name of Patentee CELLRESEARCH CORPORATION PTE LTD
Applicant Address #03-09 CLINICAL RESEARCH CENTRE, BLOCK MD-11, 10 MEDICAL DRIVE, SINGAPORE 117597, SINGAPORE.
Inventors:
# Inventor's Name Inventor's Address
1 PHAN, TOAN-THANG #03-09 CLINICAL RESEARCH CENTRE, BLOCK MD-11, 10 MEDICAL DRIVE, SINGAPORE 117597, SINGAPORE.
2 LIM, IVOR JIUN 5 LOWER KENT RIDGE ROAD, MAIN BUILDING 1, LEVEL 2, SINGAPORE 119074, SINGAPORE.
PCT International Classification Number C12N 5/06
PCT International Application Number PCT/SG2005/000174
PCT International Filing date 2005-06-03
PCT Conventions:
# PCT Application Number Date of Convention Priority Country
1 60/632,209 2004-12-01 U.S.A.
2 60/602,208 2004-08-16 U.S.A.