干细胞之家 - 中国干细胞行业门户第一站

 

 

搜索
朗日生物

免疫细胞治疗专区

欢迎关注干细胞微信公众号

  
查看: 527059|回复: 261
go

The Phenotypes of Pluripotent Human Hepatic Progenitors [复制链接]

Rank: 7Rank: 7Rank: 7

积分
威望
0  
包包
483  
楼主
发表于 2009-3-5 00:03 |只看该作者 |倒序浏览 |打印
作者:Eva Schmelzer, Eliane Wauthier, Lola M. Reid作者单位:Department of Cell and Molecular Physiology, University of North Carolina School of Medicine, Chapel Hill, North Carolina, USA 1 ?0 Y& C3 c# Z& X" D1 m( e
                  
+ U9 \% }, m% w* G- h1 Z  c                  ' h% H8 M- G9 V( \
         
/ L9 o/ U  W  \' i! J& ^, i# C                        
' _. ^9 b  E; B            $ H! F9 V# i2 q* a
            : c3 M. \! L2 v3 w9 f
            " z+ b& h- ^6 P% `3 g) d. R
            1 f' _( d: Q9 }2 o8 L
                      6 s' R) Z6 R2 S3 C& ~. w/ _
        8 `+ F6 L# I0 n% K0 @" I6 r
        
7 r; B/ h& f$ I+ z        
4 r2 _+ T+ G1 C6 r6 Z# H# O          【摘要】
/ F' ?" w$ [' d      Human livers contain two pluripotent hepatic progenitors, hepatic stem cells and hepatoblasts, with size, morphology, and gene expression profiles distinct from that of mature hepatocytes. Hepatic stem cells, the precursors to hepatoblasts, persist in stable numbers throughout life, and those isolated from the livers of all age donors from fetal to adult are essentially identical in their gene and protein expression profiles. The gene expression profile of hepatic stem cells throughout life consists of high levels of expression of cytokeratin 19 (CK19), neuronal cell adhesion molecule (NCAM), epithelial cell adhesion molecule (EpCAM), and claudin-3 (CLDN-3); low levels of albumin; and a complete absence of expression of -fetoprotein (AFP) and adult liver-specific proteins. By contrast, hepatoblasts, the dominant cell population in fetal and neonatal livers, decline in numbers with age and are found as <0.1% of normal adult livers. They express high levels of AFP, elevated levels of albumin, low levels of expression of adult liver-specific proteins, low levels of CK19, and a loss of NCAM and CLDN-3. Mature hepatocytes lack expression altogether of EpCAM, NCAM, AFP, CLDN-3, cytokeratin 19, and have acquired the well-known adult-specific profile that includes expression of high levels of albumin, cytochrome P4503A4, connexins, phosphoenolpyruvate carboxykinase, and transferrin. Thus, hepatic stem cells have a unique stem cell phenotype, whereas hepatoblasts have low levels of expression of both stem cell genes and genes expressed in high levels in mature hepatocytes. 1 Q+ h: ]3 B! H' ]
          【关键词】 Liver Stem cell Hepatoblast Progenitor Epithelial cell adhesion molecule Neural cell adhesion molecule Hepatocyte Gene expression -Fetoprotein" C; I/ E0 x# i2 K4 |8 N
                  INTRODUCTION
) z& e/ T3 `. r7 j5 W9 ~- Z. ~
: B/ A$ a0 f/ t0 c+ d  U! jDuring development, the hepatic bud arises from the foregut endoderm .% ~5 @- y- K4 B8 E+ M, y; l1 [
& c% e4 e4 e% Y
The ratio of hepatic stem cells to hepatoblasts changes depending on developmental stage, with the hepatoblasts being dominant in fetal and neonatal livers and with few if any mature parenchymal cells .6 W( l7 g% }* d9 s" \3 R
7 `/ ^. O/ o  F; l4 E4 r" s
Although the identity of hepatic stem cells and hepatoblasts in human livers has been described recently, our knowledge is incomplete on the gene and protein expression profiles of these pluripotent progenitors and whether the profiles change during development. We have shown that epithelial cell adhesion molecule (EpCAM), a 34¨C40-kDa transmembrane glycoprotein  (Schmelzer et al., manuscript submitted for publication; Bruce et al., manuscript submitted for publication). Thus, sorting for EpCAM results in only progenitor cells but in distinct ratios of hepatic stem cells to hepatoblasts, depending upon whether the tissue is fetal, neonatal, or adult. In these studies, we analyzed the gene expression profiles in hepatic stem cells and hepatoblasts isolated from fetal, neonatal, pediatric, and adult livers and compared these profiles to those of mature hepatocytes.
, \9 N9 N" k% [+ a* r3 }  t  |/ W) T! r1 P3 L& K
MATERIALS AND METHODS
$ [! \, e. G$ ~" L- d! A5 ~( r
( Q& D' w7 @, ^  W  BNeonatal, Pediatric, and Adult Liver Cell Isolation
& V, F! ]. ^3 ^0 X3 r4 @
/ H) Q# \4 v7 e5 O9 m% ^+ N- RDonated livers not suitable for orthotopic liver transplantation were obtained from federally designated organ procurement organizations. Informed consent was obtained from next of kin for use of the livers for research purposes. To isolate cells, adult and pediatric livers were perfused through the portal vein and hepatic artery with EGTA-containing buffer for 15 minutes and 125 mg/l Liberase (Roche Applied Science, Indianapolis, http://www.roche-applied-science.com) for 30 minutes at 34¡ãC. Cells were passed sequentially through filters of pore size 1,000, 500, 250, and 150 µm and centrifuged in OptiPrep density gradients at 500g to select for viable cells.
$ A, O' w  [8 b0 Q* b1 e9 h& Y2 A
2 I' ?; S% R0 e9 h) OFetal Liver Cell Isolation
5 h. h8 s2 G- W) j& u
$ @6 F* ]2 S' d7 CFetal livers were obtained from Advanced Biological Resources (Alameda, CA) and were of developmental stages between 16 and 20 weeks of gestation. Nonhepatic tissue was removed, and livers were minced by scalpel and digested at 37¡ãC by 0.06% collagenase (Sigma-Aldrich, St. Louis, http://www.sigmaaldrich.com) in RPMI 1640 (Gibco, Grand Island, NY, http://www.invitrogen.com) supplemented with 0.03% deoxyribonuclease (w/v), 0.1% (w/v) bovine serum albumin fatty acid-free, 30 nM selenium (Sigma-Aldrich), and antibiotic-antimycotic mixture (Gibco). Hematopoietic cells and nonparenchymal cells were separated from the parenchymal cell fraction by slow-speed centrifugations (30g for 5 minutes in 40 ml) until the pellet showed minimal contamination with red blood cells. Cells in suspension were filtered through a 70-µm nylon mesh, and flow through was applied on a Ficoll-Paque (Amersham Biosciences-GE Healthcare, Piscataway, NJ, http://www.amersham.biosciences.com) gradient (cells in a 5-ml suspension applied on 5 ml of 84% Ficoll, placed on 5 ml of 100% Ficoll) and centrifuged for 30 minutes at 4¡ãC at 980g. Cells forming an interphase were collected and washed once in RPMI 1640 supplemented with 0.1% bovine serum albumin fatty acid-free (w/v), 30 nM selenium, 0.054% niacinamide (w/v), insulin (0.135 USP units/ml), bovine apo-transferrin (10 µg/ml), free fatty acid mixture (2.36 µM palmitic acid, 0.21 µM palmitoleic acid, 0.88 µM stearic acid, 1.02 µM oleic acid, 2.71 µM linoleic acid, 0.43 µM linolenic acid), 1 µM M hydrocortizone, 50 µM ß-mercaptoethanol (Sigma-Aldrich), 2 mM L-glutamine, and antibiotic-antimycotic mixture (Gibco) and kept on ice for further handling.0 z# Q+ m. w& o! p. M9 r
* K* u. n: u; e2 D
Cell Sorting
# e2 X6 o, ?2 o5 T/ W' P- I! b1 X) d0 c% C$ K
Cell sorting for EpCAM  cells was done using magnetic-activated cell sorting (MACS) according to manufacturer's instructions (Miltenyi Biotec, Auburn, CA, http://www.miltenyibiotec.com). Cells were incubated in the dark at 4¡ãC with fluorescein isothiocyanate (FITC)-conjugated antibody (Miltenyi Biotec) for 20 minutes at a concentration of 50 µl for 50 x 106 total cells in 500 µl of buffer (phosphate-buffered saline ) containing 0.5% bovine serum albumin and 2 mM EDTA (Sigma-Aldrich). EpCAM  cells were labeled with magnetic beads using an anti-FITC Multisort Kit (Miltenyi Biotec) and selected by Midi- or MiniMACS columns and separation units (Miltenyi Biotec). All incubation and selection steps were performed on ice or at 4¡ãC with addition of 10% accutase (Innovative Cell Technologies, San Diego, CA) to prevent aggregation of cells.
, b+ c  D- u* H2 X; P
9 X8 A! `1 c+ S6 g7 FCell Culture5 p  F$ e4 k; _: [5 }% \* ]+ q
( N! z# _+ R( l' a5 m
Cells obtained by Ficoll gradient selection were placed in culture on plastic or collagen III (6.25 µg/cm2) (Sigma-Aldrich)-coated plastic vessels. Cultivation was done in supplemented RPMI 1640 medium (see cell isolation) with an initial 24-hour phase of 10% fetal bovine serum addition. After 24 hours, cells were cultured serum-free, and medium was changed every 4 days. Colonies that formed were picked at 10 and 20 days and collected on ice for further analyses.
: C- c$ `: Z/ o2 [0 c* f! _  `5 W) I" z* h5 v) k
Cytospins8 \0 h, `, N- z' W

3 R; _- F- B2 q0 L) U) GFollowing enzymatic digestion of fetal livers, freshly isolated cells were centrifuged at 600g for 5 minutes at 4¡ãC. Cells were then resuspended in PBS and 4% paraformaldehyde (1:1). Following fixation for 10 minutes at room temperature, cells were centrifuged as previously described. The supernatant was aspirated, and the cells were then blocked in PBS containing 2% Triton X-100 (Sigma-Aldrich), 10% goat serum (Gibco), and 2% cold water fish gelatin (Sigma-Aldrich). Cells were incubated for 1 hour at room temperature and then centrifuged as described above. Primary and secondary antibodies were incubated for 60 minutes at room temperature. Cells were washed twice in PBS plus 2% Triton, centrifuged, resuspended in blocking buffer, and cytospun at 800 rpm for 3 minutes using precoated cytoslides (Shandon Anatomical Pathology, Pittsburgh, PA). Cells were mounted in aqueous glycerol gelatin mounting medium (Sigma-Aldrich) and coverslipped. Images were visualized using a Leica SP2 AOBS laser scanning confocal microscope that was controlled by Leica SP2 TCS software (Leica, Bannockburn, IL, http://www.leica.com). Negative controls were performed by omitting primary antibody from the protocol and revealed no staining (data not shown).
9 ]* }( u; n$ ?& k, Q" @9 S* n7 v2 _' d
SDS-Polyacrylamide Gel Electrophoresis and Western Blot Analyses
( V1 S. y+ o* r+ \
+ m! ^; B6 o' U; sTotal proteins were extracted from sorted cells or from cultured cells by washing cells once with ice-cold PBS and resuspending cell pellets in ice-cold protein lysis buffer (1% Triton X-100, 10 mM TrisHCl, 150 mM NaCl, 1% protease inhibitor cocktail ). Cell suspensions were spun down at 20,000g for 5 minutes at 4¡ãC, and supernatants were frozen in aliquots for further analysis at ¨C20¡ãC.
4 l! n$ R+ {; _4 Q6 p7 I; l/ E3 q& i; B- r; _. }* s
Denaturing SDS-polyacrylamide gel electrophoresis and Western blots were done as described previously  using a Mini-Protean-3-Cell and Mini-Trans-Blot Transfer Cell (Bio-Rad, Hercules, CA, http://www.bio-rad.com). Nitrocellulose membranes were blocked in Odyssey (LI-COR, Lincoln, NE, http://www.licor.com) blocking buffer/PBS (1:1) for 1 hour, and incubated with primary and appropriate secondary antibody in blocking buffer/PBS   0.1% Tween-20 (Sigma-Aldrich). Membranes were washed four times for 5 minutes with PBS   0.1% Tween-20 after each incubation with antibody. Dried membranes were scanned using an Odyssey infrared scanner (LI-COR). Quantitative protein expression was done by loading protein standards of known concentration simultaneously on the gel, creating a standard curve that was applied to calculate unknown protein concentrations in samples using the Odyssey version 1.2 software (LI-COR).
! k# h9 u" r4 |6 A7 }) l# P' w3 C9 m% X' ~
Standards for quantitative protein analyzes were as follows: albumin (Research Diagnostics, Flanders, NJ, http://www.researchd.com), -fetoprotein (AFP) (Fitzgerald Industries International, Concord, MA, http://www.fitzgerald-fii.com), cytokeratin 19 (CK19) (Research Diagnostics), and transferrin (Fitzgerald Industries International)./ z6 ?$ g( L( e) J
* D' A7 x* R2 S
Primary antibodies were albumin, AFP (Sigma-Aldrich), CK19 (Cymbus Biotechnology, Hampshire, U.K.), transferrin (Rockland, Gilbertsville, PA), and CLDN-3 (Abcam, Cambridge, MA, http://www.abcam.com). Secondary antibodies were Alexa Fluor 680 (Western blots) and AF568 (cytospins) polyclonal goat anti-rabbit IgG (H L) and Alexa Fluor 680 (Western blots) and AF488 (Cytospins) polyclonal goat anti-mouse IgG (H L) (Molecular Probes Inc., Eugene, OR, http://probes.invitrogen.com).
. |' D$ h# y+ p
' e( n3 B7 D; J3 t8 dQuantitative Real-Time Reverse Transcription-Polymerase Chain Reaction' U3 f/ L9 h) V( w9 N- i8 k1 G

% K: f5 q9 A% L! l7 eGene expression was analyzed by quantitative real-time reverse transcription-polymerase chain reaction (RT-PCR). Gene-specific mRNAs were created as followed: total RNA from livers was extracted using the RNeasy kit (Qiagen, Valencia, CA, http://www1.qiagen.com) and reverse-transcribed by Superscript II reverse transcriptase (Invitrogen, Carlsbad, CA, http://www.invitrogen.com) and oligo-(dT)(12¨C18) primer (Gene Link, Hawthorne, NY). cDNA was used as template in conventional PCR with gene-specific primers (for sequences, see Table 1) from which the forward primer possessed a 5' overhang for T7-promotor sequence (5'-gac tcg taa tac gac tca cta tag gg). This amplified gene-specific DNA was used for in vitro transcription with T7-RNA polymerase (Promega, Madison, WI, http://www.promega.com), generating gene-specific RNA(with an additional 5'-ggg included by T7-RNA polymerase) used as standards in quantitative real-time RT-PCR using gene-specific primers without the 5' overhang; standard ranges were linear from 1 to 108 templates. Primers were selected on different exons wherever possible. Quantitative real-time RT-PCR was done in the LightCycler instrument using the LightCycler RNA Master SYBR Green I kit (Roche). RNA from samples was extracted using RNeasy mini kit (Qiagen).( E2 K5 E2 ]% M1 ]( T1 Y
  R( V( [  J; |9 F0 J$ M- ]+ t
Table 1. Primer sequences
7 F6 r0 g& q* G1 A1 ?0 y9 B! T& [6 n8 Q8 D
RESULTS  S" f" m( K7 u3 O# F( o# H% t+ g
, C( }* Q8 e7 d! }
Developmental Changes in the Percentages of EpCAM  Cells and of the Ratios of Hepatic Stem Cells Versus Hepatoblasts Within EpCAM  Cells
6 k: w2 S+ C& |* h: [5 p& ~: ^# x) {1 R5 Y7 @2 B: h* a
Data obtained in this study (Table 2) confirmed our prior finding (Bruce et al., manuscript submitted for publication) that cell suspensions prepared from neonatal and adult livers consisted of low percentages of EpCAM  cells (1.7%¨C2.5%), of which less than 0.1% are hepatoblasts. By contrast, EpCAM -sorted cells from fetal livers accounted for 12% of liver cell suspensions, of which more than 80% are hepatoblasts and 0.1%¨C0.7% are hepatic stem cells. Thus, the percentage of hepatic stem cells remains at relatively constant ratio of the total liver cells, whereas hepatoblasts exist as the dominant pluripotent cell in fetal and neonatal livers, but their numbers decline with age, occurring in high numbers in adult livers only in disease states, such as cirrhosis. In fetal livers, EpCAM-negative cells comprise nonparenchymal cells, including mainly hematopoietic and mesenchymal cells. By contrast, in postnatal livers, EpCAM-negative cells proved to be more than 95% diploid and polyploid hepatocytes (A. Bruce, L. Zhang, J.W. Ludlow, E. Schmelzer, A. Melhem, M. Kulik, L.M. Reid, and M.E. Furth, manuscript submitted for publication).
' {" u# C2 ]: a) L4 x, q$ t( C9 a" i. T/ t6 m
Table 2. Relative proportions of hepatic EpCAM  cells in donors of different ages* P. m0 b1 N7 X, @# c4 P) n  f
& P% f$ n# T, J
Expression Profiles of Hepatic Stem Cells from Livers of Donors of Different Ages
" y2 P  B) H7 r+ H+ e7 Q  t5 ^( n4 q. A  \0 [: Z
Culture-selected hepatic stem cells from fetal livers were analyzed for quantitative gene expression and used as controls to that of freshly isolated and sorted EpCAM  liver cells from livers of varying age donors (Tables 3 and 4). In general, culture-selected stem cells, whether isolated on type III collagen or culture plastic and whether from short-term (10 days) or long-term (20 days) cultures, gave similar results. Hepatic stem cells isolated from the livers of all developmental stages assayed expressed very similar gene expression profiles, with high expression of CK19, EpCAM, neuronal cell adhesion molecule (NCAM), CLDN-3, c-kit, and aquaporin 4 but low levels of expression of albumin and very low or no expression of AFP or of any liver-specific genes (e.g., connexins, PEPCK, DPP4, cytochrome P450 3A4, and transferrin). Protein profiles of hepatic stem cells from all developmental stages (Table 5) matched the mRNA expression, showing protein expression for CK19, NCAM, albumin, and CLDN-3 but not AFP. Confirmation of the unique expression of CLDN-3 in hepatic stem cells but not in hepatoblasts or mature hepatocytes could be shown also by Western blot analyses (Fig. 1) and immunocytochemistry (Fig. 2).' t. X4 v& N! Z6 V! Q) m4 b6 L. {
- r+ P: m8 e" D5 s+ S; l) B
Table 3. Comparison of quantitative gene expression from human hepatic stem cells and hepatoblasts; p, J- v% ~$ [

& q4 N' N3 }8 h$ k; e9 x+ [Table 4. Comparison of quantitative gene expression from human hepatic stem cells vs. hepatocytes5 z% X# {  y2 Z8 y

9 G$ Y5 w' t3 M( j5 fTable 5. Comparison of quantitative protein expression from human hepatic stem cells, hepatoblasts, and hepatocytes
! }3 i  G; p) t: _2 c3 m6 S7 A9 Q4 F
# k9 `. I% \# }" P! r8 O: Q! K* JFigure 1. Claudin-3 expression analysis by Western blot. (A): Adult and neonate EpCAM - and EpCAM¨C-sorted cells from two different patients each. (B): Fetal EpCAM  and EpCAM¨C-sorted cells from three different patients. (C): Fetal stem cell colonies after 10 and 20 days in culture on plastic (C). One microgram of total protein per lane. Left lanes, molecular weight markers. Abbreviations: d, days; EpCAM, epithelial cell adhesion molecule.$ R7 x) S5 l. Q1 c

# D) l4 J( ?- Z8 i" kFigure 2. Claudin-3 expression analysis by immunocytochemistry of freshly isolated, unsorted fetal liver cells on cytospin. (A¨CC): The same cells, stained for 4,6-diamidino-2-phenylindole (blue), CLDN-3 (red), and either AFP, CK19, or ALB (green). Merges show co-expression of CLDN-3 with CK19 (B) and ALB (C) but not AFP (A). Magnification x189; scale bar = 20 µm. Abbreviations: AFP, -fetoprotein; ALB, albumin; CK19, cytokeratin 19.  G8 S; e7 t2 h! ?5 x
% D0 i- f5 V. F8 O; j
Gene Expression Profiles of Hepatoblasts0 }4 A6 O4 g0 H" r  K3 c( J
- z/ z/ a- z8 U4 K- |9 v
Gene expression patterns of hepatoblasts (Table 3) are different from those of hepatic stem cells and can be characterized generally as loss of expression (CLDN-3 and NCAM) and low levels of expression (CK19 and c-kit) of the genes characteristic of the stem cell phenotype, activation of expression, albeit at low levels, of adult liver-specific genes (e.g., connexins, PEPCK, and P450s) and intense expression of AFP, a defining feature of hepatoblasts. Albumin gene expression in hepatoblasts is about 1,000-fold higher than in hepatic stem cells and still 50-fold higher in its protein expression (Table 4), which correspondents to that of hepatocytes. Some genes defining mature liver functions (e.g., cytochrome P450 3A4 and -1-antitrypsin) were not expressed by either hepatic stem cells or hepatoblasts. Although variations between different donors in gene expression profiles could be observed (variations caused not only by individual patient history but influenced by shipping and handling of the donor organ) we were able to manifest distinct trends in gene expression patterns, clearly showing different profiles for hepatic stem cells and hepatoblasts.
  }; b# z5 n# `  X9 m% @1 c. L/ p2 \5 q  ~. V. C3 N+ k
Expression Profiles of Hepatocytes from Livers of Donors of Different Ages
' ~! O2 X, y& U; I  r  i- }* C' w3 |& P2 M- l: D2 t# {
Gene expression in hepatocytes was compared with that in hepatic stem cells and is summarized in Table 4 by calculating ratios of the data from EpCAM  versus EpCAM¨C-sorted cells. Gene expression data from hepatocytes from postnatal livers proved quite variable due to variation caused by organ handling/shipping (necrosis caused by time delay until processing) and donor history (drug administration, nicotine use, obesity, etc.). Nevertheless, a pattern that proved consistent was the absence of expression of the genes defining the hepatic stem cells (e.g., NCAM, CK19, c-kit, and CLDN-3) or hepatoblasts (e.g., AFP) and high levels of expression of classic liver-specific genes (cytochrome P4503A4 and transferrin).
7 U1 I# E$ ^. B2 ]
, K4 j' Z$ Q% \; _; X. l' C3 lDISCUSSION
  R6 D, w  p9 J7 S; Y- z& B) J+ t; E* B  {! M) q& [! P# i
The gene expression profiles in terms of both RNAs and proteins of two pluripotent human hepatic progenitors, hepatic stem cells and hepatoblasts, have been compared with those of mature human hepatocytes and shown to reveal three distinct patterns: a phenotype for stem cells, a phenotype for mature hepatocytes, and one for hepatoblasts that is a mix of the other two.
! g2 q4 ^+ u" n' |5 b
. H4 W- M& g7 z4 F# \" E5 {/ qThe ability to isolate and purify specific subpopulations of human liver cells such as the human hepatic stem cells has been made possible only recently by using immunoselection for a defined antigen such as EpCAM, a transmembrane glycoprotein described by de Boer .
# F. E% ]2 s) L' m: U! \( m( V  @! a, F: z3 }
By immunohistochemical analyses, we have been able to assign EpCAM expression in nonpathologic human livers to hepatic stem cells and hepatoblasts in all developmental stages (i.e., fetal, neonatal, pediatric, and adult livers). Transplantation into the livers of mice of EpCAM -sorted cells gave rise to human liver tissue expressing human-specific liver proteins (E. Schmelzer, L. Zhang, A. Melhem, N.G. Moss, E. Wauthier, R.E. McClelland, A. Bruce, H. Yao, W.S. Turner, N. Cheng, M.E. Furth, and L.M. Reid, manuscript submitted for publication; A. Bruce, L. Zhang, J.W. Ludlow, E. Schmelzer, A. Melhem, M. Kulik, L.M. Reid, and M.E. Furth, manuscript submitted for publication). Sorting for EpCAM  cells from pediatric and adult livers resulted in a population mainly comprised of hepatic stem cells, whereas those from fetal and neonatal livers resulted in predominantly hepatoblasts.
  w. x4 L, n5 k
- w% v1 ], o: HWe show here that hepatic stem cells from livers at all developmental stages show highly similar expression profiles, defined below as the stem cell phenotype. Phenotypic characterization of gene expression at mRNA and protein levels in pluripotent human hepatic progenitors versus mature hepatocytes indicated two patterns, a stem cell phenotype and a liver-specific phenotype; a gradient from minimal levels was observed in hepatoblasts to maximal levels found in hepatocytes. The stem cell phenotype comprised expression of EpCAM, NCAM, CK19, c-kit, CLDN-3, and weak levels of albumin but no expression of AFP or adult liver-specific proteins such as transferrin, connexins, PEPCK, DPP4, or P450s. The expression of both hepatic (albumin) and biliary (CK19) markers reflects the bipotential nature of the hepatic stem cell toward both lineages of the liver, the hepatic and biliary, which are characterized by albumin and CK19 expression, respectively. In studies published elsewhere, the hepatic stem cells also express components of the Hedgehog signaling pathway, including Indian and Sonic Hedgehog and its receptor, Patched .
% v/ I9 N" i& e* H/ j) E
7 f! C- k  u" b! h& X% }' L% CThe hepatocyte phenotype comprised expression of classic liver-specific genes such as albumin, connexins, transferrin, PEPCK, DPP4, and P450s and loss of expression of the genes typifying stem cells (including EpCAM, NCAM, CK19, c-kit, and CLDN-3). Hepatoblasts proved to be cells in an intermediate position with lowered levels of the stem cell genes and with activation but low levels of adult liver-specific genes and with the unique, defining feature of AFP.
! L2 w& ]. M$ J2 W+ {# C& T+ Z: [
1 v1 X% p6 X& `8 \7 Z. W6 uA unique and novel hepatic stem cell surface marker is CLDN-3. Together with occludins, claudins form tight junction proteins . Combining our results with these previous findings, CLDN-3 is a candidate surface molecule expressed by hepatic stem cells and providing a marker in addition to NCAM, permitting selection uniquely for stem cells.+ t* x+ N$ R+ j; N- m* {

2 E: i0 y! @' B! vIn summary, this study suggests that the pluripotent hepatic progenitors are evident in both fetal and postnatal livers and remain stable in their phenotypes throughout life, particularly the hepatic stem cells, which persist in relatively constant numbers in livers at all donor ages. The primary changes with age are in the numbers of hepatoblasts that are the dominant cell type in fetal and neonatal livers and then decline in numbers to comprise 4 k  H/ c1 k& K! _, a

, h  V) T: w) t8 CDISCLOSURES
. J( V  z/ H, B8 X$ A- C3 Y% Q' o8 ~, Z6 }, [% W9 C& o
The authors indicate no potential conflicts of interest.
, F7 c% y) c' L3 r2 `
: b" q; X* d. G7 ~) `5 MACKNOWLEDGMENTS
2 ~4 M, }8 i- ^( \7 \3 o+ X$ D
0 c8 D) _( H4 q  _: o# E6 fFunding for this work was derived primarily from a sponsored research grant from Vesta Therapeutics (formerly Incara Pharmaceuticals) (Research Triangle Park, NC); funding to L.M.R. was derived from NIH grants (DK52851, AA014243, IP30-DK065933) and a Department of Energy grant (DE-FG02-02ER-63477). We thank Lucendia English and University of North Carolina Michael Hooker Microscopy Core Facility under the direction of Michael Chua for technical support. We thank National Development and Research Institutes, Inc. for providing available liver tissue. This research was made possible by the willingness of organ donor families to donate livers unsuitable for transplantation to research.
: l9 d7 l% p) t! ]          【参考文献】
. k. B, o4 x  T8 l
4 |+ u# J9 I( y3 Q5 y0 ?9 }
: s2 Q: [! v9 s/ o* ^Houssaint E. Differentiation of the mouse hepatic primordium. I. An analysis of tissue interactions in hepatocyte differentiation Cell Differ 1980;9:269¨C279.
( q) g! o  W5 @# `) V- Q" _3 E' |8 [
Jung J, Zheng M, Goldfarb M et al. Initiation of mammalian liver development from endoderm by fibroblast growth factors. Science 1999;284:1998¨C2003.
- B8 Q4 s( C+ Z' ?. f4 a8 G4 b7 v) Q, S: U
Tremblay KD, Zaret KS. Distinct populations of endoderm cells converge to generate the embryonic liver bud and ventral foregut tissues. Dev Biol 2005;280:87¨C99.' w+ P. c+ j8 U" O
+ ]- U1 ?7 k3 }9 Z
Schmelzer E, Zhang L, McClelland R et al. Hepatic stem cells and the liver's maturational lineages: Implications for liver biology, gene expression and cell therapies. In: Potten C, ed. Tissue Stem Cells: Biology and Applications. London: Marcel Dekker, 2006; in press.& G0 I: k9 P- T4 T6 w; T
1 S! B" q8 X1 h! w/ s( B: C6 Q. d
Shiojiri N, Lemire JM, Fausto N. Cell lineages and oval cell progenitors in rat liver development. Cancer Res 1991;51:2611¨C2620.
: w" Q  b6 t! _9 Q. B& F9 t0 X1 z/ [
% Y1 r4 O% a5 O- A" y1 oTheise ND, Saxena R, Portmann BC et al. The canals of Hering and hepatic stem cells in humans. Hepatology 1999;30:1425¨C1433.
" Y) M* u6 V  U5 F9 e7 p" J: `5 e5 l
Paul J, Conkie D, Freshney RI. Erythropoietic cell population changes during the hepatic phase of erythropoiesis in the fetal mouse. Cell Tissue Kinet 1969;2:283¨C294.& ]6 s. K( P" o  o
$ e" Z( v+ @0 S( K8 N. n
Balzar M, Winter MJ, de Boer CJ et al. The biology of the 17-1A antigen (Ep-CAM). J Mol Med 1999;77:699¨C712.
: W# N3 L% z+ Q
5 ]. t( M/ k' _) K: X4 {Spurr NK, Durbin H, Sheer D et al. Characterization and chromosomal assignment of a human cell surface antigen defined by the monoclonal antibody AUAI. Int J Cancer 1986;38:631¨C636.
  Q6 p8 j0 g. F5 \, Q: G
2 w  c* b) r2 Z# }! J) RStrnad J, Hamilton AE, Beavers LS et al. Molecular cloning and characterization of a human adenocarcinoma/epithelial cell surface antigen complementary DNA. Cancer Res 1989;49:314¨C317./ a9 X" g7 Z  a2 c5 R3 u3 n+ C7 p

9 w$ K. U; |+ Y% P: Y$ xSzala S, Froehlich M, Scollon M et al. Molecular cloning of cDNA for the carcinoma-associated antigen GA733¨C2. Proc Natl Acad Sci U S A 1990;87:3542¨C3546.; {: |# C3 }* i9 s/ {/ ^9 N

& c0 N) B) G8 b7 a- OLaemmli UK. Cleavage of structural proteins during the assembly of the head of bacteriophage T4. Nature 1970;227:680¨C685.
) Q6 @% o8 f8 W, O9 d7 r5 s5 _) G: ?) L) W7 \
Towbin H, Staehelin T, Gordon J. Electrophoretic transfer of proteins from polyacrylamide gels to nitrocellulose sheets: Procedure and some applications. Proc Natl Acad Sci U S A 1979;76:4350¨C4354.
$ L* l' X2 D2 N, u/ ^* F. u
( a, t6 m3 D4 V2 U0 o; ?de Boer CJ, van Krieken JH, Janssen-van Rhijn CM et al. Expression of Ep-CAM in normal, regenerating, metaplastic, and neoplastic liver. J Pathol 1999;188:201¨C206.
- X7 r. H+ [- v; x" W
! }5 @: K% T0 O. HTanimizu N, Nishikawa M, Saito H et al. Isolation of hepatoblasts based on the expression of Dlk/Pref-1. J Cell Sci 2003;116:1775¨C1786.
" f+ x+ ], X7 y( |3 d6 x" o3 d! K# {/ `4 x
Tanimizu N, Tsujimura T, Takahide K et al. Expression of Dlk/Pref-1 defines a subpopulation in the oval cell compartment of rat liver. Gene Expr Patterns 2004;5:209¨C218., M; T) E1 s/ q1 k

) Z6 m$ B4 z3 j  T, K3 ^Laborda J, Sausville EA, Hoffman T et al. dlk, a putative mammalian homeotic gene differentially expressed in small cell lung carcinoma and neuroendocrine tumor cell line. J Biol Chem 1993;268:3817¨C3820.
# H/ n; [/ B  }* I, z2 E, |" I; V( L9 D2 x7 L
Smas CM, Sul HS. Pref-1, a protein containing EGF-like repeats, inhibits adipocyte differentiation. Cell 1993;73:725¨C734.
0 F, A& b6 }5 S; b% }! X
; s' b6 G& P9 ~Sicklick JK, Li Y-X, Melhem A et al. Hedgehog signaling maintains resident hepatic progenitors throughout life. Am J Physiol Gastrointest Liver Physiol 2006;290:G859¨CG870.4 \/ U  E& H: ]: X5 R

  q: o' M0 W  L( \' f( l; PMorita K, Furuse M, Fujimoto K et al. Claudin multigene family encoding four-transmembrane domain protein components of tight junction strands. Proc Natl Acad Sci U S A 1999;96:511¨C516.6 R/ F) e) C. s- W) Q. z% a

! L; F+ R- Q) oGonzalez-Mariscal L, Betanzos A, Nava P et al. Tight junction proteins. Prog Biophys Mol Biol 2003;81:1¨C44.$ ]! c6 V% e: G" U; z0 Z2 R

. {! q  y- G5 Z1 I! u0 FTakaki Y, Hirai S, Manabe N et al. Dynamic changes in protein components of the tight junction during liver regeneration. Cell Tissue Res 2001;305:399¨C409.% ?) x' I3 U/ ^2 D

6 T- \) A5 f% S+ eSakamoto S, Yachi A, Anzai T et al. AFP-producing cells in hepatitis and in liver cirrhosis. Ann N Y Acad Sci 1975;259:253¨C258.
  \5 u8 z. s4 m; {/ M! K  q& @
$ L8 l4 s. t7 n7 z7 ~0 T. X1 p3 oKubota H, Storms RW, Reid LM. Variant forms of alpha-fetoprotein transcripts expressed in human hematopoietic progenitors. Implications for their developmental potential towards endoderm. J Biol Chem 2002;277:27629¨C27635.
& K6 L. S8 a3 P6 D( h0 F) q' p5 B+ T3 c/ H8 Z; w( ~6 ^8 Q$ i
Krusekopf S, Roots I, Kleeberg U. Differential drug-induced mRNA expression of human CYP3A4 compared to CYP3A5, CYP3A7 and CYP3A43. Eur J Pharmacol 2003;466:7¨C12., V( Y# }. k- [- D; v
3 o7 _; N- _: h& y( F3 _; `
Racine-Samson L, Scoazec JY, D'Errico A et al. The metabolic organization of the adult human liver: A comparative study of normal, fibrotic, and cirrhotic liver tissue. Hepatology 1996;24:104¨C113.

Rank: 2

积分
163 
威望
163  
包包
1852  
沙发
发表于 2015-5-26 12:41 |只看该作者
回个帖子支持一下!

Rank: 2

积分
161 
威望
161  
包包
1862  
藤椅
发表于 2015-6-29 20:41 |只看该作者
我帮你 喝喝  

Rank: 2

积分
75 
威望
75  
包包
2118  
板凳
发表于 2015-7-20 16:08 |只看该作者
干细胞之家微信公众号
干细胞疾病模型

Rank: 2

积分
116 
威望
116  
包包
1832  
报纸
发表于 2015-7-27 13:53 |只看该作者
楼上的稍等啦  

Rank: 2

积分
166 
威望
166  
包包
1997  
地板
发表于 2015-7-27 17:24 |只看该作者
拿把椅子看表演

Rank: 2

积分
75 
威望
75  
包包
2118  
7
发表于 2015-7-29 11:28 |只看该作者
(*^__^*) 嘻嘻……  

Rank: 2

积分
163 
威望
163  
包包
1852  
8
发表于 2015-8-13 12:40 |只看该作者
正好你开咯这样的帖  

Rank: 2

积分
80 
威望
80  
包包
1719  
9
发表于 2015-8-24 15:01 |只看该作者
朕要休息了..............  

Rank: 2

积分
107 
威望
107  
包包
1889  
10
发表于 2015-9-3 21:35 |只看该作者
心脏干细胞
‹ 上一主题|下一主题
你需要登录后才可以回帖 登录 | 注册
验证问答 换一个

Archiver|干细胞之家 ( 吉ICP备2021004615号-3 )

GMT+8, 2025-6-10 16:28

Powered by Discuz! X1.5

© 2001-2010 Comsenz Inc.