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: 487867|回复: 246
go

Ataxia telangiectasia-mutated, a DNA damage-inducible kinase, contributes to hig [复制链接]

Rank: 1

积分
威望
0  
包包
0  
楼主
发表于 2009-4-21 13:05 |只看该作者 |倒序浏览 |打印
Laboratory of Kidney and Electrolyte Metabolism, National Heart Lung and Blood Institute, and Oral and Pharyngeal Cancer Branch, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, Maryland8 M) K1 q2 I, e% U- R8 E
1 g  \/ a/ Z2 D- M. w; D
ABSTRACT* r+ F0 \* o# h1 x* S) z& C
) G* \* g: t$ j6 s3 E: L: a
High NaCl activates the transcription factor tonicity-responsive enhancer/osmotic response element binding protein (TonEBP/OREBP) by increasing its abundance and transactivation, the latter signaled by a variety of protein kinases. In addition, high NaCl causes TonEBP/OREBP to translocate into the nucleus, but little is known about the signals directing this translocation. The result is increased transcription of protective genes, including those involved in accumulation of organic osmolytes. High NaCl also damages DNA, and DNA damage activates ataxia telangiectasia-mutated (ATM) kinase through autophosphorylation on serine 1981. We previously found that ATM is involved in the high NaCl-induced increase in TonEBP/OREBP transactivation. The purpose of the present studies was to test whether ATM is also involved in high NaCl-induced TonEBP/OREBP nuclear translocation. We quantified TonEBP/OREBP in nuclear and cytoplasmic extracts from cultured cells by Western blot analysis. In COS-7 cells, wortmannin, an inhibitor of ATM, reduces high NaCl-induced nuclear translocation of TonEBP/OREBP. We used AT cells (in which ATM is inactive) to test the specificity of this effect. Nuclear translocation of native TonEBP/OREBP and of its recombinant NH2-terminal rel homology domain, which contains the nuclear localization signal, is reduced in AT cells and is restored when the cells are reconstituted with functional ATM. In conclusion, activation of ATM contributes to high NaCl-induced nuclear translocation of TonEBP/OREBP." \% t6 M( `+ @

  p5 F$ H9 d* I( s! R1 sataxia telangiectasia-mutated kinase; tonicity-responsive enhancer/osmotic response element binding protein; wortmannin; AT cells4 f0 w2 Q6 c# ^1 L/ U
  g$ D- |* I# A8 Q0 }) Y5 ~! o. K
NaClCONCENTRATION IS NORMALLY high in renal inner medullary interstitial fluid, where it energizes the urinary concentrating mechanism, and it varies with concentration of the urine (4). Such high levels of NaCl can be deadly for cells (4, 20, 22), yet the inner medullary cells evidently survive and function in vivo, dependent on a number of protective mechanisms, including accumulation of compatible organic osmolytes (26), which protect cells by normalizing their internal inorganic ion concentration, thus compensating for the hypertonicity caused by high NaCl (26). The renal-compatible organic osmolytes include glycine betaine (betaine), myo-inositol (inositol), and sorbitol (1). Accumulation of these renal organic osmolytes is regulated by a transcription factor, tonicity-responsive enhancer/osmotic response element binding protein (TonEBP/OREBP) (16, 21). TonEBP/OREBP initiates the accumulation of compatible organic osmolytes by increasing transcription of the betaine/-aminobutyric acid transporter (BGT1), the sodium-myo-inositol cotransporter (SMIT), and aldose reductase (AR) (5, 16, 21). BGT1 and SMIT transport betaine and inositol, respectively, into the cells, and AR catalyzes conversion of glucose to sorbitol.
$ p2 Q4 r1 ]: J2 l' ~
: m0 a, V& U; c$ _; j; |5 _3 PRegulation of TonEBP/OREBP transcriptional activity is complex. Within 30 min of hypertonicity, TonEBP/OREBP becomes phosphorylated and translocates into the nucleus (9, 16, 21). Some hours later, TonEBP/OREBP mRNA and protein abundance increase (16, 21). Also, hypertonicity increases transactivation of TonEBP/OREBP, associated with phosphorylation of its transactivation domain (TAD) (13). Several protein kinases are known to contribute to the hypertonicity-induced increase in TonEBP/OREBP transcriptional activity and transactivation, including p38 (15, 23), Fyn (15), ataxia telangiectasia-mutated kinase (ATM) (14), and protein kinase A (PKAc) (12), none of them alone being sufficient for full activation (12).
/ l# i, O6 u6 ]/ q& c2 l% c; B  H% y5 ^( d3 J
Much less is known about how high NaCl signals translocation of TonEBP/OREBP. MG-132 inhibits high NaCl-induced nuclear translocation of TonEBP/OREBP in Madin-Darby canine kidney (MDCK) (25) cells, implicating proteasome activity (25). Cyclosporin A has a similar effect in MDCK cells (24). The proteins affected by MG-132 and cyclosporin A have not yet been identified. Considering the multiplicity of kinases that signal high NaCl-induced increase in TonEBP/OREBP transactivation, it seemed probable to us that there might also be additional pathways that signal its nuclear translocation, possibly including kinases. Therefore, in the present studies we tested the effects of wortmannin on high NaCl-induced translocation of TonEBP/OREBP in COS-7 cells as a start toward identifying them. We found that wortmannin inhibits high NaCl-induced nuclear localization of TonEBP/OREBP. Because wortmannin inhibits ATM, we tested for a specific role of ATM, using AT cells, which lack functional ATM. The results confirm that ATM is involved in signaling nuclear translocation of TonEBP/OREBP but that other targets of wortmannin also play a role.) z7 C* S& ^0 {6 U/ q

8 C& [8 I% `; P- K" z& k, MMATERIALS AND METHODS
0 y  c+ a$ `$ H5 X1 b- s6 J
5 L& I/ E* n8 ~( E, fCell culture. COS-7 cells (American Type Culture Collection) were maintained in Dulbecco*s modified Eagle*s medium and AT cells, in which one allele of ATM is truncated at amino acid 1774 of 3056, the other at amino acid 2769 (GM09607 B, Coriell Cell Repositories, Camden, NJ), were maintained in Eagle*s minimum essential medium according to instructions of the suppliers. Osmolality of the basal medium was 300 mosmol/kgH2O. NaCl was added to prepare 500-mosmol/kgH2O medium, and a 200-mosmol/kgH2O medium (low NaCl) was specially prepared by BioFluids (Rockville, MD). Wortmannin (Calbiochem) or MG-132 (Sigma) was solubilized with Me2SO (DMSO), and the same final concentration of DMSO (+ u7 [  u- t% g/ }# u) ?
) ^$ ]+ Q9 C* t1 Z2 n% C! ?7 C8 y" U
Plasmids and transfection. Human TonEBP/OREBP cDNA clone KIAA0827 was a gift of T. Nagase (Kazusa DNA Research Institute, Chiba, Japan), and pcDNA3 expression vector containing wild-type Flag-ATM (6) was a gift of M. B. Kastan (St. Jude Children*s Research Hospital). A sequence coding for TonEBP/OREBP amino acids 1–547 from KIAA0827 was cloned into pcDNA6 V5-His (Invitrogen) expression vector to generate 1–547-V5. The construct was generated using standard cloning procedures and verified by restriction enzyme digestion and DNA sequencing. DNA was transfected into cells using Effectene according to the supplier*s instructions (Qiagen).
4 C# z4 F! P% E1 m! H4 E  w9 Y2 T% ]/ c  u9 l, G
Western blot analysis. Cells were lysed with Nuclear and Cytoplasmic Extraction Reagents (Pierce) according to the supplier*s instructions, with added protease inhibitor cocktail (Roche Diagnostics) and phosphatase inhibitor cocktails 1 (P2850, Sigma) and 2 (P5726, Sigma). This procedure produces separate nuclear and cytoplasmic fractions. The adequacy of the separation was confirmed by observations that 99.9% of p38 is present in the cytoplasmic fraction and 90.5% of poly(ADP-ribose) polymerase-1 is in the nuclear fraction under all the osmotic conditions that we used. Cytoplasmic protein (20 μg/lane) and nuclear protein (10 μg/lane) were separated on 4–12% Novex Tris-glycine gels and transferred to nitrocellulose membranes (Invitrogen). Western blot analysis was performed according to instructions for the Odyssey Infrared Imaging System. In brief, nonspecific binding was blocked by incubating membranes overnight at 4°C with blocking buffer (Odyssey) diluted 1:1 in phosphate-buffered saline. Membranes were then incubated with anti-NFAT5 (TonEBP/OREBP) rabbit polyclonal antibody (Affinity BioReagents) or anti-V5 mouse monoclonal antibody (Invitrogen) for 2 h at room temperature. After being washed with 0.1% Tween 20 in phosphate-buffered saline, blots were incubated with Alexa Fluor 680 goat anti-rabbit IgG or Alexa Fluor 780 goat anti-mouse IgG (Molecular Probes) for 1 h in the dark. Blots were visualized and quantitated using an LI-COR Odyssey Infrared Imager. Alternatively, membranes were washed and exposed to secondary antibody [goat anti-rabbit IgG conjugated to horseradish peroxidase (no. 31463, diluted to 1:5,000, Pierce)] for 1 h at room temperature. After being washed, bands were visualized using enhanced chemiluminescence (Amersham Biosciences) with band densities determined by laser densitometry (Personal Densitometer SI)., K/ y4 G$ ]! R" {+ ]" j
) t+ d4 L  i* a5 _: |
Calculation of nuclear/cytoplasmic ratios. From the relative quantities of TonEBP/OREBP and the relative volumes of the cytoplasmic or nuclear extract, we calculated the relative total amounts of TonEBP/OREBP in the cytoplasmic and nuclear fractions and the nuclear/cytoplasmic ratio.5 A/ l. A7 l% u9 L3 {6 O4 D

# h5 j6 ~4 b% c. X9 Z" D" J- gStatistical analysis. Data were compared by multiple comparison tests, ANOVA (followed by a Student-Newman-Keuls posttest), or false discovery rate (8). Results are expressed as means ± SE (n = number of independent experiments). Differences were considered significant for P  0.05.
1 @! k" o6 b# j  ]) `" }
# S1 F0 x0 F, h' T$ qRESULTS
8 g9 e# A5 K- K' r$ H- e1 _
( s6 B# S4 M+ C0 t/ }$ JMG-132 does not affect location of TonEBP/OREBP in COS-7 cells but does in Hep G2 cells. High NaCl causes TonEBP/OREBP to translocate from cytoplasm to nucleus (16, 21). Previously, the proteasome inhibitor MG-132 (1 μM) was found to inhibit high NaCl-induced nuclear translocation of TonEBP/OREBP in MDCK cells (25), implicating proteasome activity. We have confirmed this effect in Hep G2 cells (Fig. 1C). However, using COS-7 cells, we do not find any effect of 1 μM MG-132 on nuclear/cytoplasmic ratio of native TonEBP/OREBP at 200, 300, or 500 mosmol/kgH2O (Fig. 1A). We conclude that the effect of MG-132 on high NaCl-induced nuclear translocation of TonEBP/OREBP is cell type dependent.6 s; ?7 b$ K8 T* T: |* O5 o

  w& p+ _$ ?: ~  V: y, K- p9 L/ Y  \Wortmannin decreases high NaCl-induced TonEBP/OREBP translocation in COS-7 cells. ATM contributes to a high NaCl-induced increase in TonEBP/OREBP transcriptional activity (14). At least part of this effect is due to increased transactivation (14). In the present experiments we asked whether ATM also contributes to high NaCl-induced nuclear translocation of TonEBP/OREBP. In support of this possibility, 20 μM wortmannin, which inhibits ATM, significantly decreases nuclear/cytoplasmic ratio of native TonEBP/OREBP in COS-7 cells at 500 mosmol/kgH2O (Fig. 1B). However, because wortmannin also inhibits other kinases, including DNA-PK, phosphatidylinositol 3-kinase (PI3-K), and myosin light chain kinase (MLCK) (10), we sought to examine the possible role of ATM more specifically.6 j2 H% i+ E) J' M5 h
3 N: U3 k: O4 x& \% _! G( e1 ?
Expression of functional ATM contributes to high NaCl-induced nuclear translocation of TonEBP/OREBP. AT cells lack functional ATM, but ATM activity can be reconstituted in these cells by transfection with wild-type ATM (2). We used this system to test for a specific effect of ATM on NaCl-dependent location of TonEBP/OREBP. We transfected AT cells with either empty vector (EV) or functional ATM. In AT cells reconstituted with active ATM, the nuclear/cytoplasmic ratio of native TonEBP/OREBP is significantly greater at 500 mosmol/kgH2O than in AT cells transfected with EV, which lack ATM activity (Fig. 2, A–C). The difference exists from 30 min to at least 16 h after osmolality is increased by elevating NaCl. There is no difference at 300 or 200 mosmol/kgH2O. We conclude that ATM specifically contributes to sustained high NaCl-induced nuclear translocation of TonEBP/OREBP.$ v: B% x6 p4 _+ p

# E/ V1 W0 ]3 h& @( sATM signals high NaCl-induced nuclear translocation of recombinant TonEBP/OREBP that is truncated to contain only amino acids 1–547. High NaCl-induced translocation of TonEBP/OREBP involves its NH2-terminal third, which contains the nuclear localization signal (16, 21). Recombinant TonEBP/OREBP, containing amino acids 77–547 of TonEBP/OREBP, was previously shown to translocate into the nucleus when NaCl concentration is increased (16). We tested whether ATM might contribute to high NaCl-induced nuclear translocation of recombinant TonEBP/OREBP 1–547-V5. Increasing osmolality from 300 to 500 mosmol/kgH2O by adding NaCl increases the nuclear/cytoplasmic ratio of TonEBP/OREBP 1–547-V5 in AT cells, and the increase is greater when the cells are reconstituted with active ATM (Fig. 2D). We conclude that ATM signals increased the nuclear/cytoplasmic ratio of TonEBP/OREBP through sites within amino acids 1–547, which does not, however, exclude the possibility of modulation via effects of ATM on TonEBP/OREBP-548–1531.
$ y8 W  r& G+ H7 V% s, o) q# j$ r% U9 C) K
In the absence of functional ATM, wortmannin increases high NaCl-induced translocation of TonEBP/OREBP. For the sake of completeness, we also tested the effect of wortmannin on the nuclear-to-cytoplasmic ratio of TonEBP/OREBP in AT cells that are not reconstituted with functional ATM. To our surprise, wortmannin increases the ratio under those conditions (Fig. 3A), which is the opposite of the effect in AT cells reconstituted with functional ATM (Fig. 3B). We speculate that some wortmannin-inhibitable signaling molecule, other than ATM, acts to inhibit high NaCl-induced nuclear translocation of TonEBP/OREBP. Possible candidates are DNA-PK, PI3-K, and MLCK, all of which are inhibited by wortmannin (10).
2 a5 F" o3 K* L% O* ^8 M( V) s! Q+ ^6 N+ y/ M
DISCUSSION6 u0 [- C7 `/ a6 A* N+ w- u

9 O; ]4 ~) q. E( z; p1 R$ a4 K. |; gHigh NaCl damages DNA in cell culture and in vivo. Acute elevation of NaCl causes DNA double-strand breaks in cell culture (11, 17), and the DNA breaks persist after the cells adapt to high NaCl (11). When NaCl is lowered, however, the breaks are rapidly repaired. Renal inner medullary cells in vivo are normally exposed to a variable, but always high, level of NaCl (4). While NaCl is at its normal high level in the mouse inner medulla in vivo, the cells contain numerous DNA breaks (11), but when NaCl is lowered by the diuretic furosemide, the breaks are rapidly repaired (11). Thus both in cell culture and in vivo, high NaCl causes persistent DNA damage that is not repaired unless the level of NaCl is reduced.
! U9 O+ @- l1 @$ h0 D2 m1 x1 S. b7 H; E+ J, }- ~# K9 q% f3 a
High NaCl activates ATM. ATM protein kinase mediates responses to ionizing radiation-induced DNA damage in mammalian cells (2). Mutations of ATM that inactivate it cause the human disease ataxia-telangiectasia, in which cells are hypersensitive to DNA damaging agents. In unirradiated cells, ATM is held inactive as a dimer or higher-order multimer, with the kinase domain bound to a region surrounding serine 1981. Ionizing radiation induces rapid intermolecular autophosphorylation of serine 1981, resulting in dimer dissociation and initiation of ATM kinase activity (2). High NaCl, which damages DNA, also activates ATM via autophosphorylation on serine 1981 (14). Activated ATM phosphorylates a number of proteins, including p53, murine double minute-2, Csk homologous kinase 2, breast cancer 1, and NBS1, contributing to DNA repair, cell cycle delay, and apoptosis (3, 6, 7, 18, 19).; T4 _1 N8 \1 [% W7 K4 u. m% z* x

2 L- h0 t7 v, P* h! y- _, ?How does ATM contribute to high NaCl-induced nuclear localization of TonEBP/OREBP ATM contributes to both high NaCl-induced nuclear translocation of TonEBP/OREBP (Fig. 2, A–C) and high NaCl-induced increase in TonEBP/OREBP transactivation (14). The effect on translocation is mediated by sites within amino acids 1–547 of TonEBP/OREBP (Fig. 2D), whereas the effect on transactivation is mediated by sites within amino acids 548–1531 (14). We were able to identify consensus ATM phosphorylation sites at amino acids serine 1197, serine 1247, and serine 1367 of TonEBP/OREBP (14), and transcriptional activity of TonEBP/OREBP is increased less by high NaCl when these sites are mutated to prevent their phosphorylation (14). However, we are unable to identify clear consensus ATM phosphorylation sites within amino acids 1–547 of TonEBP/OREBP. We are left with the possibilities that ATM is affecting high NaCl-induced nuclear translocation of TonEBP/OREBP indirectly, through some intermediary signaling protein, or, if directly, through an ATM phosphorylation site that we do not recognize.) _4 B+ _$ k1 M6 x) D& n

2 F3 u! D$ l8 k( y8 Q: U" p4 T! IUnexpected wortmannin-induced increase in TonEBP/OREBP translocation in cells lacking ATM activity. ATM contributes to high NaCl-induced nuclear translocation of TonEBP/OREBP (Fig. 2, A–C). Wortmannin inhibits ATM, which can explain its inhibition of high NaCl-induced nuclear localization of TonEBP/OREBP in cells expressing active ATM (Figs. 1B and 3B). However, it also increases high NaCl-induced nuclear localization of TonEBP/OREBP in AT cells (Fig. 3A), which presumably involves some wortmannin-inhibitable signaling molecule "other" than ATM. Other known targets of wortmannin include DNA-dependent protein kinase catalytic subunit (DNA-PKcs), PI3-K, and MLCK (10). The question is why inhibition by wortmannin of a signal that enhances NaCl-induced nuclear translocation of TonEBP/OREBP (via ATM) does not cancel the effect of a signal that inhibits it (the "other"). We speculate that interaction of full-length ATM with the other normally protects the other from wortmannin, so that this effect of wortmannin is only evident in AT cells.
" }3 K* H  }1 X/ P) Y! s1 K5 s: U0 n% j
FOOTNOTES
. x- c/ F; @' k4 f
! k. s( F, L6 s' @9 A! b+ \3 j2 U+ h2 _The costs of publication of this article were defrayed in part by the payment of page charges. The article must therefore be hereby marked "advertisement" in accordance with 18 U.S.C. Section 1734 solely to indicate this fact.7 c3 Q! i( A2 `5 d( i& O, h9 @

1 R+ \/ _7 [7 N% k9 f, m( RREFERENCES
; a7 W' u" J% J' Z: `! z8 Q( ]8 K# O) o0 I4 H* U: E9 h
Bagnasco S, Balaban R, Fales HM, Yang YM, and Burg M. Predominant osmotically active organic solutes in rat and rabbit renal medullas. J Biol Chem 261: 5872–5877, 1986.. }- c& ?/ _) n

1 A% o3 A* f% k. M! b7 V# ~7 YBakkenist CJ and Kastan MB. DNA damage activates ATM through intermolecular autophosphorylation and dimer dissociation. Nature 421: 499–506, 2003.
2 @9 P4 J( ~" w
, L/ _1 r" L: D3 j- ?6 OBanin S, Moyal L, Shieh S, Taya Y, Anderson CW, Chessa L, Smorodinsky NI, Prives C, Reiss Y, Shiloh Y, and Ziv Y. Enhanced phosphorylation of p53 by ATM in response to DNA damage. Science 281: 1674–1677, 1998.! F8 \- l$ x: D7 |
6 n! B1 ~% V  |* H4 M& f
Bankir L. Urea and the kidney. In: The Kidney, edited by Brenner BM and Rector F. Philadelphia, PA: Saunders, 1996, p. 571–606.
7 [4 Q" l( x5 |9 o; Z; U! ^
. p) u) R1 B9 k# P% XBurg MB, Kwon ED, and Kultz D. Regulation of gene expression by hypertonicity. Annu Rev Physiol 59: 437–455, 1997.
" F8 ^7 S* Y* V) f8 o0 i2 a7 S
% @/ v! I  V2 w- sCanman CE, Lim DS, Cimprich KA, Taya Y, Tamai K, Sakaguchi K, Appella E, Kastan MB, and Siliciano JD. Activation of the ATM kinase by ionizing radiation and phosphorylation of p53. Science 281: 1677–1679, 1998.
7 j! G6 t& n" e8 ]: }1 J. Y- E/ _/ u# ?9 F$ a% _
Chehab NH, Malikzay A, Appel M, and Halazonetis TD. Chk2/hCds1 functions as a DNA damage checkpoint in G1 by stabilizing p53. Genes Dev 14: 278–288, 2000.4 S# t4 `' p  {  j4 F8 F  d
: s3 O' B- F$ O- a- i. x
Curran-Everett D. Multiple comparisons: philosophies and illustrations. Am J Physiol Regul Integr Comp Physiol 279: R1–R8, 2000." G3 o9 K/ V" w$ S" Z3 t8 K' {1 X

8 v. R% Y* _1 Y% M# cDahl SC, Handler JS, and Kwon HM. Hypertonicity-induced phosphorylation and nuclear localization of the transcription factor TonEBP. Am J Physiol Cell Physiol 280: C248–C253, 2001.# Y  _6 y+ ^# z4 h
( p9 \6 U% [6 E% k, L
Davies SP, Reddy H, Caivano M, and Cohen P. Specificity and mechanism of action of some commonly used protein kinase inhibitors. Biochem J 351: 95–105, 2000.' ]) I9 T0 h+ j- E" h. C

0 C0 m0 P* S, C! R9 o* ODmitrieva NI, Cai Q, and Burg MB. Cells adapted to high NaCl have many DNA breaks and impaired DNA repair both in cell culture and in vivo. Proc Natl Acad Sci USA 101: 2317–2322, 2004." l/ I! G# f. x& y+ ]0 w2 |

$ S' x0 I3 u5 O( \  F! lFerraris JD, Persaud P, Williams CK, Chen Y, and Burg MB. cAMP-independent role of PKA in tonicity-induced transactivation of tonicity-responsive enhancer/osmotic response element-binding protein. Proc Natl Acad Sci USA 99: 16800–16805, 2002.
4 S2 g* V( \6 g" m' x
) K6 g8 H- v1 G/ t' o9 BFerraris JD, Williams CK, Persaud P, Zhang Z, Chen Y, and Burg MB. Activity of the TonEBP/OREBP transactivation domain varies directly with extracellular NaCl concentration. Proc Natl Acad Sci USA 99: 739–744, 2002.
8 |' e: u% W  I2 S
  A' R+ a: J! B, AIrarrazabal CE, Liu JC, Burg MB, and Ferraris JD. ATM, a DNA damage-inducible kinase, contributes to activation by high NaCl of the transcription factor TonEBP/OREBP. Proc Natl Acad Sci USA 101: 8809–8814, 2004.
8 R) W7 q4 U$ q* S3 G% J3 g
. j( M- b. n" u6 U: l  S, uKo BC, Lam AK, Kapus A, Fan L, Chung SK, and Chung SS. Fyn and p38 signaling are both required for maximal hypertonic activation of the OREBP/TonEBP. J Biol Chem 277: 46085–46092, 2002.
% c" K, O% H, g, {: |1 @
5 c: L( Z9 g; r0 O8 d. M1 wKo BC, Turck CW, Lee KW, Yang Y, and Chung SS. Purification, identification, and characterization of an osmotic response element binding protein. Biochem Biophys Res Commun 270: 52–61, 2000.8 }  U7 F, H9 Z
" o4 J( z2 t, @9 q4 ~6 }7 j
Kultz D and Chakravarty D. Hyperosmolality in the form of elevated NaCl but not urea causes DNA damage in murine kidney cells. Proc Natl Acad Sci USA 98: 1999–2004, 2001.
) V/ @: p( ^. d: U! H6 _
* M+ Q3 F) P9 iMatsuoka S, Rotman G, Ogawa A, Shiloh Y, Tamai K, and Elledge SJ. Ataxia telangiectasia-mutated phosphorylates Chk2 in vivo and in vitro. Proc Natl Acad Sci USA 97: 10389–10394, 2000.% W& |! s; M6 _4 i9 T& f

7 N: \. l8 H9 r- f; k8 ~. BMaya R, Balass M, Kim ST, Shkedy D, Leal JF, Shifman O, Moas M, Buschmann T, Ronai Z, Shiloh Y, Kastan MB, Katzir E, and Oren M. ATM-dependent phosphorylation of Mdm2 on serine 395: role in p53 activation by DNA damage. Genes Dev 15: 1067–1077, 2001.
6 b( u$ q* g; k" Z/ e- p; ^. e2 ~/ C
0 w4 ~. B6 `  @& e- cMichea L, Ferguson DR, Peters EM, Andrews PM, Kirby MR, and Burg MB. Cell cycle delay and apoptosis are induced by high salt and urea in renal medullary cells. Am J Physiol Renal Physiol 278: F209–F218, 2000.
0 P0 Z) @8 G  U
2 O" o0 W5 F2 M4 n  [, T  X, U# dMiyakawa H, Woo SK, Dahl SC, Handler JS, and Kwon HM. Tonicity-responsive enhancer binding protein, a Rel-like protein that stimulates transcription in response to hypertonicity. Proc Natl Acad Sci USA 96: 2538–2542, 1999.1 M4 L5 E' q& {$ U4 m* X

: k3 V1 J5 O/ GSantos BC, Chevaile A, Hebert MJ, Zagajeski J, and Gullans SR. A combination of NaCl and urea enhances survival of IMCD cells to hyperosmolality. Am J Physiol Renal Physiol 274: F1167–F1173, 1998.
( X- |0 }+ Q* {1 x) y+ R. D8 m
; m8 R/ N& A9 K; f5 VSheikh-Hamad D, Di Mari J, Suki WN, Safirstein R, Watts BA III, and Rouse D. p38 Kinase activity is essential for osmotic induction of mRNAs for HSP70 and transporter for organic solute betaine in Madin-Darby canine kidney cells. J Biol Chem 273: 1832–1837, 1998.0 F0 L7 k  _) Q, O/ ]

7 l- O7 W6 }3 l( u9 u2 E0 q4 \Sheikh-Hamad D, Nadkarni V, Choi YJ, Truong LD, Wideman C, Hodjati R, and Gabbay KH. Cyclosporine A inhibits the adaptive responses to hypertonicity: a potential mechanism of nephrotoxicity. J Am Soc Nephrol 12: 2732–2741, 2001.
5 W% l( h9 _( X8 O: I# q" f( N! y) y0 u. ~( I5 ^/ `1 @% J6 @
Woo SK, Maouyo D, Handler JS, and Kwon HM. Nuclear redistribution of tonicity-responsive enhancer binding protein requires proteasome activity. Am J Physiol Cell Physiol 278: C323–C330, 2000.& F- E5 _# A( c1 f
2 l6 e" W8 {) ?# a* t
Yancey PH, Clark ME, Hand SC, Bowlus RD, and Somero GN. Living with water stress: evolution of osmolyte systems. Science 217: 1214–1222, 1982.(Zheng Zhang, Joan D. Ferr)

Rank: 2

积分
116 
威望
116  
包包
1832  
沙发
发表于 2015-6-18 21:33 |只看该作者
嘿...反了反了,,,,  

Rank: 2

积分
64 
威望
64  
包包
1734  
藤椅
发表于 2015-6-25 14:11 |只看该作者
内皮祖细胞

Rank: 2

积分
163 
威望
163  
包包
1852  
板凳
发表于 2015-7-25 10:51 |只看该作者
干细胞之家微信公众号
呵呵 哪天得看看 `~~~~  

Rank: 2

积分
122 
威望
122  
包包
1876  
报纸
发表于 2015-7-30 22:34 |只看该作者
说的真有道理啊!

Rank: 2

积分
79 
威望
79  
包包
1769  
地板
发表于 2015-8-31 14:42 |只看该作者
希望可以用些时间了~````  

Rank: 2

积分
68 
威望
68  
包包
1752  
7
发表于 2015-9-9 18:34 |只看该作者
感谢党和人民的关爱~~~  

Rank: 2

积分
72 
威望
72  
包包
1730  
8
发表于 2015-9-10 18:11 |只看该作者
好啊,,不错、、、、  

Rank: 2

积分
75 
威望
75  
包包
2118  
9
发表于 2015-12-9 21:35 |只看该作者
留个脚印```````  

Rank: 2

积分
75 
威望
75  
包包
2118  
10
发表于 2015-12-24 10:01 |只看该作者
哈哈 瞧你说的~~~  
‹ 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, 2024-4-29 06:51

Powered by Discuz! X1.5

© 2001-2010 Comsenz Inc.