  
- 积分
- 0
- 威望
- 0
- 包包
- 483
|
作者:Murielle Mimeault, Surinder K. Batra作者单位:Department of Biochemistry and Molecular Biology, Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, Nebraska, USA " U; i& e* N; ~, q: P9 P# I
. R9 ^3 b9 f: }/ C
k1 \: r& g t) K# U
% Z, V: a; f7 B' N, H) t
3 O8 y# c5 g; K% B . `0 V9 L9 k. P( V2 D
( p/ J% U' p- G- y5 v( a" ~! p
1 h# U7 O/ L7 U0 j
: d1 E% N$ v9 b( [% }6 O
, Z$ ]9 b$ k7 ]) W% Y# ]- k; f/ v
& ?% N- f& c9 k" ~2 [ # ?$ }4 c& V0 G% ^. J, v
9 }* _; R' ^6 c6 i1 A; ] 【摘要】/ M+ R+ j' [! I$ C
In this study, we report on recent advances on the functions of embryonic, fetal, and adult stem cell progenitors for tissue regeneration and cancer therapies. We describe new procedures for derivation and maturation of these stem cells into the tissue-specific cell progenitors. The localization of the adult stem cells and their niches, as well as their implication in the tissue repair after injuries and during cancer progression, are also described. The emphasis is on the interactions among certain developmental signaling factors, such as hormones, epidermal growth factor, hedgehog, Wnt/ß-catenin, and Notch. These factors and their pathways are involved in the stringent regulation of the self-renewal and/or differentiation of adult stem cells. Novel strategies for the treatment of both diverse degenerating disorders, by cell replacement, and some metastatic cancer types, by molecular targeting multiple tumorigenic signaling elements in cancer progenitor cells, are also illustrated. 4 S0 k7 w$ ?* A
【关键词】 Embryonic stem cells Adult stem cells Tissue regeneration Oncogenic transformation Stem cell-based therapies) ]. d; L( k; X ]/ k5 `( Q1 r
INTRODUCTION
" F+ P/ y% F0 O& y8 B2 r Z2 ?4 @+ F9 K! X* w) c, c
There is great interest in the biology of adult stem cells because of their capacity to self-renew and their high plasticity. These traits enable adult stem cells to produce diverse mature cell progenitors that actively participate in the maintenance of homeostatic processes by replenishing the cells that repopulate the tissues/organs during a lifespan and regenerate damaged tissues during injury . Parkinson and Alzheimer diseases, muscular degenerative disorders, chronic liver and heart failures, and type 1 and 2 diabetes, as well as skin, eye, kidney, and hematopoietic disorders, could be treated by the stem cell-based therapies (Table 1).
* i) h ]; f; A4 d! E) s' s8 ^$ l5 s
Table 1. Therapeutic applications of embryonic, umbilical cord blood, and adult stem cell progenitors: H* y" L8 ^8 l4 P
8 f, c3 I, V9 f5 h l+ z1 z+ q
Genetic alterations and/or sustained activation of distinct developmental mitogenic cascades occurring in a minority of adult stem cells and their progenitors might also lead, in certain cases, to their oncogenic transformation . Molecular targeting of these oncogenic signaling elements, therefore, constitutes a promising approach for the development of novel combination therapies against these metastatic and incurable forms of cancer. In this review, we focus on the growth factor signaling cascades that might be implicated in the control of the self-renewal and differentiation of embryonic, fetal, umbilical cord, and adult stem cells and their progenitors in vitro and in vivo. Moreover, we report a description of the in vivo localization and biological functions of adult stem cells. We also describe recent advances in potential therapeutic applications of stem cell progenitors in regenerative medicine and new combination therapies for cancer treatment.
( \9 u6 l |! x# Q1 O; j5 x7 [# S! q2 q/ m6 s
STEM CELL TYPES! _% X, b1 r9 @% V& R% E( j
@8 j3 x; v2 |, [
Distinct stem cell types have been established from embryos and identified in the fetal tissues and umbilical cord blood (UCB), as well as in specific niches in many adult mammalian tissues and organs, such as bone marrow (BM), brain, skin, eyes, heart, kidneys, lungs, gastrointestinal tract, pancreas, liver, breast, ovaries, prostate, and testis . All stem cells are undifferentiated cells that exhibit unlimited self-renewal and can generate multiple cell lineages or more restricted progenitor populations that can contribute to tissue homeostasis by replenishment of cells or regeneration of tissue after injury.
2 X7 i J& p9 F; O2 N5 @3 k( c( L9 I4 q6 @: Q! O
Several investigations have been carried out with isolated embryonic, fetal, and adult stem cells in a well-defined culture microenvironment to define the sequential steps and intracellular pathways that are involved in their differentiation into specific cell lineages. More particularly, different methods for in vitro culture of stem cells have been developed, including the use of cell feeder layers; cell-free conditions; extracellular matrix (ECM) molecules, such as collagen, gelatin, and laminin; and diverse growth factors and cytokines . We report here the structural and functional features of embryonic, umbilical cord, and adult stem cells and their niches, as well as the procedures that are used for their differentiation into particular cell lineages in vitro and in vivo.
5 G9 `% t! f4 O- e7 l
- g! w$ {% C! @9 ~0 sEmbryonic Stem Cells
: d" E; w, S- L U
+ Y/ u3 [& T# E; D% B. TSeveral mammalian pluripotent embryonic stem cell (ESC) lines derived from blastocyst-early-stage embryos have been established (Fig. 1) . Moreover, all ESC lines generally exhibit high levels of telomerase expression and activity for prolonged periods in culture. In this matter, ESCs possess the dual ability to undergo unlimited self-renewal and to differentiate in all fetal and adult stem cells and their more differentiated progenitors. Therefore, they represent a useful source of stem cells for investigating the molecular events that are involved in normal embryogenesis and generating a large number of specific differentiated progenitors for cellular therapies./ e( n1 r/ _5 o% e" s, S
' K7 \! T: M% p! ]% [9 P& _
Figure 1. Schematic diagram showing the possible differentiation pathways of ESCs. The ESCs can give rise to three germ layers, endoderm, mesoderm, and ectoderm, during embryonic development. Similarly, ESC progenitors derived through the formation of EBs might also form teratomas corresponding to the complex structures containing the cell types from three germ layers in vivo. Moreover, the in vitro expansion and differentiation of ESC-derived progenitors in the presence of specific growth factors and cytokines in culture medium may also generate mature cell progenitors possessing the particular markers and biological features of cells constituting the tissues/organs of endodermal, mesodermal, or ectodermal origin. Abbreviations: EB, embryoid body; ESC, embryonic stem cell./ }, T2 v6 M. I9 y7 `
2 R7 p$ I- A4 I2 j
In Vitro Derivation of ESC Progenitors. ESCs are generally isolated from the inner cell masses (ICMs) of blastocysts, which consist of pluripotent cell populations that are able to generate the primitive ectoderm during embryogenesis (Fig. 1). More specifically, in normal embryonic development, the primitive ectoderm gives rise during the gastrulating process to the primary germ layers, including ectoderm, mesoderm, and endoderm. These three germ layers might subsequently generate a variety of organized tissue structures involving complex epithelial-mesenchymal interactions. Similarly, the injection of ESC-derived progenitors into severe combined immunodeficient (SCID) mice might also result in the formation of teratomas corresponding to the complex structures containing the differentiated cell types from three germ layers . However, mouse and human ESCs express different marker profiles and might respond differently to certain growth factors, giving rise to distinct cell lineage progenitors. Therefore, these interspecies differences underline the importance of further establishing the particular differentiation pathways of hESC-derived progenitors for their clinical applications in humans.
' S$ U# d# N$ k1 s/ A+ R; y. q/ }0 ?' L: p: R4 p- f7 c( C
One of the critical steps in the purification procedure appears to be the enrichment of EB-derived progenitor cells by the elimination of pluripotent and undifferentiated stem cells. Indeed, the elimination of undifferentiated stem cells that may form teratomas or teratocarcinomas in vivo appears to be essential for generating transplantable sources of differentiated stem cell progenitors for the treatment of diverse disorders .
* b! {& G5 ~" \" q. a
2 G5 m! p0 R- l8 c" c. {& tHence, together these studies have contributed to the development of new procedures for the derivation and enrichment of hESCs. These new procedures can be used for studying the developmental pathways involved in hESCs maturation into their commitment lineages, as well as generating a variety of differentiated hESCs progenitor types for their in vitro and in vivo use. First, the development of novel purification methods to obtain a level of 100% purity of hESC-generated progenitors in the transplants seems to be necessary before they can be used in the clinic.& O7 u! G& a0 U3 I4 y! c7 U
! ~- z8 p3 P9 J9 {6 n- T
Embryonic and Primordial Germ Cell Carcinomas. Several works have revealed that pluripotent ESCs, as well as primordial embryonic germ cells (EGCs) that are derived from embryonic gonads, might also give rise to teratocarcinomas in vivo. As a matter of fact, the injection of adapted human ESC-derived progenitors that are obtained after prolonged culture in vitro into SCID mice might result in the formation of teratocarcinomas whose tumors are composed of cells derived from the three germ-like structures . In fact, based on the hypothesis that certain adult cancer forms might also derive from the malignant transformation of tissue-specific adult stem cells into cancer progenitor cells, the genetic alterations observed in embryonic carcinoma cells might also provide information on oncogenic gene products that might be involved in initiation of certain adult cancer types.% c% L( y" d8 |8 z6 C* V
9 ]2 F i$ W- s( I2 XAmniotic Epithelial Cells
* [' {+ d7 p4 d& u/ S' Q# b4 o2 M, s( Z) \( v# a7 n8 {: K
Amniotic epithelial cells (AECs) derived from the amniotic membrane in human term placenta also express the markers that are present on pluripotent ESCs and EGCs, such as Oct-4, Nanog, and alkaline phosphatase . Of therapeutic interest, AECs do not express the telomerase and form no teratomas after transplantation in vivo. Therefore, AECs constitute a source of pluripotent stem cells that might be used in transplantation for tissue regeneration. q: j! Z( ]7 {6 H
' R2 y! I! s% O0 J+ u! l. c t9 FFetal Stem Cells2 H5 M) @. L) y' r( W1 M
( b6 f2 ^0 n5 T" O! kMultipotent fetal stem cells (FSCs) are generally more tissue-specific than ESCs. Therefore, FSCs are able to generate a more limited number of progenitor types. One of the particular therapeutic advantages of FSCs as compared with ESCs is the fact that FSCs do not form teratomas in vivo. Moreover, the FSCs obtained up to week 12 offer the possibility of transplanting these primitive stem cells without frequent rejection reactions, in contrast to UCB and BM stem cell transplants. As a matter of fact, recent work has revealed the possibility of using FSCs or their progenitors, isolated from particular tissues, for multiple therapeutic applications involving tissue regeneration .7 N L3 T5 y% y, A) N
* M0 l, Z: r# AOn the other hand, it is interesting to note that a reciprocal fetomaternal trafficking of cells and nucleic acids has also been shown through the placental barrier during pregnancy, which might contribute to tissue repair mechanisms in different maternal organs and the growing fetus . This reflects the high plasticity and migratory potential of FSCs, which represent major advantages for their use in transplantation. More specifically, the establishment of the structural features of FSCs that enable them to cross both the placental and blood-brain barrier could allow for the improvement of therapeutic applications of neural stem cell progenitors in allowing their administration by i.v. infusion for repair of diverse brain disorders.
: W. x9 B( U2 m# _& V/ g9 u0 _7 \3 u8 ^( P$ ?
Umbilical Cord Stem Cells
( J% _- I' t5 c0 v, Y
t0 X9 _; Y% r% a, j8 `Umbilical cord epithelium (UCE), which appears to derive from amniotic membrane epithelium, and UCB represent other sources of multipotent stem cells that might be used for generating diverse differentiated cell types . Hence, these dendritic cells may be used as an adjuvant in immunotherapy for diverse disorders and cancers.8 Q$ r+ O0 r$ u N A# |% e
9 ^; m& }; t3 B* f+ ZIn addition, human UCB also contains a more primitive subpopulation of mesenchymal stem cells than adult BM, the immature cells of which express adhesion molecules, such as CD13, CD29, CD44, CD90, CD95, CD105, CD166, and major histocompatibility complex class, but not the antigens of hematopoietic differentiation, such as CD34 . Thus, it appears that the differentiation of UCB stem cells into tissue-specific adult stem cell progenitors might constitute an alternative strategy for cellular therapies of diverse disorders (Table 1).; b. q' h) N5 W! u
* K- S! J/ h$ K' J2 W
Adult Stem Cells
: `* d- T1 {0 A6 @0 h! b% m+ k, X# Y/ p# }
Numerous studies have revealed that a population of adult stem cells and supporting cells reside within specific areas designated as niches in most of adult mammalian tissues/organs, including BM, heart, kidneys, brain, skin, eyes, gastrointestinal tract, liver, pancreas, lungs, breast, ovaries, prostate, and testis (Fig. 2) .
5 {/ k5 \; ~ x; h1 m7 Y3 w/ ~* z2 o5 h' T
Figure 2. Scheme showing the tissues/organs constituting the potent targets for tissue regeneration by stem cell-based therapies. The localization of tissue-specific stem cells and their niches is shown. The tissue-specific degenerating disorders and diseases that might be treated by the transplantation of stem cell progenitors derived from embryonic stem cells, fetal stem cells, umbilical cord blood, and adult tissues/organs, including bone marrow (BM), are also indicated. The possibility of the mobilization of BM stem cells and their progenitors in the bloodstream by using the mobilizing agents is also shown. Abbreviations: bESC, bulge epithelial stem cell; CESC, corneal epithelial stem cell; CNS, central nervous system; CSC, cardiac stem cell; CXCR, CXC-chemokine receptor; eNCSC, epidermal neural crest stem cell; G-CSF, granulocyte colony-stimulating factor; HOC, hepatic oval cell; HSC, hematopoietic stem cell; KSC, keratinocyte stem cell; MPC, mesodermal progenitor cell; MSC, mechenchymal stem cell; NSC, neural stem cell; PSC, pancreatic stem cell; RSC, retinal stem cell; SDF, stromal-derived factor; SKP, skin-derived precursor.
2 `. ^+ z8 G; e5 p8 R/ c0 y7 @8 Y" u! a
Figure 3. Proposed model of the cellular events associated with the tissue regeneration by adult stem cells and cancer progression originating in their malignant transformation into cancer progenitor cells. The cellular events that are implicated in the tissue regeneration after injury, including the asymmetric division of adult stem cells into transit-amplifying cells, which in turn may give rise to the mature cells that repopulate the tissue, are shown. Moreover, the malignant transformation of adult stem cells and/or their progenitors into cancer progenitor cells induced through genetic and epigenetic alterations (gene mutations, deletions, amplifications, and chromosomal rearrangements or change in DNA methylation) and whose genetic modifications might lead to the generation a heterogeneous cancer cell population is shown. The activation of numerous growth factor signaling cascades and tumorigenic signaling elements in cancer cells, which are implicated in cancer progression, as well as their molecular targeting by using the selective inhibitors for the development of novel combination therapies against the aggressive cancer forms, are also indicated. Abbreviations: COX, cyclooxygenase; CXCR, CXC-chemokine receptor; ECM, extracellular matrix; EGFR, epidermal growth factor receptor; MMP, matrix metalloproteinase; SDF, stromal-derived factor; uPA, urokinase-type plasminogen activator; VEGF, vascular endothelial growth factor; VEGFR, vascular endothelial growth factor receptor.
$ P5 i! E6 p" F+ P) J( e7 T# v& L0 r5 e2 @ }. @3 M0 ^+ _: ~! c
Certain concepts, based on the possibility of symmetric and asymmetric divisions or populational asymmetry, have also been proposed for explaining the balance between stem cell self-renewal and differentiation , skin, and eyes) origin and the molecular events that might be implicated in their decision to undergo sustained proliferation or adopt a specific differentiation pattern under normal or pathological conditions. More particularly, the emphasis is on the adult stem cells that can give rise to a broad range of progenitors. These adult stem cells are also able to trans-differentiate in different mature cell lineages in vitro and in vivo.
; o: w2 x, U" _3 {
2 p3 u" g+ @. Y. c% mAdult Stem Cells of Endodermal Origin9 W4 z: j/ T8 Z
, |% q) w7 w# T8 v& g: z) bPulmonary Epithelial Stem Cells. The multipotent pulmonary epithelial stem cells are able to differentiate into ciliated, secretory, intermediate, and basal cells and generate the submucosal glands. Although their specific markers and niche(s) have not yet been established precisely .$ g3 f: R2 Z( `3 {* M7 P H
; |$ R* B. d& ?% H" F
Gastrointestinal Tract Stem Cells. The replenishment of epithelial cell lineages within the gastrointestinal tract is a frequent process, occurring every 2¨C7 days under physiological conditions. This process may contribute to the generation of new cell progenitors, which repopulate the damaged tissues during diverse pathological disorders, such as inflammation and ulceration ./ w& v# p0 a. }; X) Y, L
5 I! J% X; @8 y1 ~* {" f! YPancreatic Stem Cells. The mammalian adult pancreas has three tissue types: the ductal tree, the exocrine acini, which produces digestive enzymes, and the endocrine islets of Langerhans, composed of insulin-producing ß-cells, glucagon-producing -cells, somatostatin-producing -cells, and pancreatic polypeptide-producing -cells (Fig. 2). Much evidence indicates the presence of putative pancreatic stem cells (PSCs) in ductal and/or islet regions of the mammalian pancreas. The localization of the niche(s) of these putative PSCs is not yet precisely known. In this matter, the exocrine and endocrine cells appear to be produced from the differentiation of ductal cell progenitors during embryonal development . These ICCs can give rise to diverse pancreatic cell lineages, including insulin-secreting cells.
* ?+ Y }, o) n" t. q; x# e d2 V# N1 \- x& g2 h4 v
In addition, the stem cells of other sources expressing the pancreas-developing markers, including nestin, K8, K18, neurogenin-3, and nuclear transcription factors such as IDX-1, Pax-4, Pax-6, and Isl-1, which are important for the ß-cell differentiation, might be expanded and trans-differentiated under specific culture conditions in vitro. More specifically, the differentiation of ESCs, fetal and UCB stem cells, and adult stem cells from the liver, BM, spleen, and nervous system into pancreatic insulin-producing ß-cell-like progenitors has been performed in vitro by using growth factors such as bFGF, SCF, nicotinamide, betacellulin, glucagon-like peptide, and activin A (Table 1) .
% N/ y* r; t" h+ j5 ^# _4 a9 s( A4 @
Hepatic Oval Cells. Mature hepatocytes can undergo several cell division cycles and are responsible for continuous hepatic cell replacement during extensive liver injury. A small population of stem cells designated hepatic oval cells (HOCs) may also be involved in tissue regeneration . Thus, additional works are essential to establishing the factors that control the migration, incorporation, and homing of extrahepatic stem cells in the liver under specific in vivo conditions.! v$ _ S& z- x1 F, `5 \
% A' U& v+ V; Y8 X
Urogenital Stem Cells$ H. r* P0 }$ v/ `. p( k% r
4 s8 q8 P' A9 H6 m9 G# k* ^% ^/ PMammary and Prostatic Gland Stem Cells. The regulation of the self-renewal, differentiation, and migration of urogenital stem cells and their progenitors that are localized in the mammary and prostatic glands appears to be assumed through distinct developmental signaling pathways such as hormones, EGF, hedgehog, Wnt/ß-catenin, Notch, and/or BNPs .7 H( \9 [, q5 ^( u
# s+ v0 a. j( o, B1 h% eSimilarly, a stem cell population found in the embryonic urogenital sinus epithelium from which prostatic epithelial buds develop also appears to persist in the adult prostatic epithelium and gives rise to basal and luminal epithelial cells. More specifically, a small pool of the adult prostate-specific stem cells, which show unlimited growth in a specific microenvironment and generate multiple more-differentiated epithelial cells due their striking plasticity, has recently been isolated from the proximal regions of prostatic ducts ." ^ v+ }/ j) R9 W$ J/ h! |
, I6 b |4 @. J9 d7 U
Ovarian and Testicular Stem Cells. Although several lines of evidence have indicated the presence of putative multipotent stem cells, which give rise to diverse differentiated epithelial and germ cells constituting the adult mammalian ovaries and testes, their precise localization needs to be identified. As a matter of fact, the multipotent stem cells have been established from neonatal gonads, testes, and ovaries in mouse and human . These primary follicles that were formed by the oocytes and granulosa cells derived from the OSE cells could also give rise to the mature secondary oocytes in vitro. Therefore, these findings may constitute the basis for future stem cell-based research in the development of novel in vitro fertilization strategies; however, additional work is necessary to establish the functional properties of these in vitro generated human secondary oocytes before their putative use in the clinic.
5 y- K7 K' T ?1 x' {& J7 c
+ Y+ l# a- r8 `3 P7 @2 z/ HGenetic modifications in testicular and ovarian somatic stem cells and germ cells in mice and humans may also lead to tumor formation. As a matter of fact, a tumor stem cell population showing stem cell-like characteristics has been established from human ovarian tumors, which proliferate and form tumors in animal models . Additional investigations are necessary to establish the specific functions of these stem cell markers, as well as other genetic alterations that may occur in adult ovary and testicular stem cells and germ cells and that may be responsible for tumor formation in humans.5 ^! S: D3 u$ i
3 ~1 V' U x6 `% g6 [" TAdult Stem Cells of Mesodermal Origin
% h3 F# D1 v& |2 {' n$ w* c1 M1 Z4 Z3 Z- ^, ?* o
Bone Marrow. BM is a well-organized tissue composed of the basic elements from the stroma and hematopoietic system and located at the center of large bones (Fig. 2). BM contains HSCs and stromal stem cells that collaborate in a reciprocal manner at all stages, leading to the generation of different BM and bloodstream cell lineages .
8 G# y3 l0 S) H, N) a& T6 U# L# d( l9 B' Q
Hematopoietic Stem Cells. In the adult BM, the HSCs are localized near endosteal bone surface and sinusoidal endothelium, suggesting that these sites could constitute the principal niches for their homing in BM under physiological conditions and for engraftment of HSCs after systemic transplantation (Fig. 1) . Further investigation is necessary to establish the specific signals that govern the rate of proliferation, differentiation, and migration of HSCs under physiological and pathological conditions. This should allow us to identify the specific functions that are associated with each type of HSC that is localized in BM and other distant sites, such as spleen and liver.
; N3 |+ L. O7 }" ^/ D) i2 B" ^) E4 x7 a4 d; q2 h6 ~
In addition, HSCs and their progenitors may also be obtained from different sources, including ESCs, FSCs, and UCB as well as adult BM and MPB, and differentiate in vitro and ex vitro into different hematopoietic cell lineages .# z3 L) o0 m" }
" j. n4 A6 l% x3 W! j
Stromal Stem Cells. The BM stroma is a highly vascularized, complex structure containing mesenchymal stem cells and extracellular matrix elements supporting for hematopoiesis .! V# E8 x* }7 y# Z; k" ]& F i
% m2 K* d3 r3 J7 e, q
It has been reported that the stromal stem cells from BM may be involved in continuous bone remodeling, as well as in the regeneration of injured bone, cartilage, and other distant tissues, such as the liver, lungs, gut, and heart, in adults .& j- H5 _' j/ W( U6 k
) i* R# J* |, |0 z( j3 g
Cardiac Stem Cells. The clusters of multipotent cardiac stem cells (CSCs) expressing the cell surface antigen Sca-1, KIT, and/or multidrug resistance transporter gene 1 (MDR1) in variable combinations have recently been identified and localized throughout the myocardium, and more particularly, at the atria and lower region of the left ventricle of the heart, termed apex (Fig. 2) . Thus, it appears that the multipotent quiescent CSCs and neural crest stem cells that reside in the adult mammalian heart may contribute to the regeneration of different cardiac cell lineages following injury.% C+ T0 ~% G0 y3 Z4 f
6 A7 J/ ]: T/ t0 }Stem Cells of Ectodermal Origin+ n5 k, T6 c+ }0 l( H8 `5 w8 ~
* g( O* u& w: d4 q( E e
Neural Stem Cells. During prenatal development of the mammalian CNS, the neural stem cells (NSCs) and their progenitors may expand and give rise to the functional neurons and glial cells that constitute the growing brain . Hence, the generation of new differentiated neural cells from adult neural stem cells in the specific brain regions during a lifespan might assume the maintenance of CNS homeostasis and functions, particularly for learning and memory.. U. e* t/ R" G, O! w9 y+ F
5 p, @6 P3 f8 U' h: a" k, m
In Vivo Proliferation and Differentiation of NSCs. Several studies have been undertaken to establish the microenvironmental and intrinsic factors that might influence the behaviors of adult NSCs in vivo. Among the numerous growth factors and adhesion molecules that may be involved in the regulation of proliferation, maturation, and/or migration of adult NSCs, there are EGF, bFGF, SHH, Wnt/ß-catenin, Notch 1 ligand jagged 1, platelet-derived growth factors (PDGFs), ciliary neutrophic factor, VEGF, thyroid hormone T3, dopamine, NGF, neuregulins, BMPs, TGF-ß, Ephrins/Ephs, leukemia inhibitory factor (LIF), and integrins . This suggests that the endogenous VEGF from endothelial cells might also contribute of paracrine fashion to the NSC activation in vivo. Based on the knowledge of the factors involved in the regulation of embryonic and adult NSC growth, survival and differentiation in vivo, several new methods for in vitro expansion and differentiation of embryonic and adult NSCs have been conceived.
6 l9 C) n; ~7 j' ~# { X
, i3 i+ C8 G- SIn Vitro Expansion and Differentiation of NSCs. Results from numerous studies have revealed that the human and rodent NSC progenitors derived from ESCs, UCB, fetal brain, MSCs, or skin-derived stem cells or isolated from adult brain tissues might be expanded in vitro or ex vivo in floating clusters called neurospheres in the presence of exogenous EGF, bFGF, SHH, and/or LIF. Moreover, the withdrawal of these mitogens and the addition of serum, RA, BNP, TGF-ß type III, and/or ascorbic acid may promote their differentiation in the three major neuronal cell types, including neurons, astrocytes, and oligodendrocytes (Table 1) . Hence, together these works have identified certain growth factors and cytokines that might promote the expansion and differentiation of embryonic, fetal and adult NSCs into the specific neuronal cell lineages and possibly constitute the basis for the development of novel therapies for a variety of CNS disorders.: C( f; r8 G+ o# y4 z0 }9 w
8 m9 P2 `5 \1 M; B
Skin Stem Cells. In adult mammalian skin, the epithelial compartment consists of the interfollicular epidermis (IFE) and its related appendages, such as the hair follicles and sebaceous glands. Numerous studies have revealed that the upper region of hair follicles, the bulge area, constitutes the principal niche of multipotent stem cells, which are responsible for the long-term growth of the hair follicles and epidermis regeneration after injury (Fig. 2) .- f& y: {* m9 n/ n1 f, a9 p4 c
' v) V( n* `) K! m, ?The bulge area in adult mammalian hair follicle also contains a pluripotent epidermal neural crest stem cell (eNCSC) population that shows several properties similar to embryonic neural crest stem cells (Fig. 2) .
* B% |% Y, \3 X7 l5 z
- F+ j3 }7 z& l. Y3 EIn addition, the keratinocytes constituting the basal layer of epidermis also need to undergo a continuous expansion to replenish the terminally differentiated epithelial cells in the corneous layer of normal epidermis, which are shipped out of skin during the desquamation process . In this matter, since bESCs might also give rise to cells constituting IFE after injury, additional investigations appear necessary to determine the specific implication of bulge stem cells versus KSCs residing near the basement membrane during the different stages leading to the epidermis regeneration after trauma and carcinogenesis. X: p7 h! ^7 z i/ K
9 d! o! C/ c3 y: Q9 a7 p- mOcular Stem Cells. The human ocular surface epithelium includes the corneal, limbal, and conjunctival stratified epithelia. Several recent lines of evidence have revealed that the corneal epithelial stem cells (CESCs) are localized at the basal cell layer of the peripheral cornea, and particularly at the limbus within the limbal epithelial crypts (Fig. 2) . These stem cell-derived TA cells could also migrate at the endothelial periphery, suggesting their possible implication in eye endothelium regeneration.% f0 o7 N6 T; }- n( \; P* y
( k% X7 @* o* h$ OA small population of mitotic quiescent neural stem cells has also been identified in the ciliary epithelium (CE) region adjacent to the retina in adult mammalian eyes, which may proliferate in response to retinal injury in vivo or after treatment with specific exogenous growth factors in vitro (Fig. 2) . Hence, the discovery in the adult eyes of limbal CESCs and RSCs in CE, which possess immature epithelial and neural cell properties, respectively, offers the possibility to use these stem cell types for the repair of corneal epithelium and retinal damages.
7 t; g+ U3 N3 r$ f2 a! K
4 y/ D; T4 D# Y# a! f7 h( _STEM CELL-BASED THERAPIES$ n y. k: t4 w: G' `" O
5 b1 Y( {4 Q! W) T
The possibility of using stem cells and their more differentiated progenitors to treat numerous degenerative disorders has stimulated great interest in developing safe transplantable sources of stem cells that are unable to form the teratomas but able to repopulate damaged tissues. Many recent investigations have provided interesting clinical findings about the possibility of using the progenitors derived from ESCs, AECs, FSCs, and UCB and BM stem cells, as well as other adult tissue-specific stem cells in genetic and cellular therapies for a wide variety of pathological processes, including degenerative, autoimmune, and genetic diseases. Clinical transplantation procedures for stem cells, which depend on patient state and diagnosis, generally involve the i.v. injection or subcutaneous administration of a specific number of stem cells directly into therapeutically targeted areas (Fig. 2). Among the disorders that might benefit from stem cell-based therapy are diabetes, acute liver and heart failures, muscular disorders, arthritis, brain damages and disorders, vision disorders, renal disorders, and hematopoietic and immune diseases, as well as acute leukemia and lymphoma and diverse solid tumor types . In addition, molecular targeting of tumorigenic cascade elements in tissue-specific cancer progenitor cells, which are derived from the malignant transformation of adult stem cells, also represents a novel approach for the treatment of diverse metastatic and incurable cancer types by combination therapies. We report the recent advances on the more promising stem cell-based strategies that have been developed in past years for the treatment of numerous degenerative disorders and aggressive cancer types./ l+ G C9 z; b
8 H6 n. @; G E6 M) s2 S; X" ^1 M
Regenerative Medicine
2 N1 a9 p" A# S; v5 v3 w
8 q+ u$ y; c& ?* S. c' lPancreatic Diseases. Numerous studies have been undertaken to establish the insulin-producing ß-cell-like progenitors for the development of new treatments of type 1 and 2 diabetes mellitus. In fact, the replacement of insulin-producing pancreatic islet ß-cells that are destroyed or reduced in number results in the insulin deficiency and fasting hyperglycemia and constitutes a promising strategy. The ß-cell-like progenitors derived from human and mouse ESCs, fetal and UCB stem cells, and adult stem cells from pancreas and BM, showing the capacity to secrete insulin, have been obtained in vitro . This observation then suggests that HOCs could also constitute another source of pancreatic cells for the treatment of diabetes. Thus, it now appears possible to conceive the development of cell replacement therapies for human diabetic patients by using human stem cell-derived ß-cells. Nevertheless, additional work on the identification of specific factors that are involved in the migration and trans-differentiation of BM and liver stem cells into insulin-producing islet ß-cell-like progenitors in vivo is needed. In addition, the functional properties of ex vivo and in vitro expanded ß-cells in distinct animal models in vivo are essential. Optimization of these protocols is essential before their translation into clinical applications for humans. In this matter, the establishment of in vitro methods for obtaining a large amount of functional ß-cells and the optimization of delivery site are also important factors that must be considered for improving their therapeutic efficacy in the long-time treatment of patients.
+ D; q4 \: G8 }6 a( S/ E& M% r/ Y% \$ A
5 s/ Y; C1 o" O" b( Z3 OCardiac Diseases. The generation of new myocytes from endogenous multipotent CSC pools offers the possibility for maintaining cardiac homeostasis and tissue regeneration after heart hypertrophies and failures. The stimulation of the proliferation, survival, and/or differentiation of the endogenous CSCs and their progenitors may be performed by using the specific growth factors that are involved in the control of cardiogenesis and neo-angiogenesis, such as VEGF, HGF, IGF-1, EPO, TGF-ß, HIF-1, and IL-8 . Despite this advance, the lack of knowledge of the molecular mechanisms that are responsible for human CSC (hCSC) senescence, as well as the difficulties associated with the procedures of the isolation and in vitro expansion of functional hCSCs, limit their clinical applications as effective therapies for myocardial regeneration.6 q: \+ q; {- U; P4 ~( V* J& `* S
5 f$ j4 Y' A0 p, _0 N
In addition, another alternative may consist of the expansion and trans-differentiation of other stem cell types into cardiac cell progenitors in vitro or in vivo under specific conditions that may also enable them to engraft and regenerate the functional cardiomyocytes and endothelial cells for the reconstitution of damaged myocardium after injury. More particularly, several recent investigations indicated that the transplantation of ESC- and BM stem cell-derived cardiac cell progenitors might result in stable and functional intracardiac grafts in human and rodent, thereby improving cardiac functions by regenerating the infarcted myocardium . Hence, it appears that the ESC- and BM stem cell-derived cardiac cell progenitors could constitute a more accessible source of stem cells than CSCs for the treatment of diverse cardiac disorders. Additional long-term trials on large populations in human are necessary to establish the functional properties of these cardiac progenitors and their efficacy for the recovery of cardiac function after transplantation.
* V- S/ B5 o! o% p' Y6 H: q& N! u/ j0 _, E1 n% ?
CNS Disorders and Diseases. The stimulation of adult neurogenesis in vivo by supplying specific growth factors and cytokines, such as G-CSF and SCF, that stimulate intrinsic NSC progenitors and BM-derived neuronal cells or delivery of exogenous NSC progenitors constitute the novel strategies that are very promising for restoring the impaired functions in numerous damaged brain areas after ischemia, seizures, and traumas or in diverse degenerative disorders (Table 1; Fig. 2) .
/ r1 }- ?, J& |' ]6 J) m0 h/ g& C. T( R) N9 A
Parkinson and Alzheimer Diseases. The motor dysfunctions that are associated with Parkinson disease result from the progressive loss of dopaminergic neurons in the substantia nigra pars compacta, a region of brain that controls muscle movement. Therefore, cell replacement therapy by delivering new dopaminergic neurons represents a putative strategy for the treatment of this neurodegenerative disease .
3 C+ S a. X/ J/ z3 ?" G: p3 y
# P( @, J4 z) k# @1 x. ISpinal Cord Injuries. Since the major cause of neurological disability in spinal cord injuries is oligodendrocyte death leading to demyelization and axonal degeneration, rescuing the oligodendrocytes and preserving myelin should result in a significant improvement in the functional outcome after such injuries. It has been reported that the addition of oligodendrogenic factors such as SHH, bFGF, and PDGF might result in the differentiation of NSCs into oligodendrocyte progenitor cells (OPCs) in vitro. However, the rate of OPC differentiation of NSCs under these conditions was only approximately 10%. Interestingly, the transient expression of oligodendrogenic helix-loop-helix transcription factor Oligo 2 in NSCs, which is essential for oligodendrocyte lineage specification, may induce a high rate (approximately 55%) of OPC differentiation and generate the mature and functional oligodendrocytes expressing the transcription factor Nkx2.2 and all major myelin-specific proteins . Thus, on the basis of these observations, it now appears possible to conceive novel stem cell-based therapies for a variety of brain disorders. At this time, there exist only palliative treatments for these diseases. In this matter, the development of new strategies for the delivery of NSCs and their progenitors in the specific brain damaged areas or selective activation of endogenous NSC-derived progenitor subset should also contribute to improving their potential therapeutic applications in humans.
! f8 w4 I; [6 O/ h E, T+ K& z# N: {! _+ O
Ocular Disorders and Diseases. CESCs and RSCs, isolated from the limbal epithelium and CE of adult eyes, respectively, are able to incorporate and reconstitute the damaged tissues in vivo. Therefore, they may be transplanted for the repair and regeneration of ocular surface damages. Several types of treatment for ocular disorders have been developed based on the use of limbal CESC transplants and bioengineered ocular surface tissue-equivalents for the replacement of ocular damaged tissues . Further investigation of the behaviors of endogenous CESCs and RSCs and their progenitors in eyes should allow us to conceive new methods for the functional recovery of partial ocular defects by their stimulation by diverse exogenous growth factors. Additional in vivo trials on the functional properties of ex vivo and in vivo expanded CESCs and RSCs, as well as the precise functions of amniotic membrane, are also necessary to establish the molecular mechanisms that are responsible for their therapeutic effects. These studies should contribute to improving the long-term survival rate of these adult stem cells after engraftment and thereby to their beneficial effects for the treatment of eye diseases.
; W. v$ a F8 `$ h0 y
. ^$ k8 \: U/ p) HBlood and Immune System Disorders. HSCs and their progenitors, which might be obtained from different sources including UCB, fetus, BM, and MPB, represent potent cells for autologous or allogeneic transplantation in the patients with hematological and autoimmune diseases .
$ _2 W1 ~3 }9 N Y: R$ a2 m% k( J% J ^) p: F
Table 2. Deregulated gene products in cancer cells and their molecular targeting as anticarcinogenic therapies
/ l8 v+ }: c' B) O1 j- K, K$ d0 t8 C) l4 a9 t7 q, J
Cancer Therapies
0 v7 {$ y( u; A1 \; ^- L- c- v, C8 g1 X6 q1 t0 \; C
Numerous studies have indicated that several human cancer types, including those of the blood, brain, skin, lung, kidneys, gastrointestinal tract, pancreas, liver, ovarian, prostate, and testis, might arise from the malignant transformation of stem cells and their progenitors into cancer progenitor cells . In fact, it has been observed that only these malignant cells were able to give rise to a heterogeneous population of more differentiated cancer cells in vitro and to reconstitute the original cancer-like tumors in NOD mice in vivo. Thus, on the basis of these observations, it appears that the elimination of this minority of cancer progenitor cells with stem cell-like properties, which are responsible for tumor formation, is essential for the development of more effective curative treatments against these aggressive cancer types.
! m! {4 |+ w! Q- S4 F& R% O. c! n9 V1 A; X, t3 Z" z4 z
Several in vitro and in vivo studies have been carried out with a variety of cancer cell line types and on different animal models to identify new therapeutic targets to block the growth and/or survival of the cancer cells. Among them, the molecular targeting of distinct oncogenic signaling elements, which are activated in the cancer cells during the progression of numerous cancer, represents a promising strategy for the development of new chemopreventive treatments and combination therapies against some aggressive and metastatic cancers (Table 2; Figs. 3, 4). The aberrant expression and/or activity of diverse hormones, growth factors, cytokines and chemokines (androgens, estrogens, EGF and TGF-/EGFR, IGF/IGFR, SHH/SMO, Wnt/ß-catenin, Notch, TGF-ß, and SDF-1/CXCR4), and tumorigenic signaling elements (telomerase, phosphatidylinositol 3-kinase . Therefore, their molecular targeting is of importance to the elimination of cancer progenitor cells, thereby inducing a complete tumor regression and cancer remission. We report here a brief description of new therapeutic drugs that are able to block the specific growth factor signaling cascades that are frequently deregulated in the stem cell-derived cancer progenitor cells, as well as the advantages that are associated with the use of high-dose chemotherapy (HDCT) with hematopoietic cell support.
W3 P6 @% K! [! r! I
) y0 w; f6 C- B3 v6 BGrowth Factor Signaling Inhibitors
# Y, ^4 F" O7 i7 r7 q4 d: g- @! I2 E- _5 s+ I% j& l1 F
EGFR Family Member Inhibitors. Numerous preclinical and clinical trials have indicated that the selective blockade of the EGFR family member signaling, and particularly EGFR (ErbB1) or erbB2 (Her2), might represent a potent strategy, alone or in combination with other conventional treatments for numerous aggressive cancer forms . The inhibition of EGFR was also accompanied by cancer cell growth inhibition.
: D5 o5 p, I J; v B4 d) [7 N! Y' H* @3 D$ r, t
Figure 4. Proposed strategies for the development of novel combination therapies by targeting distinct oncogenic signaling pathways in cancer cells. The possible mitogenic and anti-apoptotic cascades induced through EGFR, hedgehog, Notch, and Wnt/ß-catenin signaling pathways, which can be involved in the stimulation of sustained growth, survival, and/or migration of cancer cells, are shown. Moreover, the possible growth inhibitory and/or apoptotic effects induced by the selective inhibitors of EGF-EGFR system (gefitinib), Smoothened hedgehog signaling element (cyclopamine), Notch signaling (-secretase inhibitor), and Wnt cascade by using anti-Wnt antibody on the cancer cells are also indicated. Abbreviations: APC, adenomatous polyposis coli; CDK, cyclin-dependent kinase; CoA, coactivators; Cyt c, cytochrome c; Dsh, Dishevelled; EGF, epidermal growth factor; EGFR, epidermal growth factor receptor; Fzd, Frizzled receptor; GSK, glycogen synthase kinase; ICN, intracellular domain of Notch; LEF, lymphocyte enhancer factor; LPR, lipoprotein coreceptor; MAPK, mitogen-activated protein kinase; MEK, extracellular signal-related kinase kinase; NF-B, nuclear factor-B; PI3K, phosphatidylinositol 3-kinase; PLC, phospholipase C; PTCH, Sonic hedgehog-patched receptor; SHH, Sonic hedgehog; SMO, Smoothened; TCL, T-cell factor.& @1 m8 Q% J/ z+ R+ A
; C( j, a0 _# \: X6 R6 K
Hedgehog, Wnt/ß-Catenin, and Notch Signaling Inhibitors. The inactivating mutations in the hedgehog receptor patched (PTCH) and/or activating mutations in the hedgehog signaling element, SMO, might increase the incidence of BCC, medulloblastoma, and rhabdomyosarcoma .: q1 V% W0 P, ]3 X! z, Q1 Y
& [8 y0 i {1 u; b) m/ K
In addition, molecular targeting of the canonical Wnt/ß-catenin signaling elements (which may contribute to the malignant transformation of cancer progenitor cells and the progression of numerous cancers, including upper gastrointestinal tract, colorectal, pancreatic, mammary, prostatic, and epidermal cancers, as well as non-small-cell lung carcinoma, hepatoblastoma, medulloblastomas, and multiple myeloma) constitutes another anticarcinogenic strategy for the treatment of these cancer forms .& o+ n Z% r' s( L0 F$ ~1 g; ?
5 Q; F" r: I r/ I/ V( _Combination Therapies. The simultaneous inhibition of diverse hormone and growth factor signaling pathways, including ER, AR, IGFR, EGFR, hedgehog, Wnt/ß-catenin, Notch, and/or G-protein-coupled receptors, as well as VEGFR and PDGFR cascades, which can act in cooperation by stimulating the growth, invasion, and metastatic spread of cancer cells at distant sites during the different stages of cancer progression, may also constitute more effective therapies against the aggressive and highly metastatic cancer forms . f1 R3 G- Q+ K1 [/ f. U9 U
8 N, G5 h, X* r- n' d3 q) @. F0 }Since the metastatic spread of diverse tumor cells, including those from glioblastomas, melanomas, and pancreas, breast, and prostate cancers to other specific tissues/organs, such as lymph nodes, bone, lungs, and/or liver, appears to be governed by the expression of diverse angiogenic factors, such as VEGF-VEGFR system, matrix metalloproteinases, urokinase-type plasminogen activator (uPA), cyclooxygenase-2 (COX-2), chemokines, and surface adhesion molecules, their molecular targeting may also constitute another adjuvant cancer therapy (Table 2; Fig. 3) . Altogether, these recent studies have indicated that molecular targeting of EGFR signaling, alone or in combination with other cytotoxic agents, may constitute a putative strategy for conceiving more effective clinical treatments against a variety of aggressive cancers.6 C4 H+ }; \, l- @( ^9 K1 T
+ C- Y4 p) O. z( `. {& n4 THigh-Dose Cancer Therapy Plus HSCs. Stem cell transplantation may also constitute an option as adjuvant therapy for cancer, particularly in the patients receiving high doses of chemotherapeutic agents and/or radiation that, along with killing cancer cells, cause the severe damage to normal tissues and/or destroy the hematopoietic cells. Thus, the stem cell transplants might replace the endogenous stem cells destroyed by high-dose cancer treatment, thereby producing healthy hematopoietic cell lineages and improving the immune system defense. The autologous or allogeneic transplantation of UCB, BM, or MPB stem cells and their progenitors might be effectuated in combination with HDCT for numerous aggressive cancer forms to replace BM and blood-forming cells that have been destroyed by chemotherapy. AML and high-grade lymphoma are among the principal types of cancer that are usually treated with hematopoietic cell support as adjuvant therapy .
7 O) N/ D: [. u7 r/ o. V) X- |& j: d A
In addition, transplantation or mobilization of HSCs and their progenitors in systemic circulation is often used as immune support in combination with HDCT for the treatment of patients with certain highly aggressive solid tumors, and more particularly in advanced and metastatic stages of germinal cell tumors, retinoblastoma, myeloma, brain, lung, kidney, breast, and ovarian cancers .
1 T" M3 u* }: v8 [4 R
p4 m$ O2 A# M7 DCONCLUSIONS6 a4 m0 v* I! `6 w# n
" A5 q. v3 A; I5 B9 v: nThese recent works in the field of stem cell biology have identified intrinsic mitogenic signaling cascades that are activated in mammalian embryonic, fetal, and adult stem cells during the normal process of self-renewal and differentiation. These cellular events may also be implicated in the regenerating process after tissue injuries. Hence, this offers the possibility of differentiating these stem cell types into the specific mature cell lineages in vitro, ex vivo, and in vivo by using appropriate growth factors and cytokines for their use in basic research, as well as in transplantation for diverse degenerative disorders. Several molecular targeting therapies may also be conceived by blocking distinct developmental signaling cascade elements, such as EGFR, hedgehog, Wnt/ß-catenin, and/or Notch pathways, which are frequently upregulated in cancer progenitor cells during the initiation and development of a variety of aggressive and metastatic cancers.4 N# q4 _6 v% W- l8 L4 H
- `4 e. M; m- ?: R, F$ n, k: a1 QPERSPECTIVES AND FUTURE DIRECTIONS
! W( q" V# I3 D0 L: b1 m2 ^4 u( I, g! t
Further in vitro and in vivo investigations with the human embryonic, fetal, UCB, and adult stem cells and their more differentiated progenitors in different systems, animal models, and humans appear to be essential to determine their functional properties during long time periods. Detailed analyses of structural and biological properties of differentiated progenitors established from human stem cells in vitro in comparison with those of endogenous adult stem cells are highly needed before they are used in the clinic. In this direction, additional studies on human adult stem cell biology appear to be particularly essential because of the interspecies differences that may influence their capacity to give rise to the specific cell lineages under well-defined conditions in vitro and regenerate a particular tissue in vivo. In addition, the optimization of purification and delivery methods of stem cells and their progenitors appear generally to be important before their possible use under safe conditions in cellular therapies. More specifically, the establishment of the procedures of derivation from EB-derived progenitor cells in culture for the elimination of residual pluripotent and undifferentiated hESCs, which are able to form the teratomas or teratocarcinomas in vivo, is necessary before they are used in transplantation for diverse degenerative disorders in humans. In addition, further work is important to identify the specific biomarkers of adult stem cells and their progenitors, as well as the microenvironmental factors that might induce their self-renewal, maturation, and migration during each stage, leading to tissue regeneration, and during their relocation to other distant damaged tissues. These future studies should elucidate the sequential cellular events that are implicated in the maintenance of tissue homeostasis, thereby establishing the deregulated signaling that may contribute to diverse disorders and cancer types. Furthermore, since certain lines of evidence have also indicated that certain adult stem cells might trans-differentiate into mature and specialized cells from other tissue types under specific culture conditions in vitro and after transplantation in vivo, it will be important to establish more precisely the factors influencing their incorporation in healthy and damaged tissues/organs in vivo.
* c% K+ r$ Y3 Z9 e
9 w5 \/ {' V; L4 W( [Several similarities appear to exist between the behavior of adult stem cells and the cancer progenitor cells, including their high capacity to self-renew and give rise to a heterogeneous population of more differentiated cells and their high capacity to migrate at distant sites, where they may establish their novel homing through interactions with their new microenvironment. Therefore, the knowledge that will be acquired on the adult stem cell biology under physiological and pathological conditions should aid particularly in the design of new cancer therapies targeting cancer progenitor cells. In this matter, it will be important to establish the specific oncogenic alterations occurring in cancer progenitor cells versus their more differentiated progenitors within the global tumor cell population during localized tumor development and dissemination to other distant tissues. Additional analyses of possible interactions between diverse developmental signaling (such as hormones, EGFR, hedgehog, Wnt/ß-catenin, and Notch pathways that are involved in the regeneration of numerous tissues and frequently activated during the malignant transformation of adult stem cells into cancer progenitor cells) are notably of particular interest. The possibility that the pharmacological agents acting on the cancer progenitor cells may also alter the behaviors of normal stem cells also emphasizes the importance of using these types of agents at lower doses in combination with conventional cancer treatments.
% T! T( S. j- Z% l& [4 ~$ w- w1 X4 j# d0 }
Altogether, these future works should establish molecular changes occurring in adult stem cells and their progenitors during tissue repair and etiopathogenesis. Hence, these further studies could lead to the development of more effective treatments for numerous genetic and degenerative disorders by cell replacement. Moreover, the identification of specific markers and targeting distinct tumorigenic cascades in cancer progenitor cells should also contribute to developing novel early detection methods and combination therapies for diverse aggressive and lethal cancers derived from the malignant transformation of adult stem cells.
$ }- i' G U2 Q% K5 H" E4 b$ r# i/ p R) u# E
DISCLOSURES9 s( m' C: ^8 F' n6 |( F
% ]( Y9 V( X0 s) } c4 z/ ~
The authors indicate no potential conflicts of interest.1 M* B. {. l( o% v
# J, x6 n. j- a9 w( y# U
ACKNOWLEDGMENTS
0 ]* r( [$ n U+ n5 i' p' ?
, L; k7 O" x* W W" zThis work was supported by U.S. Department of Defense Grants PC040493, PC04502, and OC04110 and NIH Grants CA78590, CA72712, and CA111294. We thank Kristi L. Berger for editing the manuscript.
7 w) }' d5 m2 T1 D 【参考文献】' E" O. t; ] t* G7 ]
) H( d& Q9 Z M
^% C0 ^5 Q7 J; h6 p2 [: |
Brittan M, Wright NA. Gastrointestinal stem cells. J Pathol 2002;197:492¨C509.% b1 K9 ]# V# _% p) Q
" T" F M" r* ]3 z, {Modlin IM, Kidd M, Lye KD et al. Gastric stem cells: An update. Keio J Med 2003;52:134¨C137.
. h8 W, h% d. S2 W" S$ _ ?
' U! n3 P$ Z {3 {- A6 xLavker RM, Tseng SC, Sun TT. Corneal epithelial stem cells at the limbus: Looking at some old problems from a new angle. Exp Eye Res 2004;78:433¨C446.
) V; c, t( ?3 j7 _( S* b2 ^1 ?: H. r/ f+ D8 s; H
Tumbar T, Guasch G, Greco V et al. Defining the epithelial stem cell niche in skin. Science 2004;303:359¨C363.
7 U' u; h k+ o
5 r" ~' c' i6 C& K0 S0 l# M; G$ YReya T, Clevers H. Wnt signalling in stem cells and cancer. Nature 2005;434:843¨C850.
* Z: n+ r% f8 Z `/ U4 J
, |7 |5 O6 `0 P4 e. _' Q& [Watts C, McConkey H, Anderson L et al. Anatomical perspectives on adult neural stem cells. J Anat 2005;207:197¨C208.
2 k- v- L/ [+ Q. e! R0 R. ^/ @' z, Q: N
Woodward WA, Chen MS, Behbod F et al. On mammary stem cells. J Cell Sci 2005;118:3585¨C3594.1 K9 I7 u& X5 }
; `! Q" y- T- b; e/ M8 q1 _- J' s( k
Liu S, Dontu G, Wicha MS. Mammary stem cells, self-renewal pathways, and carcinogenesis. Breast Cancer Res 2005;7:86¨C95.
. f( d, M8 R3 ?6 }7 @6 y1 z) C; A
2 y$ n r, p7 U! H9 M2 SKim CF, Jackson EL, Woolfenden AE et al. Identification of bronchioalveolar stem cells in normal lung and lung cancer. Cell 2005;121:823¨C835.( a6 z/ [! W) f; X8 p- M
/ r/ j7 P+ _; Q- sLi L, Xie T. Stem cell niche: Structure and function. Annu Rev Cell Dev Biol 2005;21:605¨C631.
: I: D# E& t) @9 I8 P8 A( O
. [; N2 p# @5 v; WGuettier C. Which stem cells for adult liver? Ann Pathol 2005;25:33¨C44.
7 D- ?2 w0 E: P% z5 Y2 Y
' g. y+ r# D4 K6 ^) N' \Heissig B, Ohki Y, Sato Y et al. A role for niches in hematopoietic cell development. Hematology 2005;10:247¨C253.
! j" o5 Q/ e& ]; r$ y9 r1 J1 v' @! |5 s7 L! B! \
Leri A, Kajstura J, Anversa P et al. Cardiac stem cells and mechanisms of myocardial regeneration. Physiol Rev 2005;85:1373¨C1416.
9 A# H- T: e1 r6 u
- r: x6 Q! C3 j3 UOrtiz-Gonzalez XR, Keene CD, Verfaillie CM et al. Neural induction of adult bone marrow and umbilical cord stem cells. Curr Neurovasc Res 2004;1:207¨C213.
1 t+ H8 C6 [, j" }5 ?
& s6 y$ _3 d0 V; NRollini P, Kaiser S, Faes-van't Hull E et al. Long-term expansion of transplantable human fetal liver hematopoietic stem cells. Blood 2004;103:1166¨C1170.2 f' k b) a1 W$ i3 X8 s& a
. k. Y7 ~- f/ K& zLindvall O, Kokaia Z, Martinez-Serrano A. Stem cell therapy for human neurodegenerative disorders-how to make it work. Nat Med 2004;10 (suppl):S42¨CS50.* B! P& `& ?. _$ g& R. c
p( g5 j, k4 X- W- L- p) b
Perillo A, Bonanno G, Pierelli L et al. Stem cells in gynecology and obstetrics. Panminerva Med 2004;46:49¨C59.! r1 U2 k7 Z6 c7 q- z2 ?" v
. S+ j! O2 H- L1 A' e3 n! j( ]Mimeault M, Batra SK. Recent advances on multiple tumorigenic cascades involved in prostatic cancer progression and targeting therapies. Carcinogenesis 2006;27:1¨C22.) `1 B" O3 d a$ n1 J9 z
: u6 A4 c6 q/ P0 `# r; I
Bonner-Weir S, Weir GC. New sources of pancreatic beta-cells. Nat Biotechnol 2005;23:857¨C861.
8 ^# d: G! e9 [7 f( X; z: o e+ E/ T: r
Sanmano B, Mizoguchi M, Suga Y et al. Engraftment of umbilical cord epithelial cells in athymic mice: In an attempt to improve reconstructed skin equivalents used as epithelial composite. J Dermatol Sci 2005;37:29¨C39.
( H9 K; ~. n3 q! R: [
) y }; D! `. B) v9 bYao S, Chen S, Clark J et al. Long-term self-renewal and directed differentiation of human embryonic stem cells in chemically defined conditions. Proc Natl Acad Sci U S A 2006;103:6907¨C6912.0 w; p& e6 F3 r7 M/ K4 t) j
; O6 h" `/ z; I4 c5 ^. ~Trounson A. The production and directed differentiation of human embryonic stem cells. Endocr Rev 2006;27:208¨C219.6 I5 L" K6 p! q5 ~ w
1 @# M6 Q5 o9 y9 NErices AA, Allers CI, Conget PA et al. Human cord blood-derived mesenchymal stem cells home and survive in the marrow of immunodeficient mice after systemic infusion. Cell Transplant 2003;12:555¨C561.
5 v. ]1 [3 r" t8 g4 x
8 Y: R7 g. `% Q8 O cRoche E, Reig JA, Campos A et al. Insulin-secreting cells derived from stem cells: Clinical perspectives, hypes and hopes. Transpl Immunol 2005;15:113¨C129.
8 @3 q0 F3 ?5 Q" x8 `: c* n* L9 g7 W9 h2 \6 M
Oe H, Kaido T, Mori A et al. Hepatocyte growth factor as well as vascular endothelial growth factor gene induction effectively promotes liver regeneration after hepatectomy in Solt-Farber rats. Hepatogastroenterology 2005;52:1393¨C1397.; u2 [9 G# Z* E R+ v
4 ~' I! k: ]; FSuen PM, Leung PS. Pancreatic stem cells: A glimmer of hope for diabetes? JOP 2005;6:422¨C424.
6 P: o; C' G! E$ K9 y' b0 ?* J
5 L. w3 }$ ^1 {9 q3 t: ^0 y, ~* r- w. PCohen Y, Nagler A. Umbilical cord blood transplantation¡ªhow, when and for whom? Blood Rev 2004;18:167¨C179.' M% o; o3 @; `9 B: B
/ F0 M! x/ Y1 e" H3 ~+ ^% z8 o
Giuliano CJ, Kerley-Hamilton JS, Bee T et al. Retinoic acid represses a cassette of candidate pluripotency chromosome 12p genes during induced loss of human embryonal carcinoma tumorigenicity. Biochim Biophys Acta 2005;1731:48¨C56.* k$ }- K, M9 _) i2 F- I
; _$ ]. t- s) G; E [, ~
Baik I, Devito WJ, Ballen K et al. Association of fetal hormone levels with stem cell potential: Evidence for early life roots of human cancer. Cancer Res 2005;65:358¨C363.
3 K! z3 Y, l. h' Q! G3 [$ \$ R; V$ [% g! J) f! m
Fang D, Nguyen TK, Leishear K et al. A tumorigenic subpopulation with stem cell properties in melanomas. Cancer Res 2005;65:9328¨C9337.5 M: o7 p6 b' ~% S# n3 S
0 P, |2 |2 U. G" j
Berns A. Stem cells for lung cancer? Cell 2005;121:811¨C813.4 Y ]6 a8 b. c" k" Q
y x7 X& Z3 q, x* g
Schier S, Wright NA. Stem cell relationships and the origin of gastrointestinal cancer. Oncology 2005;69:9¨C13.$ F6 U: U( \" P" r& C/ d6 G
5 g! s3 n" a( [7 S5 L) VBeachy PA, Karhadkar SS, Berman DM. Tissue repair and stem cell renewal in carcinogenesis. Nature 2004;432:324¨C331.' I% C' J; x. T- Y+ ]- T2 _1 b
: ?0 r5 L- M& _& SBapat SA, Mali AM, Koppikar CB et al. Stem and progenitor-like cells contribute to the aggressive behavior of human epithelial ovarian cancer. Cancer Res 2005;65:3025¨C3029.
& H+ `, N9 E# p' i$ q; k, l; ` {) Y0 u# ]6 |
Mimeault M, Bonenfant D, Batra SK. New advances on the functions of epidermal growth factor receptor and ceramides in skin cell differentiation, disorders and cancers. Skin Pharmacol Physiol 2005;17:153¨C166.( I2 e6 c" K9 F
' L& K: f. S5 H2 G" P* Q% v$ f
Mimeault M, Brand RE, Sasson AA et al. Recent advances on the molecular mechanisms involved in pancreatic cancer progression and therapies. Pancreas 2005;31:301¨C316.2 p; H, p0 {7 Q' L7 r; a
0 F0 }5 a Y ]Wang JC, Dick JE. Cancer stem cells: Lessons from leukemia. Trends Cell Biol 2005;15:494¨C501.
6 e" v P0 p( S% H
( F& v0 B* A* c7 n) w" X) b3 B2 zGalderisi U, Cipollaro M, Giordano A. Stem cells and brain cancer. Cell Death Differ 2006;13:5¨C11.
" i5 k5 \' m$ T8 d( u: ~& L
# }- ^& z* {+ \/ [1 J1 a8 i8 YLiu X, Driskell RR, Engelhardt JF. Airway glandular development and stem cells. Curr Top Dev Biol 2004;64:33¨C56.
. ~+ N3 x3 Z& U3 }1 v) B9 ~8 u0 m6 [5 C' {2 P
Seaberg RM, Smukler SR, Kieffer TJ et al. Clonal identification of multipotent precursors from adult mouse pancreas that generate neural and pancreatic lineages. Nat Biotechnol 2004;22:1115¨C1124.
. A a* d* X8 H: s* L7 e+ W3 }9 Q# X! w. W( A
Griffiths MJ, Bonnet D, Janes SM. Stem cells of the alveolar epithelium. Lancet 2005;366:249¨C260." Z' q" ] s1 V$ A% a
- @* C3 |1 G9 r
Schuldiner M, Yanuka O, Itskovitz-Eldor J et al. Effects of eight growth factors on the differentiation of cells derived from human embryonic stem cells. Proc Natl Acad Sci U S A 2002;97:11307¨C11312.
5 ^% T5 Q, M3 r, V! t) a3 p; W7 I1 M% U; E1 A
D'Amour KA, Agulnick AD, Eliazer S et al. Efficient differentiation of human embryonic stem cells to definitive endoderm. Nat Biotechnol 2005;23:1534¨C1541.
: ?) n1 i1 R& ?( B( {: M! k7 A$ y2 k! X6 S1 ^# D
Richards M, Tan SP, Tan JH et al. The transcriptome profile of human embryonic stem cells as defined by SAGE. STEM CELLS 2004;22:51¨C64.% _, r$ [8 ]8 z# m& D% P2 O- E
- ] ~% H" u& ~! B+ I T4 j
Chambers I, Smith A. Self-renewal of teratocarcinoma and embryonic stem cells. Oncogene 2004;23:7150¨C7160.( ^% |% r' ?8 B. i9 F
* [" n1 [2 n% g* D( H* H9 fZeng X, Cai J, Chen J et al. Dopaminergic differentiation of human embryonic stem cells. STEM CELLS 2004;22:925¨C940.
# U# B! e. p5 H2 {4 y# r' X& D8 `" d1 O7 S0 Q4 I
Brolen GK, Heins N, Edsbagge J et al. Signals from the embryonic mouse pancreas induce differentiation of human embryonic stem cells into insulin-producing beta-cell-like cells. Diabetes 2005;54:2867¨C2874.
! d; ]* R; z0 i' w3 J2 o6 u: _" c, r0 U" |2 A9 w
Banin E, Obolensky A, Idelson M et al. Retinal incorporation and differentiation of neural precursors derived from human embryonic stem cells. STEM CELLS 2006;24:246¨C257.; s/ k; r4 @2 q1 X8 @$ S' S6 e& E
, G e5 ~- z4 U. r4 W$ b
Karp JM, Ferreira LS, Khademhosseini A et al. Cultivation of human embryonic stem cells without the embryoid body step enhances osteogenesis in vitro. STEM CELLS 2006;24:835¨C843.6 P: R v+ Z0 l2 `6 @- e
5 U1 j. ~5 w2 @, g# Z# ]
Zambidis ET, Peault B, Park TS et al. Hematopoietic differentiation of human embryonic stem cells progresses through sequential hematoendothelial, primitive, and definitive stages resembling human yolk sac development. Blood 2005;106:860¨C870.
8 M4 J! Z% k/ Y2 P' ^& {8 ^) @: r$ n9 L0 I! H7 q0 g; K
Arnhold S, Klein H, Semkova I et al. Neurally selected embryonic stem cells induce tumor formation after long-term survival following engraftment into the subretinal space. Invest Ophthalmol Vis Sci 2004;45:4251¨C4255.
( F" r- ^/ Z. L* k# O8 G; V5 k
' e! \: x' @8 W/ s2 }' h4 _/ [* _Andrews PW, Matin MM, Bahrami AR et al. Embryonic stem (ES) cells and embryonal carcinoma (EC) cells: Opposite sides of the same coin. Biochem Soc Trans 2005;33:1526¨C1530.2 u# J0 n1 ]1 f1 @. p" x
- [% G: u# ]3 K; @0 ]4 ^' ~/ lFujikawa T, Oh SH, Pi L et al. Teratoma formation leads to failure of treatment for type I diabetes using embryonic stem cell-derived insulin-producing cells. Am J Pathol 2005;166:1781¨C1791.
1 ~2 F0 c% v# I% T6 c
3 S: S, A3 X" y. d% y4 J( r$ MBieberich E, Silva J, Wang G et al. Selective apoptosis of pluripotent mouse and human stem cells by novel ceramide analogues prevents teratoma formation and enriches for neural precursors in ES cell-derived neural transplants. J Cell Biol 2004;167:723¨C734.) G* X& r* G2 a4 Y v
. C* [' |+ B3 s5 u9 A0 _( I
Kumashiro Y, Asahina K, Ozeki R et al. Enrichment of hepatocytes differentiated from mouse embryonic stem cells as a transplantable source. Transplantation 2005;79:550¨C557.6 Q/ d$ t6 b7 D6 N
# i7 R0 g4 R/ M( iPrzyborski SA, Christie VB, Hayman MW et al. Human embryonal carcinoma stem cells: Models of embryonic development in humans. Stem Cells Dev 2004;13:400¨C408.
) v5 M: H1 o- n! n
1 U- l. T' U$ C" D& T+ r |9 A1 w. WTamagawa T, Ishiwata I, Saito S. Establishment and characterization of a pluripotent stem cell line derived from human amniotic membranes and initiation of germ layers in vitro. Hum Cell 2004;17:125¨C130.
( D6 ?. E, F: e9 h( P' h
: F; ?; A- P* t4 {& \$ ^Miki T, Lehmann T, Cai H et al. Stem cell characteristics of amniotic epithelial cells. STEM CELLS 2005;23:1549¨C1559.
7 \: D* [ L4 v) d
" [- I" |; E! S- g) RTarasenko YI, Yu Y, Jordan PM et al. Effect of growth factors on proliferation and phenotypic differentiation of human fetal neural stem cells. J Neurosci Res 2004;78:625¨C636.# o1 v. N" b3 J( T9 N2 ?- y( g( D
5 }" b0 c0 E; I1 |
Bianchi DW, Romero R. Biological implications of bi-directional fetomaternal cell traffic: A summary of a National Institute of Child Health and Human Development-sponsored conference. J Matern Fetal Neonatal Med 2003;14:123¨C129.
3 o1 L+ {1 G* O( @& ^2 G! U+ K# M( p6 e; [ |" e, Z
Tan XW, Liao H, Sun L et al. Fetal microchimerism in the maternal mouse brain: A novel population of fetal progenitor or stem cells able to cross the blood-brain barrier? STEM CELLS 2005;23:1443¨C1452.& a/ z) @+ z& ]$ E) H. h4 T. P4 z* i5 T
& W* ^, n; @; H* x" p
Khosrotehrani K, Bianchi DW. Multi-lineage potential of fetal cells in maternal tissue: A legacy in reverse. J Cell Sci 2005;118:1559¨C1563." I1 _% O! |2 L! i% b2 d
. ` {- j1 ~' ?4 ] I5 }: L' B/ zIshikawa F, Drake CJ, Yang S et al. Transplanted human cord blood cells give rise to hepatocytes in engrafted mice. Ann NY Acad Sci 2003;996:174¨C185.
8 n# H9 m0 {; D, z( S3 T, m( @$ N$ ^) \6 y1 q4 E0 E' N; i! T( d
Tanavde VM, Malehorn MT, Lumkul R et al. Human stem-progenitor cells from neonatal cord blood have greater hematopoietic expansion capacity than those from mobilized adult blood. Exp Hematol 2002;30:816¨C823.
0 [5 H2 k8 }5 W% n' y
- M. `% `2 D1 @5 Q. B! v5 RLian SM, Wang XB, Xue ZG et al. Differentiation and increase of dendritic cells from umbilical cord blood in vitro. Zhongguo Shi Yan Xue Ye Xue Za Zhi 2004;12:615¨C619.- A o6 `% L. b5 P9 a6 D0 Y! y* u2 P
" Q1 n( W" s* n1 _
Pessina A, Eletti B, Croera C et al. Pancreas developing markers expressed on human mononucleated umbilical cord blood cells. Biochem Biophys Res Commun 2004;323:315¨C322.
( L) e2 n! S' G* }4 b" t1 l; I. t" d' u, |5 O7 N8 C
Chang YJ, Shih DT, Tseng CP et al. Disparate mesenchyme-lineage tendencies in mesenchymal stem cells from human bone marrow and umbilical cord blood. STEM CELLS 2006;24:679¨C685.
" v' ]1 x ^: @. u0 I3 Q: f( r0 X+ F) k" H5 k! M% b
Blau HM, Brazelton TR, Weimann JM. The evolving concept of a stem cell: Entity or function? Cell 2001;105:829¨C841.$ g# ]# d0 Q( _0 f
7 m( l* E5 b& C- l x7 vArai F, Hirao A, Ohmura M et al. Tie2/angiopoietin-1 signaling regulates hematopoietic stem cell quiescence in the bone marrow niche. Cell 2004;118:149¨C161.
- K4 T8 c; L/ e2 d, ~
. [; [) a$ h2 F7 ^8 l- gFuchs E, Tumbar T, Guasch G. Socializing with the neighbors: Stem cells and their niche. Cell 2004;116:769¨C778.
! S8 @% E2 q i
* o! b% R4 g# [* G# o. N0 b, {& sHirao A, Arai F, Suda T. Regulation of cell cycle in hematopoietic stem cells by the niche. Cell Cycle 2004;3:1481¨C1483. i1 f2 t m6 ?: _. E, ]. \ H& ^- ?
7 Q' b9 p, D2 I4 T a
Hope KJ, Jin L, Dick JE. Acute myeloid leukemia originates from a hierarchy of leukemic stem cell classes that differ in self-renewal capacity. Nat Immunol 2004;5:738¨C743.. z q# R' ?9 C6 V" j+ X( ^% G2 H
% O' h" J5 s2 L0 l. {9 D
Wilson A, Trumpp A. Bone-marrow haematopoietic-stem-cell niches. Nat Rev Immunol 2006;6:93¨C106.% t+ X% U$ ]. E3 y
* k: W) T4 ~2 {5 oKopen GC, Prockop DJ, Phinney DG. Marrow stromal cells migrate throughout forebrain and cerebellum, and they differentiate into astrocytes after injection into neonatal mouse brains. Proc Natl Acad Sci U S A 1999;96:10711¨C10716.
5 ~ H7 ?* q4 @% m j3 U
$ G5 e- W/ T3 r3 SJiang Y, Jahagirdar BN, Reinhardt RL et al. Pluripotency of mesenchymal stem cells derived from adult marrow. Nature 2002;418:41¨C49.
2 j/ b' O: w' i8 m, C3 R# ]: e% ]7 p! L: X3 x i9 c
de Vries EG, Vellenga E, Kluin-Nelemans JC et al. The happy destiny of frozen haematopoietic stem cells: From immature stem cells to mature applications. Eur J Cancer 2004;40:1987¨C1992.
! k: s$ o8 S! V; b2 f+ W) n/ W
9 s9 _8 K0 t8 o8 j# \: YShelton JG, Steelman LS, Abrams SL et al. The epidermal growth factor receptor gene family as a target for therapeutic intervention in numerous cancers: What's genetics got to do with it? Expert Opin Ther Targets 2005;9:1009¨C1030.
" d# `6 b3 L4 h$ H! H* h( O4 ~. d4 M; B6 }
Bouwens L, Rooman I. Regulation of pancreatic beta-cell mass. Physiol Rev 2005;85:1255¨C1270.
2 S6 A: C, ?; ?2 D9 a* w5 R3 y; d( i
. K* P; i* `8 jZulewski H, Abraham EJ, Gerlach MJ et al. Multipotential nestin-positive stem cells isolated from adult pancreatic islets differentiate ex vitro into pancreatic endocrine, exocrine, and hepatic phenotypes. Diabetes 2001;50:521¨C533.0 A# Q$ i% e. \( h
: l B4 J* f# d; N! v4 ?8 xSaxena R, Theise N. Canals of Hering: Recent insights and current knowledge. Semin Liver Dis 2004;24:43¨C48.
# L; m- d, E' F! q+ r y7 U+ f7 h) O1 C
Mishra L, Shetty K, Tang Y et al. The role of TGF-beta and Wnt signaling in gastrointestinal stem cells and cancer. Oncogene 2005;24:5775¨C5789.1 p: y8 u9 S% f3 q$ F
5 K- P3 q) r& \' {( v2 ~5 s
Lagasse E, Connors H, Al-Dhalimy M et al. Purified hematopoietic stem cells can differentiate into hepatocytes in vivo. Nat Med 2000;6:1229¨C1234.* G6 E+ e2 ?& R
- Y; m- q Q n0 h- |0 pSmalley M, Ashworth A. Stem cells and breast cancer: A field in transit. Nat Rev Cancer 2003;3:832¨C844.
9 E1 n q5 F# C# V" w0 E' Q' ?, b$ C
Clayton H, Titley I, Vivanco M. Growth and differentiation of progenitor/stem cells derived from the human mammary gland. Exp Cell Res 2004;297:444¨C460.- J' q. B; t- r! q9 _4 v' ?
4 C5 w7 b$ U: y7 F! I4 ZDontu G, El-Ashry D, Wicha MS. Breast cancer, stem/progenitor cells and the estrogen receptor. Trends Endocrinol Metab 2004;15:193¨C197.
4 T$ J' q1 E, ?$ [* M& `. k' B" \1 L: I
Petersen OW, Gudjonsson T, Villadsen R et al. Epithelial progenitor cell lines as models of normal breast morphogenesis and neoplasia. Cell Prolif 2003;36:33¨C44.
6 O9 `/ y) K# a9 Q! @
1 e4 s0 N' M; u1 TClarke RB, Spence K, Anderson E et al. A putative human breast stem cell population is enriched for steroid receptor-positive cells. Dev Biol 2005;277:443¨C456.
# M( u9 l: a+ Y8 s3 f3 m* D0 e6 e G+ L
Dontu G, Abdallah WM, Foley JM et al. In vitro propagation and transcriptional profiling of human mammary stem/progenitor cells. Genes Dev 2003;17:1253¨C1270.
% y- w3 ^, g: n9 T+ P, u/ x/ l0 s2 U& H, @! }/ |$ s( r& l4 S* N
Burger PE, Xiong X, Coetzee S et al. Sca-1 expression identifies stem cells in the proximal region of prostatic ducts with high capacity to reconstitute prostatic tissue. Proc Natl Acad Sci U S A 2005;102:7180¨C7185.
" L. S) j3 {2 f1 f, H$ E. t9 u1 a" f) x/ _ Z: {8 b
Xin L, Lawson DA, Witte ON. The Sca-1 cell surface marker enriches for a prostate-regenerating cell subpopulation that can initiate prostate tumorigenesis. Proc Natl Acad Sci U S A 2005;102:6942¨C6947.0 O7 }0 `9 V+ X; [6 k
' V6 J: x" Y X# G& H+ j( MAl-Hajj M, Wicha MS, Ito-Hernandez A et al. Prospective identification of tumorigenic breast cancer cells. Proc Natl Acad Sci U S A 2003;100:3983¨C3988.
/ b! Q: d ]8 E, I$ n/ f. q. u% u e& r( O: `. y' V
Kurbel S. Selective reduction of estrogen receptor (ER) positive breast cancer occurrence by estrogen receptor modulators supports etiological distinction between ER positive and ER negative breast cancers. Med Hypotheses 2005;64:1182¨C1187." b. b# I3 ?4 e
" ~* D6 ^) z3 J# }8 `: V
Kanatsu-Shinohara M, Inoue K, Lee J et al. Generation of pluripotent stem cells from neonatal mouse testis. Cell 2004;119:1001¨C1012.
, @4 l- H/ n1 u
7 r$ P5 i. C! ?/ u0 n Z8 [3 `von Schalburg KR, McCarthy SP, Rise ML et al. Expression of morphogenic genes in mature ovarian and testicular tissues: Potential stem-cell niche markers and patterning factors. Mol Reprod Dev 2005;73:142¨C152.
# x, J# @1 }+ Y& A" b0 r1 c' Y5 t+ l
4 c( I. G7 p- G; D8 _9 ^Bukovsky A, Caudle MR, Svetlikova M et al. Oogenesis in adult mammals, including humans: A review. Endocrine 2005;26:301¨C316.1 H( @' \! ?- y+ A9 p
- d2 R- |- c5 }4 @8 e+ {
Bukovsky A, Svetlikova M, Caudle MR. Oogenesis in cultures derived from adult human ovaries. Reprod Biol Endocrinol 2005;3:17.4 \1 A" X& p" K8 v
: Y% ], O" x6 BNeiva K, Sun YX, Taichman RS. The role of osteoblasts in regulating hematopoietic stem cell activity and tumor metastasis. Braz J Med Biol Res 2005;38:1449¨C1454.
2 G( ?& \. {, t9 ]" `& i0 }, Q2 x' G. P0 I
Maillard I, Fang T, Pear WS. Regulation of lymphoid development, differentiation, and function by the Notch pathway. Annu Rev Immunol 2005;23:945¨C974.
2 f& B, k4 J; b& n, D0 M g
1 o Z3 l* ^0 I9 @1 UMurphy MJ, Wilson A, Trumpp A. More than just proliferation: Myc function in stem cells. Trends Cell Biol 2005;15:128¨C137.
7 U8 c, S( s: f' g; ~; V
: @8 P9 K% ?8 o" qKondo M, Scherer DC, Miyamoto T et al. Cell-fate conversion of lymphoid-committed progenitors by instructive actions of cytokines. Nature 2000;407:383¨C386.
# ^; m) I$ { V" a ^
2 A$ y* D/ `9 M: I$ h( [Schatteman GC, Awad O. Hemangioblasts, angioblasts, and adult endothelial cell progenitors. Anat Rec A Discov Mol Cell Evol Biol 2004;276:13¨C21.% s& ?1 d7 ]& ]4 C3 ^# U/ T
9 Z) ]+ E( }+ v3 z9 B+ o% o2 O9 nTavian M, Zheng B, Oberlin E et al. The vascular wall as a source of stem cells. Ann NY Acad Sci 2005;1044:41¨C50.& o9 p' e" K1 y0 r9 a$ W
$ G7 c, W9 j( N1 @3 bKiel MJ, Yilmaz OH, Iwashita T et al. SLAM family receptors distinguish hematopoietic stem and progenitor cells and reveal endothelial niches for stem cells. Cell 2005;121:1109¨C1121.
8 R }9 c7 `! {( g* f$ o; t3 \& H$ g. @6 @0 w, m
Papayannopoulou T, Craddock C. Homing and trafficking of hemopoietic progenitor cells. Acta Haematol 1997;97:97¨C104.
5 y: B: ?' B$ j/ a# P" U3 q5 y! Z& b" j. P
Vermeulen M, Le Pesteur F, Gagnerault MC et al. Role of adhesion molecules in the homing and mobilization of murine hematopoietic stem and progenitor cells. Blood 1998;92:894¨C900.: t1 P' p9 ^& I! n, ]1 U" @/ S# ?$ F
* l H/ t& O! o, l. ^% }- T
Ueda T, Tsuji K, Yoshino H et al. Expansion of human NOD/SCID-repopulating cells by stem cell factor, Flk2/Flt3 ligand, thrombopoietin, IL-6, and soluble IL-6 receptor. J Clin Invest 2000;105:1013¨C1021.
8 X) @" R# t, W
6 b& u6 h- u2 B S% c9 {Gammaitoni L, Bruno S, Sanavio F et al. Ex vivo expansion of human adult stem cells capable of primary and secondary hemopoietic reconstitution. Exp Hematol 2003;31:261¨C270.
1 a9 n& @/ O0 p$ ?1 h" f, _) Q/ F6 N+ k9 J# x% D' O; [
Wang J, Zhao HP, Lin G et al. In vitro hematopoietic differentiation of human embryonic stem cells induced by co-culture with human bone marrow stromal cells and low dose cytokines. Cell Biol Int 2005;29:654¨C661.1 @6 P0 l( q: {0 l0 K
8 @7 o' @1 v' E. c* b1 D- vGeorgantas RW III, Tanadve V, Malehorn M et al. Microarray and serial analysis of gene expression analyses identify known and novel transcripts overexpressed in hematopoietic stem cells. Cancer Res 2004;64:4434¨C4441.- _4 m8 l2 d* y) \% s
# c% S* P* i! ?# I! x5 d
Shi S, Gronthos S. Perivascular niche of postnatal mesenchymal stem cells in human bone marrow and dental pulp. J Bone Miner Res 2003;18:696¨C704.
/ D+ T1 a* Z$ T% n' G/ _$ c; T, ?$ }3 i3 ]( |
Reyes M, Lund T, Lenvik T et al. Purification and ex vivo expansion of postnatal human marrow mesodermal progenitor cells. Blood 2001;98:2615¨C2625., z/ y+ `! K( B( W
1 W$ t* }0 V$ h/ h* m j$ l
Pittenger MF, Mackay AM, Beck SC et al. Multilineage potential of adult human mesenchymal stem cells. Science 1999;284:143¨C147.
5 u! u; m) D1 `0 I! Q
& R0 M: ~% K& O' g5 ?Sanchez-Ramos J, Song S, Cardozo-Pelaez F et al. Adult bone marrow stromal cells differentiate into neural cells in vitro. Exp Neurol 2000;164:247¨C256.% g2 z- _% y0 k" y0 x% C
% |3 R1 J3 |- W' DHanada K, Dennis JE, Caplan AI. Stimulatory effects of basic fibroblast growth factor and bone morphogenetic protein-2 on osteogenic differentiation of rat bone marrow-derived mesenchymal stem cells. J Bone Miner Res 1997;12:1606¨C1614.0 G. e9 x5 E; ]+ K
: c# d4 X1 B* H( [: j
Krampera M, Pasini A, Rigo A et al. HB-EGF/HER-1 signaling in bone marrow mesenchymal stem cells: Inducing cell expansion and reversibly preventing multilineage differentiation. Blood 2005;106:59¨C66.
9 Z; {+ t7 F. d2 J, B& X$ @& h8 x5 y5 O8 b, a
Calvi LM, Adams GB, Weibrecht KW et al. Osteoblastic cells regulate the haematopoietic stem cell niche. Nature 2003;425:841¨C846.6 w2 U8 @! M5 M" V9 q) B' n
, G* Q& U! Y# i- ^9 B. y. BPeled A, Grabovsky V, Habler L et al. The chemokine SDF-1 stimulates integrin-mediated arrest of CD34( ) cells on vascular endothelium under shear flow. J Clin Invest 1999;104:1199¨C1211.( ]& E/ o. m' w5 v$ w
) V3 c, _" f8 K4 z6 T* Q' x
De Clercq E. Potential clinical applications of the CXCR4 antagonist bicyclam AMD3100. Mini Rev Med Chem 2005;5:805¨C824.
& m1 O( B, X- M- E2 j- m- y& h) X+ c8 ]/ i8 R6 ]
Beltrami AP, Barlucchi L, Torella D et al. Adult cardiac stem cells are multipotent and support myocardial regeneration. Cell 2003;114:763¨C776.9 z+ z$ X! k; G9 Q1 i3 `
f6 ?0 q& P# x& K( q3 ]
Urbanek K, Rota M, Cascapera S et al. Cardiac stem cells possess growth factor-receptor systems that after activation regenerate the infarcted myocardium, improving ventricular function and long-term survival. Circ Res 2005;97:663¨C673.
/ t4 O! a, l+ O* T; ]3 y1 V* _9 Z. G- d2 S$ ^- N0 q
Tomita Y, Matsumura K, Wakamatsu Y et al. Cardiac neural crest cells contribute to the dormant multipotent stem cell in the mammalian heart. J Cell Biol 2005;170:1135¨C1146.8 j: Y- d, E+ O! r6 k
) T8 ?8 }1 R6 E9 bLillien L, Raphael H. BMP and FGF regulate the development of EGF-responsive neural progenitor cells. Development 2000;127:4993¨C5005.
5 l6 R1 u. d* K" q8 o! M& p
1 } W; w/ g- [2 JPanchision DM, Pickel JM, Studer L et al. Sequential actions of BMP receptors control neural precursor cell production and fate. Genes Dev 2001;15:2094¨C2110.8 k5 O/ p0 B6 F \, o* @
( I& N s5 {- a7 o% y% |6 j" D7 r. b
Doetsch F. A niche for adult neural stem cells. Curr Opin Genet Dev 2003;13:543¨C550.
J+ t$ ]% ~7 y
, x9 J/ q/ i, U! @9 USeri B, Garcia-Verdugo JM, Collado-Morente L et al. Cell types, lineage, and architecture of the germinal zone in the adult dentate gyrus. J Comp Neurol 2004;478:359¨C378.# B4 Y1 ]8 l+ M6 m% Q: X0 W
t+ F1 b+ }6 k6 H: ]
Alvarez-Buylla A, Lim DA. For the long run: Maintaining germinal niches in the adult brain. Neuron 2004;41:683¨C686.4 [# a" t9 ?4 C: I
- A0 X- V) q% U% z) {7 j/ |$ r* C3 |Sanai N, Tramontin AD, Quinones-Hinojosa A et al. Unique astrocyte ribbon in adult human brain contains neural stem cells but lacks chain migration. Nature 2004;427:740¨C744. l* X6 F6 S' G) a% J& O3 {
# N: V" l% K4 q8 G6 a1 u9 J. o. t& q
Holmberg J, Armulik A, Senti KA et al. Ephrin-A2 reverse signaling negatively regulates neural progenitor proliferation and neurogenesis. Genes Dev 2005;19:462¨C471.; U; j, }+ C/ T7 o
! |, U3 M2 O, K3 O3 R' e% _# ]Campos LS. Beta1 integrins and neural stem cells: Making sense of the extracellular environment. Bioessays 2005;27:698¨C707.
* n0 c) [0 n2 a, V' j/ A
/ p/ w0 G0 z( {, x9 t( L' Y! vLie DC, Colamarino SA, Song HJ et al. Wnt signalling regulates adult hippocampal neurogenesis. Nature 2005;437:1370¨C1375.3 w7 `7 z3 |! h/ n* ~; e
; S, i( I4 @. F" w% s _3 |) O# S
Wong RW, Guillaud L. The role of epidermal growth factor and its receptors in mammalian CNS. Cytokine Growth Factor Rev 2004;15:147¨C156.
Z- L4 t6 r$ C& d- o$ L' [) K1 V! r }! z: z
Ciccolini F, Mandl C, Holzl-Wenig G et al. Prospective isolation of late development multipotent precursors whose migration is promoted by EGFR. Dev Biol 2005;284:112¨C125.- W) t0 |; F8 R+ C
9 Z9 ^& I* W% _
Palma V, Lim DA, Dahmane N et al. Sonic hedgehog controls stem cell behavior in the postnatal and adult brain. Development 2005;132:335¨C344.7 o8 i" N' \$ ?7 _( d
. S4 T7 o& D4 C+ l+ N+ N, RWu CJ, Qian X, O'Rourke DM. Sustained mitogen-activated protein kinase activation is induced by transforming erbB receptor complexes. DNA Cell Biol 1999;18:731¨C741.
" m$ M, ~: t) u5 ~5 r# B" d* c ^* B0 k W1 R% n" ~
Singh SK, Clarke ID, Terasaki M et al. Identification of a cancer stem cell in human brain tumors. Cancer Res 2003;63:5821¨C5828.' ?; s$ c/ q' Y2 E. H& |
4 A& j, M# G2 W3 JSchanzer A, Wachs FP, Wilhelm D et al. Direct stimulation of adult neural stem cells in vitro and neurogenesis in vivo by vascular endothelial growth factor. Brain Pathol 2004;14:237¨C248.
! v" P4 D) N! A6 ?" V% ]9 I- d F a; w
Liu IH, Chen SJ, Ku HH et al. Comparison of the proliferation and differentiation ability between adult rat retinal stem cells and cerebral cortex-derived neural stem cells. Ophthalmologica 2005;219:171¨C176.
% |: I/ S" u) Z+ b( ~
7 u: W0 O6 x X3 s+ SShen Q, Goderie SK, Jin L et al. Endothelial cells stimulate self-renewal and expand neurogenesis of neural stem cells. Science 2004;304:1338¨C1340.: _9 t9 x3 Y$ _5 B. C. \( g
1 \2 Q T- G% I( `! `9 s$ XMorris RJ, Liu Y, Marles L et al. Capturing and profiling adult hair follicle stem cells. Nat Biotechnol 2004;22:411¨C417.% } }3 N: @ y
4 o$ ^, Q1 |5 S; Z8 TBlanpain C, Lowry WE, Geoghegan A et al. Self-renewal, multipotency, and the existence of two cell populations within an epithelial stem cell niche. Cell 2004;118:635¨C648." e, D4 W0 F4 J+ L
- q5 _7 L" n) ~* k4 Y$ `0 W1 @7 c
Rendl M, Lewis L, Fuchs E. Molecular Dissection of Mesenchymal-Epithelial Interactions in the Hair Follicle. PLoS Biol 2005;3:1910¨C1924.
2 g+ i) Y& k+ S8 I6 O# C( R
! c. m4 z) p: J! e5 x( M* \Millar SE. An ideal society? Neighbors of diverse origins interact to create and maintain complex mini-organs in the skin. PLoS Biol 2005;3:1873¨C1877.
7 a: T' j+ a) d. B* {- @* b
8 `- I4 \/ V% v \! X! s, O) LLevy V, Lindon C, Harfe BD et al. Distinct stem cell populations regenerate the follicle and interfollicular epidermis. Dev Cell 2005;9:855¨C861.
. q% R) w9 M% N% e& u* K
7 A$ t3 \9 \' \5 Q/ L& TFlores I, Cayuela ML, Blasco MA. Effects of telomerase and telomere length on epidermal stem cell behavior. Science 2005;309:1253¨C1256.
9 s6 N1 R' T" |. t* {$ t8 O
0 o. V- N# V/ W* L. ASarin KY, Cheung P, Gilison D et al. Conditional telomerase induction causes proliferation of hair follicle stem cells. Nature 2005;436:1048¨C1052. g7 s/ O/ o" _) x, ~5 ^
* U: a# j! Y0 CVidal VP, Chaboissier MC, Lutzkendorf S et al. Sox9 is essential for outer root sheath differentiation and the formation of the hair stem cell compartment. Curr Biol 2005;15:1340¨C1351.
) D7 Y6 }% q0 v3 c+ R- z, i
g& D( T) _9 d6 d- X( z2 [+ Y+ c) }" D0 kSieber-Blum M, Grim M. The adult hair follicle: Cradle for pluripotent neural crest stem cells. Birth Defects Res C Embryo Today 2004;72:162¨C172.
4 Z. }# P; K7 D3 f9 ~0 D) x+ W, {1 _, V4 N7 R- g: b7 `
Kawase Y, Yanagi Y, Takato T et al. Characterization of multipotent adult stem cells from the skin: Transforming growth factor-beta (TGF-beta) facilitates cell growth. Exp Cell Res 2004;295:194¨C203.
5 u. x, v x7 b9 Q
/ O6 N e# K% OFernandes KJ, McKenzie IA, Mill P et al. A dermal niche for multipotent adult skin-derived precursor cells. Nat Cell Biol 2004;6:1082¨C1093.* i" ]+ B1 K8 x, T$ l" z
) y5 c, }9 t* G: I' i$ O, w" q) yBelicchi M, Pisati F, Lopa R et al. Human skin-derived stem cells migrate throughout forebrain and differentiate into astrocytes after injection into adult mouse brain. J Neurosci Res 2004;77:475¨C486.; [8 [" h7 S5 i2 b x# b" [
% ~" Y- z- y2 iLowell S, Jones P, Le R et al. Stimulation of human epidermal differentiation by delta-notch signalling at the boundaries of stem-cell clusters. Curr Biol 2000;10:491¨C500. n" P7 f5 Q) H) S: L! J# E3 l
$ X7 }0 }5 {$ ] Q! aWatt FM. Role of integrins in regulating epidermal adhesion, growth and differentiation. EMBO J 2002;21:3919¨C3926.. J2 \1 d2 P @# s; p/ m4 G) R
# J6 G* x7 J' F2 q1 \1 w. |$ hBenitah SA, Frye M, Glogauer M et al. Stem cell depletion through epidermal deletion of Rac1. Science 2005;309:933¨C935.
- Q! g0 R& W) k+ Z! y1 B6 l Y4 `9 q. ]2 u; T5 ]+ k. p+ d6 W7 `
Pasonen-Seppanen S, Karvinen S, Torronen K et al. EGF upregulates, whereas TGF-beta downregulates, the hyaluronan synthases Has2 and Has3 in organotypic keratinocyte cultures: Correlations with epidermal proliferation and differentiation. J Invest Dermatol 2003;120:1038¨C1044.) M" ~( c; K$ q/ w! u1 k8 B+ |
6 w- D3 h! r# D2 J9 e3 D5 d& O* J- _7 ^3 RPellegrini G, Dellambra E, Paterna P et al. Telomerase activity is sufficient to bypass replicative senescence in human limbal and conjunctival but not corneal keratinocytes. Eur J Cell Biol 2004;83:691¨C700." J- ?- N; z) P7 l: J! N$ ?
- V' y& }! d* H7 @Ang LP, Tan DT. Ocular surface stem cells and disease: Current concepts and clinical applications. Ann Acad Med Singapore 2004;33:576¨C580.
5 m7 X' [2 Z1 `: }" H: Z9 F0 o2 q0 K+ `2 [; c
Dua HS, Shanmuganathan VA, Powell-Richards AO et al. Limbal epithelial crypts: A novel anatomical structure and a putative limbal stem cell niche. Br J Ophthalmol 2005;89:529¨C532.
; u3 C4 o) Y) }1 G* Y- u' f; c
7 |8 t$ I7 c/ s0 A: ~# CChen Z, de Paiva CS, Luo L et al. Characterization of putative stem cell phenotype in human limbal epithelia. STEM CELLS 2004;22:355¨C366.
" m w- H0 a; M- y8 R6 ]2 p) s9 A/ u# `
) ^1 F% R& n; oZhao X, Das AV, Thoreson WB et al. Adult corneal limbal epithelium: A model for studying neural potential of non-neural stem cells per progenitors. Dev Biol 2002;250:317¨C331.' r( ^: i! f- Q, r0 R
! n* M2 O" s4 b. L+ X5 H
Whikehart DR, Parikh CH, Vaughn AV et al. Evidence suggesting the existence of stem cells for the human corneal endothelium. Mol Vis 2005;11:816¨C824.. [) V, `5 V+ O8 q: O
' h2 W* h" a3 }; _Ahmad I, Das AV, James J et al. Neural stem cells in the mammalian eye: Types and regulation. Semin Cell Dev Biol 2004;15:53¨C62.
9 N+ [ R/ [! K
: M! _; X5 z. R; r- ?Das AV, James J, Rahnenfuhrer J et al. Retinal properties and potential of the adult mammalian ciliary epithelium stem cells. Vision Res 2005;45:1653¨C1666.
8 W: {# ~; c+ w; }- P
! |# k1 n: q; f d/ UMoshiri A, Close J, Reh TA. Retinal stem cells and regeneration. Int J Dev Biol 2004;48:1003¨C1014.- z$ L" o f2 b, l# g" n1 g
0 }+ W& F* D+ {/ n
James J, Das AV, Rahnenfuhrer J et al. Cellular and molecular characterization of early and late retinal stem cells per progenitors: Differential regulation of proliferation and context dependent role of Notch signaling. J Neurobiol 2004;61:359¨C376.
) |9 ~: S7 R; ~" J* N! @ w( D- g8 s ?9 K' e$ d, |/ Y a5 Y$ |
Moshiri A, McGuire CR, Reh TA. Sonic hedgehog regulates proliferation of the retinal ciliary marginal zone in posthatch chicks. Dev Dyn 2005;233:66¨C75.
! k! L, h* ]: l6 ?; d2 E0 i" d8 f9 C
Sacco O, Silvestri M, Sabatini F et al. Epithelial cells and fibroblasts: Structural repair and remodelling in the airways. Paediatr Respir Rev 2004;5:S35¨CS40.8 Q1 }! K: @- K+ W3 X \" G
, Z+ n" o( a8 B" u2 |! V& `
Orlic D. BM stem cells and cardiac repair: Where do we stand in 2004? Cytotherapy 2005;7:3¨C15.1 P \; j0 N# m. w5 _$ H- ^1 H
6 n/ o1 i) R+ }5 o( G- q1 y
Vandervelde S, van Luyn MJ, Tio RA et al. Signaling factors in stem cell-mediated repair of infarcted myocardium. J Mol Cell Cardiol 2005;39:363¨C376.5 S! ^. z% c8 B( }' |/ U {
/ J T1 @! p& E% ^+ k' U
Hesdorffer C, Ayello J, Ward M et al. Phase I trial of retroviral-mediated transfer of the human MDR1 gene as marrow chemoprotection in patients undergoing high-dose chemotherapy and autologous stem-cell transplantation. J Clin Oncol 1998;16:165¨C172.: v$ M' R/ C. I3 m4 z
9 A# P5 S6 {( W. u) TKrebsbach PH, Zhang K, Malik AK et al. Bone marrow stromal cells as a genetic platform for systemic delivery of therapeutic proteins in vivo: Human factor IX model. J Gene Med 2003;5:11¨C17.
3 q% U. @( g" b! C- }% [
, _; A1 F; R, ]0 H' U9 w: X/ DKoh CJ, Atala A. Tissue engineering, stem cells, and cloning: Opportunities for regenerative medicine. J Am Soc Nephrol 2004;15:1113¨C1125.
7 ]: Q' s+ o- @) |: E: |3 y5 Y
8 \ V3 c9 ?( x& @Schmidt NO, Przylecki W, Yang W et al. Brain tumor tropism of transplanted human neural stem cells is induced by vascular endothelial growth factor. Neoplasia 2005;7:623¨C629.
. j$ r/ P) I1 F0 H" c0 V9 @
B4 G1 M$ g m' oRamiya VK, Maraist M, Arfors KE et al. Reversal of insulin-dependent diabetes using islets generated in vitro from pancreatic stem cells. Nat Med 2000;6:278¨C282.8 T6 O7 e& z% r( O/ u6 U
: s& e& W- @ N9 F" B
Huang H, Tang X. Phenotypic determination and characterization of nestin-positive precursors derived from human fetal pancreas. Lab Invest 2003;83:539¨C547.. A8 V5 {8 t/ R1 \" R
5 K4 t/ w& w& S) V2 lSuen PM, Li K, Chan JC et al. In vivo treatment with glucagon-like peptide 1 promotes the graft function of fetal islet-like cell clusters in transplanted mice. Int J Biochem Cell Biol 2006;38:951¨C960.' ]2 R/ N& c6 l. B/ j9 s n! E
4 [9 z+ r: T+ m+ ~
Suarez-Pinzon WL, Yan Y, Power R et al. Combination therapy with epidermal growth factor and gastrin increases beta-cell mass and reverses hyperglycemia in diabetic NOD mice. Diabetes 2005;54:2596¨C2601.
1 N+ ^& v0 ~) B8 S9 x- O0 p. g4 c5 l" a' O- x" w" X# b
Lechner A, Yang YG, Blacken RA et al. No evidence for significant transdifferentiation of bone marrow into pancreatic beta-cells in vivo. Diabetes 2004;53:616¨C623.8 O* Y2 f7 y7 E0 t9 T' |4 c4 \) x9 F5 R
% A& l O* ~% j7 [; T) POh SH, Muzzonigro TM, Bae SH et al. Adult bone marrow-derived cells trans-differentiating into insulin-producing cells for the treatment of type I diabetes. Lab Invest 2004;84:607¨C617.2 N3 f- s3 {) y* a% Z# q/ {
4 D, r9 l; m0 o% {5 P3 v
Yang L, Li S, Hatch H et al. In vitro trans-differentiation of adult hepatic stem cells into pancreatic endocrine hormone-producing cells. Proc Natl Acad Sci USA 2002;99:8078¨C8083.: h+ e7 ^8 N+ j% s5 E& Y/ x3 ?
& o8 A3 d5 J* l( j8 ?, Q
Torella D, Ellison GM, Mendez-Ferrer S et al. Resident human cardiac stem cells: Role in cardiac cellular homeostasis and potential for myocardial regeneration. Nat Clin Pract Cardiovasc Med 2006;3:S8¨CS13.: {8 y3 B2 T+ u; x* C/ q+ g1 e3 S
# G: j) o" a. j' T# b
Dawn B, Stein AB, Urbanek K et al. Cardiac stem cells delivered intravascularly traverse the vessel barrier, regenerate infarcted myocardium, and improve cardiac function. Proc Natl Acad Sci U S A 2005;102:3766¨C3771.' V% H" a9 l% K, D
0 w! s3 T8 y: f. r1 r' zKajstura J, Rota M, Whang B et al. Bone marrow cells differentiate in cardiac cell lineages after infarction independently of cell fusion. Circ Res 2005;96:127¨C137.
8 T; E6 { w7 y! B. x1 c3 @ P& h; B1 C+ X6 c; O
Mendez-Ferrer S, Ellison GM, Torella D et al. Resident progenitors and bone marrow stem cells in myocardial renewal and repair. Nat Clin Pract Cardiovasc Med 2006;30:S83¨CS89.: t8 v* N( x8 S7 g7 s
. d5 Q z; C4 C
Nakatomi H, Kuriu T, Okabe S et al. Regeneration of hippocampal pyramidal neurons after ischemic brain injury by recruitment of endogenous neural progenitors. Cell 2002;110:429¨C441.# N0 W$ E4 n7 `
5 m; R' N: M! Z7 O
Vendrame M, Cassady J, Newcomb J et al. Infusion of human umbilical cord blood cells in a rat model of stroke dose-dependently rescues behavioral deficits and reduces infarct volume. Stroke 2004;35:2390¨C2395.
0 {& n; g6 u/ x2 r8 Y$ ~5 V& {' A" J% G2 j5 v. R
Bambakidis NC, Miller RH. Transplantation of oligodendrocyte precursors and sonic hedgehog results in improved function and white matter sparing in the spinal cords of adult rats after contusion. Spine J 2004;4:16¨C26.1 z; j9 e, c* Z4 q- m9 [( [
$ W% o4 {% X2 E4 m2 E( JPicard-Riera N, Nait-Oumesmar B, Baron-Van EA. Endogenous adult neural stem cells: Limits and potential to repair the injured central nervous system. J Neurosci Res 2004;76:223¨C231.: ?# J' s- |3 J0 x; W# k, h( n( a
4 C% u1 T2 @4 [: X0 V. N+ TKawada H, Takizawa S, Takanashi T et al. Administration of hematopoietic cytokines in the subacute phase after cerebral infarction is effective for functional recovery facilitating proliferation of intrinsic neural stem/progenitor cells and transition of bone marrow-derived neuronal cells. Circulation 2006;113:701¨C710.* A: K) q/ L7 [& u: l2 j, T
+ w8 s+ x! | f2 \/ \
Lee ST, Chu K, Park JE et al. Intravenous administration of human neural stem cells induces functional recovery in Huntington's disease rat model. Neurosci Res 2005;52:243¨C249.. p( r) r% a+ z3 s! n7 ~$ ~
( D, e4 P6 }4 u/ x, O# M' }Correia AS, Anisimov SV, Li JY et al. Stem cell-based therapy for Parkinson's disease. Ann Med 2005;37:487¨C498.
) v$ M* ~* d9 z; e* C5 N3 `+ j( O% M& {6 i
Sugaya K. Possible use of autologous stem cell therapies for Alzheimer's disease. Curr Alzheimer Res 2005;2:367¨C376.! d" J! I2 U( a! y9 f2 D3 T& t
% O- l0 ^$ O3 v; w" V5 ZZhao Z, Hu H, Feng G. Learning and memory amelioration of transplantation of the neural stem cells modified with human brain-derived neurotrophic factor gene on Alzheimer disease model rat. Zhongguo Xiu Fu Chong Jian Wai Ke Za Zhi 2005;19:331¨C334.
; o7 X a# g* ~0 b; J; O/ h6 l) A2 X
: [8 c$ e$ [: \& ]6 PCopray S, Balasubramaniyan V, Levenga J et al. Olig2 overexpression induces the in vitro differentiation of neural stem cells into mature oligodentrocytes. STEM CELLS 2006;24:1001¨C1010.
/ Q3 d0 y& Q9 A% w+ ]7 i! w5 a5 @3 l) B
Lepore AC, Fischer I. Lineage-restricted neural precursors survive, migrate, and differentiate following transplantation into the injured adult spinal cord. Exp Neurol 2005;194:230¨C242.' ?0 m; g6 b) X% U$ t* S9 y- `
1 @: Y3 v7 @% x& {
Dogru M, Tsubota K. Current concepts in ocular surface reconstruction. Semin Ophthalmol 2005;20:75¨C93.( Q8 h7 m: ^* c d: L
4 [' l+ y& \, GGomes JA, Romano A, Santos MS et al. Amniotic membrane use in ophthalmology. Curr Opin Ophthalmol 2005;16:233¨C240.
" ], }7 ?* ?9 X* C0 _: Q7 e5 R8 W/ D9 c
Nakamura T, Inatomi T, Sotozono C et al. Successful primary culture and autologous transplantation of corneal limbal epithelial cells from minimal biopsy for unilateral severe ocular surface disease. Acta Ophthalmol Scand 2004;82:468¨C471.; N( g% R. K! d- X
4 P5 u* V Y% t) ~# K8 K4 bSantos MS, Gomes JA, Hofling-Lima AL et al. Survival analysis of conjunctival limbal grafts and amniotic membrane transplantation in eyes with total limbal stem cell deficiency. Am J Ophthalmol 2005;140:223¨C230.7 i7 H( b. {3 f/ ~
' }) E! i, v5 l" ZMa Y, Xu Y, Xiao Z et al. Reconstruction of chemically burned rat corneal surface by bone marrow-derived human mesenchymal stem cells. STEM CELLS 2006;24:315¨C321.
8 ~ h" n; D$ b$ I$ M' @# J8 V0 D( X$ x) P1 r) x: _2 V& P% s
Haynes T, Del Rio-Tsonis K. Retina repair, stem cells and beyond. Curr Neurovasc Res 2004;1:231¨C239.* L! I n; h; M+ Z# X; P
- e' F0 c$ V( i1 e# mWoodard P, Cunningham JM, Benaim E et al. Effective donor lymphohematopoietic reconstitution after haploidentical CD34 -selected hematopoietic stem cell transplantation in children with refractory severe aplastic anemia. Bone Marrow Transplant 2004;33:411¨C418.
) @ S0 U% Q8 N
7 w7 ~3 h( N: n$ y9 Q& tKushida T, Inaba M, Hisha H et al. Intra-bone marrow injection of allogeneic bone marrow cells: A powerful new strategy for treatment of intractable autoimmune diseases in MRL/lpr mice. Blood 2001;97:3292¨C3299.
9 ?+ P0 T6 n. g$ @& F/ w, f2 }) k4 E$ H' P; C
Petit I, Szyper-Kravitz M, Nagler A et al. G-CSF induces stem cell mobilization by decreasing bone marrow SDF-1 and upregulating CXCR4. Nat Immunol 2002;3:687¨C694.: L, n, I9 v }. w$ [' M
7 m& o9 i! q! ]) d# ]" W8 M# qNoia G, Pierelli L, Bonanno G et al. The intracoelomic route: A new approach for in utero human cord blood stem cell transplantation. Fetal Diagn Ther 2004;19:13¨C22.
# Y' i0 ?% Y" K) G& @2 `6 i. l$ `& b: ~/ `3 x9 n7 y) ^. C
Galli R, Binda E, Orfanelli U et al. Isolation and characterization of tumorigenic, stem-like neural precursors from human glioblastoma. Cancer Res 2004;64:7011¨C7021.
+ ]9 C5 t; ^+ o c5 z$ D$ D$ O% q1 M, b: M6 Z
Collins AT, Berry PA, Hyde C et al. Prospective identification of tumorigenic prostate cancer stem cells. Cancer Res 2005;65:10946¨C10951.
! s9 k6 k" y: f' I/ F
2 m4 Z. X, m& [5 j) e- ^# PKubo M, Nakamura M, Tasaki A et al. Hedgehog signaling pathway is a new therapeutic target for patients with breast cancer. Cancer Res 2004;64:6071¨C6074.& A, r5 y# p8 U4 L( Q
4 I m, v( G2 f, P2 V$ a# M
Romer J, Curran T. Targeting medulloblastoma: Small-molecule inhibitors of the Sonic Hedgehog pathway as potential cancer therapeutics. Cancer Res 2005;65:4975¨C4978.$ k; ]+ L- h' Z/ e
; O6 C+ K: U6 V {* ABudiyanto A, Bito T, Kunisada M et al. Inhibition of the epidermal growth factor receptor suppresses telomerase activity in HSC-1 human cutaneous squamous cell carcinoma cells. J Invest Dermatol 2003;121:1088¨C1094.
8 l z3 x* n" T8 @, t. W8 _
( s/ M, z: I) F3 e& J6 J" NMimeault M, Pommery N, Henichart JP. New advances on prostate carcinogenesis and therapies: Involvement of EGF-EGFR transduction system. Growth Factors 2003;21:1¨C14.
0 n+ |6 b% H/ H- E. D6 T9 F4 i c3 K3 ~8 |( t, I2 H; k
Mimeault M, Moore E, Moniaux N et al. Cytotoxic effects induced by a combination of cyclopamine and gefitinib, the selective hedgehog and epidermal growth factor receptor signaling inhibitors, in prostate cancer cells. Int J Cancer 2006;118:1022¨C1031., I0 M# L: k0 ]: g
' P* P! E9 y. ?7 m' d
Haruki N, Kawaguchi KS, Eichenberger S et al. Dominant-negative Notch3 receptor inhibits mitogen-activated protein kinase pathway and the growth of human lung cancers. Cancer Res 2005;65:3555¨C3561.$ X4 y2 C' V# }" @
1 `' l1 H9 M" u# ] N$ kChung KY, Saltz LB. Antibody-based therapies for colorectal cancer. Oncologist 2005;10:701¨C709.
( Q3 P& D8 X; v9 B& X" C9 j9 I4 \% s4 A$ t$ x( x: z: D' j& s
del Carmen MG, Rizvi I, Chang Y et al. Synergism of epidermal growth factor receptor-targeted immunotherapy with photodynamic treatment of ovarian cancer in vivo. J Natl Cancer Inst 2005;97:1516¨C1524.
& j1 V: U' f; _! J; g& c
3 D. R) l1 x; H4 E4 O8 b G. tMimeault M, Pommery N, Henichart JP. Synergistic antiproliferative and apoptotic effects induced by epidermal growth factor receptor and protein kinase a inhibitors in human prostatic cancer cell lines. Int J Cancer 2003;106:116¨C124.
% t% k2 y. e! u/ U7 R( [3 d0 K( L8 ~9 ?7 F- B/ F
Mimeault M, Jouy N, Depreux P et al. Synergistic antiproliferative and apoptotic effects induced by mixed epidermal growth factor receptor inhibitor ZD1839 and nitric oxide donor in human prostatic cancer cell lines. Prostate 2005;62:187¨C199.
6 F5 h' \6 i" `: ?9 a6 n5 [: q' @
5 q1 L* A% s( A+ F1 T7 B+ ?( bLepourcelet M, Chen YN, France DS et al. Small-molecule antagonists of the oncogenic Tcf/beta-catenin protein complex. Cancer Cell 2004;5:91¨C102.
2 S; s) i8 j' ~/ `" A+ ?" u' t
Luu HH, Zhang R, Haydon RC et al. Wnt/beta-catenin signaling pathway as a novel cancer drug target. Curr Cancer Drug Targets 2004;4:653¨C671.
R' a8 G" u+ i- X5 j# V( Y x4 T
Aster JC. Deregulated NOTCH Signaling in Acute T-Cell Lymphoblastic Leukemia/Lymphoma: New Insights, Questions, and Opportunities. Int J Hematol 2005;82:295¨C301.
* [% H8 o* _- |, K. y) j- V" l2 \; W3 X% q3 v, t
Paris D, Quadros A, Patel N et al. Inhibition of angiogenesis and tumor growth by beta and gamma-secretase inhibitors. Eur J Pharmacol 2005;514:1¨C15.8 E3 Z1 P& Q& |, H2 U3 |% O
7 q$ t; J" {1 [4 k1 O: u7 Z
Hopfer O, Zwahlen D, Fey MF et al. The Notch pathway in ovarian carcinomas and adenomas. Br J Cancer 2005;93:709¨C718.
% I) h' I" R$ @8 f3 |0 \
& a8 y$ Q$ @+ @: @9 R2 c1 x3 WSjölund J, Manetopoulos C, Stockhausen MT et al. The Notch pathway in cancer: Differentiation gone awry. Eur J Cancer 2005;41:2620¨C2629./ p( I: U5 s. }# @
0 C' @/ O2 S o. ~% j) C8 W |' N
Yokoi K, Sasaki T, Bucana CD et al. Simultaneous inhibition of EGFR, VEGFR, and platelet-derived growth factor receptor signaling combined with gemcitabine produces therapy of human pancreatic carcinoma and prolongs survival in an orthotopic nude mouse model. Cancer Res 2005;65:10371¨C10380.& G! T* l3 c2 o# Q
* I2 L; G& |9 t. J- j' _Britton DJ, Hutcheson IR, Knowlden JM et al. Bidirectional cross talk between ERalpha and EGFR signalling pathways regulates tamoxifen-resistant growth. Breast Cancer Res Treat 2006;96:131¨C146.
, k& }5 E. ^5 P# ?2 Q
& e4 Y/ v- R7 G# ^& L5 ESun YX, Schneider A, Jung Y et al. Skeletal localization and neutralization of the SDF-1(CXCL12)/ CXCR4 axis blocks prostate cancer metastasis and growth in osseous sites in vivo. J Bone Miner Res 2005;20:318¨C329.
2 Y5 u0 D' z0 G, |
: w8 }6 L# `/ b& d6 n. i: GMann JR, Backlund MG, DuBois RN. Mechanisms of disease: Inflammatory mediators and cancer prevention. Nat Clin Pract Oncol 2005;2:202¨C210.
0 p. g" B" E+ o9 o$ ^; m6 L& }: V9 {" m/ J" \+ V
Sartor CI. Mechanisms of disease: Radiosensitization by epidermal growth factor receptor inhibitors. Nat Clin Pract Oncol 2004;1:80¨C87.
% b, ?4 ^+ ~" M+ p0 c, J* _+ h/ e6 x: f& n5 G8 q- y
Morelli MP, Cascone T, Troiani T et al. Sequence-dependent antiproliferative effects of cytotoxic drugs and epidermal growth factor receptor inhibitors. Ann Oncol 2005;16 (suppl 4):iv61¨Civ68./ o1 Z2 M( x( `; d- A
0 p% Q% T4 ]- T+ W |% r) V7 J
Mimeault M. New advances on structural and biological functions of ceramide in apoptotic/necrotic cell death and cancer. FEBS Lett 2002;530:9¨C16.# Y+ s6 n. L! [$ y# ~
+ Y7 u$ `: E( Y' T6 kWang AL, He ZD, Luo Y et al. Treatment of moderate-grade and high-grade malignant lymphoma with autologous hematopoietic stem cell transplantation. Ai Zheng 2003;22:1317¨C1320.1 ?& M$ }9 v) R5 U* T
8 {( F- N3 U- R5 [
Aoudjhane M, Labopin M, Gorin NC et al. Comparative outcome of reduced intensity and myeloablative conditioning regimen in HLA identical sibling allogeneic haematopoietic stem cell transplantation for patients older than 50 years of age with acute myeloblastic leukaemia: A retrospective survey from the Acute Leukemia Working Party (ALWP) of the European group for Blood and Marrow Transplantation (EBMT). Leukemia 2005;19:2304¨C2312.
1 I. k9 ~1 E8 ?6 ]+ @$ N; o$ L* d2 t' y7 |+ d
Cui JW, Zhang XM, Wang GJ. Progress in the studies of acute myelogenous leukemia stem cell. Zhongguo Shi Yan Xue Ye Xue Za Zhi 2003;11:549¨C552.
* M2 D0 j3 [9 b2 p6 t4 H" g2 Z4 k1 |" A
Matsubara H, Makimoto A, Higa T et al. A multidisciplinary treatment strategy that includes high-dose chemotherapy for metastatic retinoblastoma without CNS involvement. Bone Marrow Transplant 2005;35:763¨C766.
& Y Q: y6 |- ^7 D* v2 a& o1 N1 {2 L5 V! K% t! O& _
Iwasaki Y, Nagata K, Nakanishi M et al. Double-cycle, high-dose ifosfamide, carboplatin, and etoposide followed by peripheral blood stem-cell transplantation for small cell lung cancer. Chest 2005;128:2268¨C2273.1 ^( M7 u" p# i& U! ^# R3 M& I4 ?
. v @* A/ P% c, E" ~2 D C% Q, iVogel W, Kopp HG, Kanz L et al. Myeloma cell contamination of peripheral blood stem-cell grafts can predict the outcome in multiple myeloma patients after high-dose chemotherapy and autologous stem-cell transplantation. J Cancer Res Clin Oncol 2005;131:214¨C218.
4 N& N" z, a# H* z2 T; Z, z' n9 {. L, V, ]+ M
den Brok MH, Nierkens S, Figdor CG et al. Dendritic cells: Tools and targets for antitumor vaccination. Expert Rev Vaccines 2005;4:699¨C710. |
|