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: 486167|回复: 243
go

Cisplatin-induced cell death is EGFR/ src /ERK signaling dependent in mouse prox [复制链接]

Rank: 1

积分
威望
0  
包包
0  
楼主
发表于 2009-4-22 08:10 |只看该作者 |倒序浏览 |打印
作者:Istvan Arany, Judit K. Megyesi, Hideaki Kaneto, Peter M. Price, and Robert L. Safirstein作者单位:1 Department of Internal Medicine, University of Arkansas for Medical Sciences and Central Arkansas Veterans Healthcare System, Little Rock, Arkansas 72205; and 2 Department of Internal Medicine and Therapeutics, Osaka University, Graduate School of Medicine, Osaka 54 Japan & i. D/ G/ i7 }3 G& z4 \; z
                  % @9 c4 P  C6 t: V1 B
                  
4 O- Q2 T1 K! R            I* R7 L9 Q  L; _- n) F; i. E7 i
                         + g; `& q! a+ ]
            
5 K0 E7 k; K& G% y& s! ^# j            
9 M, [, q$ o) c  h1 X% V# x$ A8 R+ V            
9 I9 |- X* R* K, ]1 ^  r( q6 _) a            1 }9 _0 z0 O6 U$ \( B4 s
                      8 f/ ~# V# y) ^( n9 H1 {7 Z9 ^, p
        / w6 m# R1 R- J( A- ^! y
        
1 J4 g# h' Q' N        
# e9 V9 L9 W: V8 R, ^. V          【摘要】
3 K  u% W8 C" x6 @8 ?2 l      Cisplatin treatment induces extensive death of the proximal tubules in mice. We also demonstrated that treatment of immortalized mouse proximal tubule cells (TKPTS) with 25 µM cisplatin induces apoptotic death in vitro. Here, we demonstrate that members of the MAPKs such as ERK, JNK, and p38 are all activated after cisplatin treatment both in vivo and in vitro. Because MAPKs mediate cell survival and death, we studied their role in cisplatin-induced cell death in vitro. Apoptosis was confirmed by cell morphology, fluorescence-activated cell-sorting analysis, annexin V/propidium iodide binding, and caspase-3 activation in TKPTS cells. Inhibition of ERK, but not JNK or p38, abolished caspase-3 activation and apoptotic death, suggesting a prodeath role of ERK in cisplatin-induced injury. We also determined that cisplatin-induced ERK as well as caspase-3 activation are epidermal growth factor receptor (EGFR) and c- src dependent because inhibition of these genes inhibited ERK and caspase-3 activation and attenuated apoptotic death. These results suggest that caspase-3 mediates cisplatin-induced cell death in TKPTS cells via an EGFR/src/ERK-dependent pathway. We also suggest that the prodeath effect of ERK is injury type dependent because during oxidant injury, ERK supports survival rather than death in the same cells. We propose that injury-specific outcome diverges downstream from ERK in cisplatin- or H 2 O 2 -mediated cell survival and death.
2 |8 w  J% x1 |. _          【关键词】 apoptosis mitogenactivating protein kinase epidermal growth factor receptor c src8 Y. I( ?' f* a! t, G" d- P
                  THE CHEMOTHERAPEUTIC AGENT cisplatin is commonly used against various solid tumors. Despite its potent antitumor activity, its clinical use is hampered by nephrotoxicity. Recent surveys indicate that 20% of acute renal failure (ARF) cases among hospitalized patients is due to cisplatin nephrotoxicity ( 5 ).( E% g, _' ~. C5 u; K

) b0 X* z9 j! C9 T% sThe cisplatin-related nephrotoxicity is most probably due to its high accumulation in the kidney, especially in the S3 segment, which undergoes extensive necrosis ( 3 ). Mice injected with cisplatin undergo ARF ( 21 ), and immortalized mouse proximal tubule cells (TKPTS) undergo extensive death 24 h after cisplatin treatment ( 27 ).3 q5 S' z4 U( Z4 d

$ ~0 x* t% i. `0 v  b+ F* WVarious types of stress signals activate MAPKs that serve to coordinate cellular responses to those stimuli ( 8, 19 ). Generally, activation of ERK confers survival advantage to cells during most stress conditions ( 6, 8 ), whereas activation of JNK or p38MAPK is associated with cell death ( 9, 37 ). Cisplatin treatment has been found to activate ERK, JNK, and p38 in various systems ( 25, 29, 35, 38 ) including primary cultures of mouse proximal tubule cells ( 23 ). The protective role of ERK in cisplatin-induced apoptosis was suggested by studies that demonstrated pharmacological inhibition of ERK-sensitized cells to cisplatin ( 20, 25, 26 ), although other studies demonstrated a prodeath function of ERK as well ( 23, 35 ). Still, other groups proposed that activation of JNK, but not ERK, was responsible for apoptotic effects of cisplatin ( 15, 16, 29, 38 ), although its protective role has also been demonstrated ( 15 ). These studies relied heavily on pharmacological inhibition of MAPKs and thus may not have the necessary specificity.; T' L' j/ J) Q

9 q. G$ E' v, X+ J' r& XAccordingly, the aim of this study was to reexamine the role of MAPKs in the death of mouse proximal tubule cells following cisplatin treatment by using additional direct approaches. We also sought to define the upstream regulation of MAPK activation.- K9 Y9 @1 E; s% A
* W. E! @" G" _8 w8 P: D* }  b
MATERIALS AND METHODS
; D+ x1 R. q9 q7 k1 s# L) {% s! _) l* F# K
Animals and treatment. Six- to eight-week-old male 129Sv mice received a single intraperitoneal injection of 20 mg/kg cisplatin ( 27 ). Control and treated mice were killed 1, 2, and 3 days after treatment. The animals used in these studies (IACUC protocol 5-02-8) were housed at the Veterinary Medical Unit at the John McClellan Veterans Hospital at Little Rock. Animals were painlessly killed in accordance with methods of euthanasia approved by the Panel of Euthanasia of the American Veterinary Medical Association.9 K! \4 L4 K" U
/ d( t) X8 G1 p% u! E
Cells and treatment. The immortalized mouse proximal tubule cell line (TKPTS) was a gift from Dr. Bello-Reuss ( 11 ) and was maintained as described previously ( 2, 10 ). Subconfluent cultures were treated with 25 µM cisplatin for the time points described in RESULTS.0 T" Q0 R" P+ z4 X' x0 m
  R" N1 u3 ~4 T% P" X1 _9 T) M6 W: T9 r
Western blotting. Kidneys were removed and homogenized in a RIPA buffer that contained 100 µg/ml phenylmethylsulfonyl fluoride (Sigma, St. Louis, MO), 100 mM sodium orthovanadate (Sigma), and 50 µl/ml of proteinase inhibitor cocktail (Sigma) as described earlier ( 2 ). The total lysates were used for Western blotting. Similarly, monolayers of TKPTS cells were lysed in a RIPA buffer. Protein content was determined by using a Bio-Rad Protein Determination Assay (Bio-Rad, Hercules, CA) as described earlier ( 2 ). One-hundred micrograms of protein from cell or tissue lysates were separated by SDS-PAGE electrophoresis and transferred to a PVDF membrane (Bio-Rad). The filters were hybridized with the appropriate primary antibodies followed by a horseradish peroxidase (HRP)-conjugatedsecondary antibody. The bands were visualized by an ECL method (Amersham) and quantified by densitometry (UnScan-It, Silk Scientific, Ore, UT). Antibodies against ERK1/2, phospho-ERK1/2 (Thr202/Tyr204), JNK, p38, phospho-p38 (Thr180/Tyr182), and caspase-3 were purchased from Cell Signaling Technology (Beverly, MA). Antibodies against phospho-JNK (Thr183/Tyr185), phospho-epidermal growth factor receptor (EGFR) (Tyr1173), and EGFR were purchased from Santa Cruz Biotechnology (Santa Cruz, CA). Anti-rabbit HRP-conjugated secondary antibody was purchased from Cell Signaling Technology (Beverly, MA), whereas anti-mouse and anti-goat HRP-conjugated secondary antibodies were purchased from Santa Cruz Biotechnology.9 d; v2 N+ d  }& t/ X8 [

1 Z+ m: V8 K2 @! B) d: RCell cycle analysis and quantitation of apoptosis. TKPTS cells were collected after trypsinization and fixed in 70% ethanol. After RNase treatment, cells were incubated with 5 µg/ml propidium iodide (PI) and analyzed with a Becton Dickinson FACSCalibur analyzer. The cell cycle distribution was determined using the CellQuest software. Cells in the subG1 phase were considered apoptotic ( 27 ).
! T6 M- l8 W8 D, A$ p; q4 Q2 L' ^& _$ u. G- d
Apoptosis in TKPTS cells was further confirmed using an annexin V/PI binding assay (BD Biosciences). PI and annexin V-FITC fluorescence was quantified by flow cytometry. For each sample, 10,000 events were counted. Cells positive for annexin V but negative for PI were considered apoptotic.1 m% C/ L& n( k1 G" M
% }1 I0 Q6 k% _( o5 Q" T; _/ w
Manipulation of gene activities before cisplatin treatment. To inhibit EGFR or c- src, TKPTS cells were treated with 20 µM AG-1478 or 20 µM PP1 (Biomol Research Laboratories, Plymouth Meeting, PA), respectively, 1 h before cisplatin treatment. To inhibit ERK, cells were pretreated with 50 µM U-0126 (Cell Signaling Technology) 1 h before cisplatin treatment. To inhibit JNK or p38, cells were infected with 50 MOI adenoviruses that contain dominant-negative JNK ( 13 ) or dominant-negative p38 ( 36 ) as described earlier.8 ?$ k5 c' R& B2 {

; o% F1 u5 c2 |  g/ s( F8 c; Q& yStatistical analysis. Statistical differences between the treated and control groups were determined by Student's paired t -test. Differences between means were considered significant if P
1 R- N- e& h( g/ C* B$ T# e
& _+ J  b" Y0 c* e+ ^( N" L+ gRESULTS: O/ f, [& r/ R; q2 t

0 B  W/ E6 V0 J) M5 X  L' L- RCisplatin activates MAPKs in the kidney of cisplatin-treated mice. Mice treated intraperitoneally with a single dose of 20 mg/kg cisplatin were killed 1, 2, or 3 days after treatment. Phosphorylation status of ERK, JNK, and p38 was determined by Western blotting ( Fig. 1 ). Our results show that all three MAPKs are activated by cisplatin compared with the control animals. The activation was sustained during the observation period and preceded the development of ARF ( 21 ). It is important to note that both inhibition of GFR and extensive death of proximal tubules occur only after day 3 of cisplatin treatment ( 21 ).
4 d" r. a: c0 k, B" ]! t' e) c; f) m+ d7 j- l
Fig. 1. Effects of cisplatin treatment on phosphorylation of various MAPKs in the mouse kidney. 129Sv mice were treated with a single intraperitoneal injection of 20 mg/kg cisplatin. Kidneys were removed from control as well as 1-, 2-, and 3-day-treated animals and homogenized. A : total lysates were subjected to SDS-PAGE electrophoresis and Western blotting as described in MATERIALS AND METHODS. Data are representative of 3 independent experiments. B : densitometric analysis of Western blots described in A. Results are expressed as fold increase in phosphorylation compared with the appropriate controls. Phosphorylation is calculated as ratios of the phosphorylated vs. nonphosphorylated forms (means ± SD, n = 3). Dashed line represents the control value. * P
5 d' H& g- i2 w' \8 M# b) A7 }3 A3 [  T3 ]3 J2 i- m: k$ F
Cisplatin induces apoptosis and activates MAPKs in TKPTS cells. Cisplatin treatment damaged proximal tubule cells in vitro as well ( 27 ). Cisplatin induced apoptosis of TKPTS cells 24 h after exposure ( Fig. 2 A ) as was demonstrated previously ( 27 ). The number of apoptotic cells (subG1 fraction) increased from 2.96 to 45.3% 24 h after cisplatin treatment as determined by FACS analysis ( Fig. 2 B and Table 1 ). Also, cisplatin treatment significantly increased the number of annexin V-stained (apoptotic) cells (from 8 to 43%; Fig. 2 C ) and activated caspase-3 ( Fig. 2 D ) 24 h after treatment. Cisplatin treatment, similar to the kidney, significantly increased phosphorylation of all three major MAPKs ( Fig. 3 ), although with different kinetics. A high level of ERK phosphorylation was already apparent at 1 h after treatment, whereas JNK and p38 phosphorylation was elevated later at 6 h after treatment. The elevated phosphorylation was maintained at 24 h posttreatment in all cases. Thus MAPK activation occurred well before obvious cell death in a similar manner both in vivo and in vitro. This allowed us to study the role of MAPKs in cisplatin-induced cell death in vitro.
0 c! L5 M. q6 k, i1 Z5 b' N" z+ v% E  x& l: e/ |% S
Fig. 2. Effects of cisplatin on viability and caspase-3 (casp-3) activation in TKPTS cells. TKPTS cells were treated with 25 µM cisplatin for 24 h. A : cell morphology after cisplatin treatment was determined by light microscopy (magnification x 100). B : FACS analysis was performed to demonstrate and quantitate cell death. The percentage of dead (apoptotic) cell is given. Results are representative of 3 independent experiments. C : annexin V and propidium iodide (PI) staining was determined by flow cytometry. The percentage of apoptotic cells (annexin V positive and PI negative) was calculated. Results are representative of 3 independent experiments. D : activation of caspase-3 was determined by Western blotting as described in MATERIALS AND METHODS. C0, C24, appropriate time controls; T1, T6, and T24, treated for 1, 6, and 24 h. Results are representative of 3 independent experiments.- a3 l6 I( N& d9 ^5 _& g

1 ^% i$ Y# O: ?7 V, `* _; l; ^Table 1. Percentage of apoptotic (subG1) cells after CP treatment  e) `3 @( ^* h! u' a# A% w

3 p1 h( [3 {. G1 A& R% e9 y7 mFig. 3. Activation of MAPKs by cisplatin in TKPTS cells. A : TKPTS cells were treated with 25 µM cisplatin for the time indicated. Western blot analysis was applied to determine phosphorylation of ERK, JNK, and p38 together with the unphosphorylated forms. Data are representative of 3 independent experiments. B : densitometric analysis of Western blots described in A. Results are expressed as fold increase in phosphorylation compared with the appropriate controls. Phosphorylation is calculated as ratios of the phosphorylated vs. nonphosphorylated forms (means ± SD, n = 3). Dashed line represents the control value. * P % q4 G' u) q) F, {6 Z
3 Z# g5 A2 \+ m- y; T8 u, ]
Effects of inhibition of various MAPKs on cisplatin-induced apoptosis and caspase-3 activation in vitro. TKPTS cells were pretreated with 50 µM U-0126 for 1 h before cisplatin treatment to inhibit ERK activation. Previously, we demonstrated complete inhibition of endogenous and injury-induced ERK phosphorylation using the MEK inhibitor U-0126. Also, activation of JNK and p38 was prevented by infection of TKPTS cells with dominant-negative JNK or p38 adenoviruses (data not shown) 24 h before cisplatin treatment. After treatment of these pretreated cells with 25 µM cisplatin for 24 h, inhibition of ERK, but not JNK or p38, ameliorated cisplatin-induced morphological changes 24 h after treatment ( Fig. 4 ). FACS analysis demonstrated that the percentage of cisplatin-induced apoptotic cells (45.3%) was significantly reduced in U-0126-pretreated (28.6%) cells but not in cells that were pretreated with either adeno-dnJNK or adeno-dnp38 ( Table 1 ). Neither U-0126, adeno-dnJNK, nor adeno-dnp38 induced apoptosis when applied alone ( Table 1 ). U-0126 significantly decreased annexin V-positive cells from 43 to 18% ( Fig. 5 A ). Neither adeno-dnJNK nor adeno-dnp38 infection affected the annexin V-positive cell number. Furthermore, ERK inhibition, but not JNK or p38, also inhibited caspase-3 expression and activation ( Fig. 5 B ).
# }+ f5 |, c7 [% a  u  g! n7 }% v1 y$ I5 n
Fig. 4. Effects of inhibition of various MAPKs on cell morphology after cisplatin (CP) treatment. TKPTS cells were treated with 25 µM CP for 24 h. Some cultures were pretreated with 50 µM U-0126 for 1 h or infected with adeno-dn-JNK or adeno-dn-p38 for 24 h before CP treatment. Cell morphology was determined by light microscopy (magnification x 100). Results are representative of 3 independent experiments./ N% x0 r: q/ _+ j+ V/ B
9 S# e0 X) j0 g6 o! |  }2 U/ x
Fig. 5. Effects of inhibition of various MAPKs on apoptosis and caspase-3 activation in TKPTS cells. A : TKPTS cells were treated with 25 µM CP for 24 h. Some cultures were pretreated with 50 µM U-0126 for 1 h or infected with adeno-dn-JNK or adeno-dn-p38 for 24 h before CP treatment. Annexin V/PI binding was determined by flow cytometry. Percentage of apoptotic cells (annexin V positive and PI negative) was calculated. Values were compared with the control value. Significance of differences between the CP-treated (CP) and the CP   U-0126-treated (CP   U-0126) groups was also computed. Results are representative of 3 independent experiments. B : activation of caspase-3 was determined 24 h after CP treatment by Western blotting as described in MATERIALS AND METHODS. Results are representative of 3 independent experiments.. y. q, k) X5 [& l) X% a' d

- |/ z+ y  u' xEffects of inhibition of EGFR or c-src on cisplatin-induced apoptosis as well as caspase-3 and ERK activation in vitro. Because cisplatin treatment is capable of activating the EGFR/ src pathway upstream from ERK ( 4 ), we determined the role of this pathway in survival of TKPTS cells treated with cisplatin. First, we determined that cisplatin treatment activates (phosphorylates) EGFR both in vivo and in vitro ( Fig. 6 ). Second, we determined that application of the EGFR inhibitor AG-1478 (20 µM AG-1478 1 h before cisplatin treatment), c- src inhibitor PP1 (20 µM 1 h before cisplatin treatment), or MEK inhibitor U-0126 (50 µM 1 h before cisplatin treatment) efficiently inhibited endogenous phosphorylation of ERK before cisplatin treatment. In contrast, phosphorylation of JNK or p38 was unchanged under the same conditions (data not shown). Accordingly, inhibition of EGFR activation by AG-1478 significantly improved cell morphology after cisplatin treatment ( Fig. 7 ). FACS analysis of AG-1478-pretreated cells showed reduced numbers of apoptotic cells (subG1 fraction from 45.3 to 24.1%) after cisplatin administration ( Table 1 ). Annexin V binding was also lower in cells that were pretreated with AG-1478 (from 43 to 23%) before 24-h cisplatin treatment ( Fig. 8 A ). Moreover, cisplatin-induced caspase-3 activation was also, albeit partially, inhibited by AG-1478 pretreatment ( Fig. 8 B ). Similarly, pretreatment with the c- src inhibitor PP1 significantly improved cell morphology ( Fig. 7 ), apoptosis ( Table 1 ), and annexin V binding ( Fig. 8 A ) 24 h after cisplatin treatment. PP1 also inhibited cisplatin-induced caspase-3 activation ( Fig. 8 B ). These changes were also accompanied by inhibition of ERK phosphorylation ( Fig. 8 B ), suggesting that ERK is downstream of EGFR and src.
. J: N, f$ t8 Q2 P3 W/ W" w; ?4 U- L* @6 D6 x
Fig. 6. Phosphorylation of the epidermal growth factor receptor (EGFR) after CP administration and phosphorylation of ERK after treatment with various kinase inhibitors. A : phosphorylation of the EGFR was determined by Western blotting in total lysates originated from the kidney of untreated or 1-day CP-treated mice. A representative of 3 independent experiments is shown. B : phosphorylation of the EGFR was determined by Western blotting in total lysates of TKPTS cells 30 min after CP treatment. A representative of 3 independent experiments is shown. C : endogenous phosphorylation of ERK was determined after 1-h treatment with 20 µM AG-1478, 20 µM PP1, or 50 µM U-0126, respectively. The blot shown is a representative of 3 independent experiments. U0, U-0126-MEK inhibitor; AG, AG-1478-EGFR inhibitor; PP1-c-src inhibitor.
  t, x. c) q5 W! a2 A/ Q5 ~. k+ s+ w7 r3 l
Fig. 7. Effects of inhibition of EGFR or c- src on cell morphology after CP treatment. TKPTS cells were treated with 25 µM CP for 24 h. Some cultures were pretreated with 20 µM AG-1478 or PP1 for 1 h before CP treatment. Cell morphology was determined by light microscopy (magnification x 100). Results are representative of 3 independent experiments.
- h% ^: z; M. h8 L/ g1 M6 m7 d
: A. ~; \2 Z# A+ f! ZFig. 8. Effects of inhibition of EGFR or c- src on CP-induced apoptosis. TKPTS cells were treated with 25 µM CP for 24 h. Some cultures were pretreated with 20 µM AG-1478 or PP1 for 1 h before CP treatment. A : annexin V/PI binding was determined by flow cytometry. Percentage of apoptotic cells (annexin V positive and PI negative) was calculated. Results are representative of 3 independent experiments. B : activation of caspase-3 as well as phosphorylation of ERK was determined 24 h after CP treatment by Western blotting as described in MATERIALS AND METHODS. TKPTS cells were treated with 25 µM CP for 1 h. Some cultures were pretreated with 20 µM AG-1478 or PP1 for 1 h before CP treatment. Results are representative of 3 independent experiments." ~+ {! B& R' }& s( c

1 A: D4 p1 @2 }3 |DISCUSSION
5 p0 n, g% W  R; i5 I7 T5 i2 k# ?  |4 U5 P( f
Cisplatin treatment damages the proximal tubules both in vivo ( 21 ) and in vitro ( 23, 27 ). Morphological changes in the proximal tubules of the kidney are observed 3 days after cisplatin administration ( 21 ). Immortalized mouse proximal tubule cells (TKPTS) treated with 25 µM cisplatin for 24 h undergo apoptosis ( 27 ) as demonstrated by the morphological changes ( Fig. 2 A ), FACS analysis ( Fig. 2 B ), annexin V-binding ( Fig. 2 C ), as well as caspase-3 activation ( Fig. 2 D ). These changes were preceded by activation of ERK, JNK, and p38 both in vivo and in vitro ( Figs. 1 and 3 ). We next sought to determine the role of each member of the MAPK cascade in either survival or death of these cells exposed to cisplatin.
( R3 O+ p% Y/ ]4 y5 E9 ?: f3 Y& i8 O' G: v% s) R- P+ @
Using specific inhibitors of those MAPKs in vitro, we found that inhibition of ERK, but not JNK or p38, significantly improved survival of cells 24 h by inhibiting apoptosis after cisplatin treatment ( Figs. 4 and 5, A and B, and Table 1 ). These data suggested that ERK, but not JNK or p38, mediates cisplatin-induced cell death.* ]  Y8 D$ ~3 m  {0 ~/ T
3 `0 _4 u$ D" K5 ~
Because cisplatin activates EGFR in various types of cells ( 4 ) including the kidney ( 18 ), we next sought to determine whether EGFR mediates some of these effects evoked by cisplatin. Indeed, cisplatin treatment enhanced phosphorylation of EGFR both in vivo and in vitro ( Fig. 6 A ). Pretreatment of TKPTS cells with the EGFR inhibitor AG-1478 before cisplatin treatment significantly improved cell survival ( Fig. 7 ) and attenuated apoptosis ( Table 1 and Fig. 8 A ). Also, inhibition of EGFR diminished cisplatin-induced ERK activation ( Fig. 8 B ). These results suggest that cisplatin-induced cell death is EGFR dependent. Interestingly, this receptor activation is ligand independent ( 4 ) but involves activation of c- src and ERK. Such activation of ERK is usually prosurvival ( 4 ) but has been shown recently to participate in cisplatin-mediated ERK activation in the death of HeLa cells ( 35 ).
( x- t: O3 E" d- `
6 u. P6 Z- ]6 j- a  gEGFR is present in the proximal tubules ( 7, 24 ) but not in the thick ascending limb or distal convoluted tubules of the kidney. Also, in the regenerating proximal tubules, EGFR expression is lost in cisplatin-treated animals ( 18 ). Similarly, cells derived from the proximal tubules are significantly more sensitive to cisplatin cytotoxicity than those derived from the distal convoluted tubules in vitro ( 17 ). As the proximal tubules, but not the thick ascending limb or distal convoluted tubules, undergo extensive cell death after cisplatin treatment ( 31 ), these data together suggest a role of EGFR in cisplatin-induced cell death. Small-molecule tyrosine kinase inhibitors, monoclonal antibodies to EGFR or pan-EGFR inhibitors, are safe and well tolerated by patients with cancer ( 33 ). Thus targeting EGFR could be a useful measure in preventing cisplatin-induced renal cytotoxicity in addition to the treatment of cancer.# M4 `# a& o* d! e' ?! [5 c3 k: `
2 k. N! J8 Y  U/ ^
Inhibition of c- src by the inhibitor PP1 before cisplatin treatment also abolished cell death ( Figs. 6 and 7 A and Table 1 ) and inhibited cisplatin-induced ERK activation ( Fig. 8 B ). Thus cisplatin-induced ERK activation, and thus cell death, is EGFR/src dependent. A recent study suggested that c- src functions as a kinase in EGFR phosphorylation after cisplatin treatment ( 4 ), although c- src could be downstream from the EGFR ( 12 ). It will be necessary to perform additional studies to determine whether c- src is downstream or upstream of the EGFR in cisplatin-induced cell death signaling.3 G2 L5 @1 y1 S6 ]

& ^$ X; k; Y! C4 M. d! [  P1 N, j4 hHowever, ERK activation is not sufficient to stimulate the death pathway in TKPTS cells. Our earlier studies showed that the survival of TKPTS cells exposed to moderate levels of H 2 O 2 was ERK dependent ( 2 ). Obviously, cisplatin activates additional signal(s) that render ERK to stimulate the death rather than the prosurvival pathway. Wang et al. ( 35 ) proposed that sustained ERK activation by cisplatin might be responsible for the prodeath function of ERK in HeLa cells. In at least one study, the duration of ERK activity and resultant biological responses were traced to the effects of prolonged ERK activity on the stability and phosphorylation of immediate early gene products such as c- fos ( 22 ). Consistent with this view, cisplatin induced a longer-lasting ERK activation than H 2 O 2 treatment in these cells. Revealing the mechanism of this phenomenon, however, will require additional studies.
* ]" F  e6 }0 s* y* |, C2 H- p$ e% N# T: V6 c( y, S
Our data showed that inhibition of ERK also inhibited caspase-3 activation ( Fig. 8 B ). A link between ERK and caspase-3 activation has been described before ( 14, 28, 30 ), although the mechanism of this link is not fully understood. It is still unknown if ERK activates caspase-3 directly or indirectly. Generally, ERK inhibits rather than activates caspase-3; this inhibition has been associated with NF- B activation ( 32 ), activation of the X-linked inhibitor of apoptosis ( 34 ), or with direct phosphorylation of caspase-9, and subsequent inhibition of caspase-3 activation ( 1 ). Determining whether any of these factors is responsible for the opposing effects of ERK activation in cell survival in those two conditions is also an area of future attention.' n" {0 G& N0 `6 {9 x7 u

0 @4 g5 O# r2 q3 R1 ]In summary, our data suggest that the cisplatin-induced ERK activation is prodeath in nature and that this pathway is activated by EGFR and c- src. Thus manipulation of this pathway might be useful in ameliorating the outcome of cisplatin-induced ARF.# d1 }4 @; s7 n5 Q# n2 U. a
  c& V& r, s3 ^
GRANTS
- x4 }/ d4 H; e3 Q- r3 U; ?* j( D: q8 Y* ~. S
This material is the result of work supported in part with resources and the use of facilities at the Central Arkansas Veterans Healthcare Center (Little Rock, AR) and was supported by National Institutes of Health/National Institute of Diabetes and Digestive and Kidney Diseases Grant PO1-DK-58324-01A1 to R. L. Safirstein.. g8 i3 d+ K( n* w4 d

4 n) ^* ]; P9 s4 d1 mACKNOWLEDGMENTS
. g3 Q0 Q" e3 n
) L1 T) [. \; r% ^The authors acknowledge Dr. E. Bello-Reuss (Dept. of Internal Medicine, UTMB, Galveston, TX) for the TKPTS cells. We also thank Dr. J. Han (Department of Immunology, The Scripps Research Institute, La Jolla, CA) for the dominant-negative p38 adenovirus.& C6 I4 l; v4 J+ K
          【参考文献】
& o' \; ^6 N4 o- S/ L; D  Z  i Allan LA, Morrice N, Brady S, Magee G, Pathak S, and Clarke PR. Inhibition of caspase-9 through phosphorylation at Thr 125 by ERK MAPK. Nat Cell Biol 5: 647-654, 2003.) C3 |  _3 q) V+ }* e; r! v

7 C/ |: `" n8 g/ w/ T* u$ J% U: _/ R( W) ^
3 S0 Q+ F0 S6 m% y8 k
Arany I, Megyesi JK, Kaneto H, Tanaka S, and Safirstein RL. Activation of ERK or inhibition of JNK ameliorates H 2 O 2 cytotoxicity in mouse renal proximal tubule cells. Kidney Int 65: 1231-1239, 2004.3 M" g) T+ D7 J! Z) ]

. T+ ?3 U, Y% n2 W4 X- b$ p" X0 j9 \/ a9 C

) Y7 X$ T: w( z# y- y- e( ]; j. w3 bArany I and Safirstein RL. Cisplatin nephrotoxicity. Semin Nephrol 23: 460-464, 2003.
6 x* J5 O: m9 u/ w* o; |; h& C& n, y+ u8 [5 y) }1 r; k0 f* D
/ ^7 t$ \# ^) o$ ~

0 k) K0 y6 q( W5 p3 lBenhar M, Engelberg D, and Levitzki A. Cisplatin-induced activation of the EGF receptor. Oncogene 21: 8723-8731, 2002.
3 G2 ?% K! }" ~8 z0 q& ~0 L
6 e1 x% V! B7 |) a! I+ U. L9 p/ v4 [3 E

0 Q5 v# j2 B8 nBerns JS and Ford PA. Renal toxicities of antineoplastic drugs and bone marrow transplantation. Semin Nephrol 17: 54-66, 1997.
- J7 C1 W" x8 ~/ @, Y* Z% r/ v' d4 S& d, `$ N$ P

8 ]* R' Q2 u  C
. F" I  d' C. }+ b3 K8 `3 z  {Bonni A, Brunet A, West AE, Datta SR, Takasu MA, and Greenberg ME. Cell survival promoted by the Ras-MAPK signaling pathway by transcription-dependent and -independent mechanisms. Science 286: 1358-1362, 1999.
/ Z  D" N/ E1 r  N! y0 w) i8 O! Q$ s( L4 E, z/ O

4 T1 v0 b: r2 c8 E& n
. M' o& H1 `: }0 j' C7 ?4 ^5 dBreyer MD, Redha R, and Breyer JA. Segmental distribution of epidermal growth factor binding sites in rabbit nephron. Am J Physiol Renal Fluid Electrolyte Physiol 259: F553-F558, 1990.  R! j+ L0 ~6 ^% E. v% _* ^# z5 s8 L
7 m9 [/ n) D( K
, Y& }9 T4 Y8 K

5 ^4 Z+ `3 ^! LCobb MH. MAP kinase pathways. Prog Biophys Mol Biol 71: 479-500, 1999.2 f- T+ s# v( o% V& h% T
$ i, X' O- [1 n- o

! z/ S: L  C* Q+ f& j7 F/ @, j( Z# G9 k8 q2 ^0 u" A
Davis RJ. Signal transduction by the JNK group of MAP kinases. Cell 103: 239-252, 2000.
: f. G- U8 I, ?  q9 O
2 a5 x! ^# r+ m* k! |! o9 m1 L& N
4 J1 @( [( e* N7 S" w  o* A% a3 y2 Z! c+ r2 d9 S$ O3 b1 I
Di Mari JF, Davis R, and Safirstein RL. MAPK activation determines renal epithelial cell survival during oxidative injury. Am J Physiol Renal Physiol 277: F195-F203, 1999.
5 t- c) K# r3 o8 G) t- S6 N9 T! R" U

2 x, A% x& q6 K' }- L# t, `
) q( a! T- _+ G4 f* ^Ernest S and Bello-Reuss E. Expression and function of P-glycoprotein in a mouse kidney cell line. Am J Physiol Cell Physiol 269: C323-C333, 1995.6 n* I- _- Y3 _, j/ l: ~2 p
& s- {2 i' G. ^# P

, z! U) O+ ?; \) ?! ]
6 j2 c, f/ P' ^/ SJorissen RN, Walker F, Pouliot N, Garrett TP, Ward CW, and Burgess AW. Epidermal growth factor receptor: mechanisms of activation and signalling. Exp Cell Res 284: 31-53, 2003.
+ j0 i  ]9 f% U" E5 B+ U8 P. e
8 [3 F2 T% b  C- v$ I7 S' L  g8 t; j
- y) d& J" {- O0 H7 p
Kaneto H, Xu G, Fujii N, Kim S, Bonner-Weir S, and Weir GC. Involvement of c-Jun N-terminal kinase in oxidative stress-mediated suppression of insulin gene expression. J Biol Chem 277: 30010-30018, 2002.
# u) J2 _# D: N" }2 @7 e" u1 {" }
2 `1 R* ?2 r1 w  N" `
# R3 m# P7 i* d: B+ c
$ l  U0 _( a9 m/ L! `Kim GS, Hong JS, Kim SW, Koh JM, An CS, Choi JY, and Cheng SL. Leptin induces apoptosis via ERK/cPLA2/cytochrome c pathway in human bone marrow stromal cells. J Biol Chem 278: 21920-21929, 2003.
% a( X  ~! a( G
3 p5 }3 ~9 w0 n# W
" u3 p; f& O: s' i9 Y
( p9 R5 {0 g  i8 U* mKoo MS, Kwo YG, Park JH, Choi WJ, Billiar TR, and Kim YM. Signaling and function of caspase and c- jun N-terminal kinase in cisplatin-induced apoptosis. Mol Cell 13: 194-201, 2002.: Z, \( s$ E( G2 ^9 @
( d) x0 H( }& q8 n

7 S9 w% c/ B' z# w! E% ]: o  }1 B7 U4 v+ x$ i
Krilleke D, Ucur E, Pulte D, Schulze-Osthoff K, Debatin KM, and Herr I. Inhibition of JNK signaling diminishes early but not late cellular stress-induced apoptosis. Int J Cancer 107: 520-527, 2003., F8 S, l' ]/ y7 }( A' W
! Y8 J4 _7 z& v

( [4 t( _1 z. c4 e- D& y  [$ ^! ?/ k, W% ]1 ]5 b; K7 Y) @- ~  i
Kroning R, Jones JA, Hom DK, Chuang CC, Sanga R, Los G, Howell SB, and Christen RD. Enhancement of drug sensitivity of human malignancies by epidermal growth factor. Br J Cancer 72: 615-619, 1995.
5 S" x0 J7 r) o) c  v' \/ j8 x( S3 ~7 x# q8 R
* @' u( D+ c: _3 o4 E  C$ D2 X

( M9 Z# W  C4 I6 X2 F# qLeonard I, Zanen J, Nonclercq D, Toubeau G, Heuson-Stiennon JA, Beckers JF, Falmagne P, Schaudies RP, and Laurent G. Modification of immunoreactive EGF and EGF receptor after acute tubular necrosis induced by tobramycin or cisplatin. Ren Fail 16: 583-608, 1994.& C( t; }- V$ \5 G
! S# R# W) X7 d! R9 o3 ^% b4 A
0 e* d! \( ~. a; y
4 j! D. ^( s, X5 H# l' U  [9 i# p
Lewis TS, Shapiro PS, and Ahn NG. Signal transduction through MAP kinase cascades. Adv Cancer Res 74: 49-139, 1998.
7 X( k9 {1 G- r. p% U# P4 n/ }3 N0 |- }+ M) T
. v* g% H- {5 N) s' o# g

1 @0 i$ a% Z9 Q6 G! jMandic A, Viktorsson K, Heiden T, Hansson J, and Shoshan MC. The MEK1 inhibitor PD98059 sensitizes C8161 melanoma cells to cisplatin-induced apoptosis. Melanoma Res 11: 11-19, 2001.6 ^- A% s; X9 Y8 b

6 k/ M7 l, G( H0 @. P' G+ b# J9 j3 Q
# a; G3 c" C+ y
* i2 T" w, ~9 H3 U  }Megyesi J, Safirstein RL, and Price PM. Induction of p21WAF1/CIP1/SDI1 in kidney tubule cells affects the course of cisplatin-induced acute renal failure. J Clin Invest 101: 777-782, 1998./ M2 G* R3 V, L* ^/ v" s5 C
; m" Z: L* g) _9 L3 ]4 I6 N) m  W) S

7 Y" e% u$ ?# ~" a
; G* O! c# o, u/ s9 ]Murphy LO, Smith S, Chen RH, Fingar DC, and Blenis J. Molecular interpretation of ERK signal duration by immediate early gene products. Nat Cell Biol 4: 556-564, 2002.. l$ `; T) ?# c( d' [# c

/ s; m. o5 W# b' ^/ _) r% |& t& c5 X+ A

6 |  S- D; C6 R& u9 DNowak G. Protein kinase C- and ERK1/2 mediate mitochondrial dysfunction, decreases in active Na   transport, and cisplatin-induced apoptosis in renal cells. J Biol Chem 277: 43377-43388, 2002.
" P" `; z: }0 h; B+ n# P0 p) l$ ]
4 D5 z! S  c& e, d/ S* f  P5 L' B- e
! T: x7 a+ \/ O4 l4 @# V' \' {& M/ F% n9 v4 Z) _6 q  u/ ^
Okada T, Iwamoto A, Kusakabe K, Mukamoto M, Kiso Y, Morioka H, Sasaki F, and Morikawa Y. Perinatal development of the rat kidney: proliferative activity and epidermal growth factor. Biol Neonate 79: 46-53, 2001.
7 q  L, {/ K! F' {' H: }6 {  l/ y. x/ x) @8 J2 m

, X# {0 j/ s$ n5 S( W6 H+ Z8 v3 c/ A, |; B
Persons DL, Yazlovitskaya EM, Cui W, and Pelling JC. Cisplatin-induced activation of mitogen-activated protein kinases in ovarian carcinoma cells: inhibition of extracellular signal-regulated kinase activity increases sensitivity to cisplatin. Clin Cancer Res 5: 1007-1014, 1999.9 S5 l# P: l% l* _$ v( z
' ^1 l$ N+ t9 u- k/ b% S
, @% D% c. K( R1 `

0 J( r# N; c$ ?2 dPersons DL, Yazlovitskaya EM, and Pelling JC. Effect of extracellular signal-regulated kinase on p53 accumulation in response to cisplatin. J Biol Chem 275: 35778-35785, 2000.
3 X" V; V/ \2 ]' v; J+ M
0 K( U" g, @! V: O/ E
9 z+ t" s; C: M% e* m
: t+ q2 q' W6 \" kPrice PM, Safirstein RL, and Megyesi J. Protection of renal cells from cisplatin toxicity by cell cycle inhibitors. Am J Physiol Renal Physiol 286: F378-F384, 2004.
# U: I* J* U% ^% u2 P. F. B  r; X$ m; j1 `4 R0 A! A! y

5 x  n$ Q1 F: {
3 v1 X" d! @' l7 ORice PL, Goldberg RJ, Ray EC, Driggers LJ, and Ahnen DJ. Inhibition of extracellular signal-regulated kinase 1/2 phosphorylation and induction of apoptosis by sulindac metabolites. Cancer Res 61: 1541-1547, 2001.4 `7 K" f0 a% U8 u! n

5 B0 ^% }/ \" r7 a# A2 X- Q: @7 g. i0 L3 }

9 Y  P7 n. \! @Sanchez-Perez I, Murguia JR, and Perona R. Cisplatin induces a persistent activation of JNK that is related to cell death. Oncogene 16: 533-540, 1998.# R4 B4 j: Q  g% K

" [# ]7 Q: u7 @' q5 P& T" j9 q$ O# A5 Z7 F$ j8 Y; V5 M
  A; y" q4 H2 ^8 F4 ]- ]
Shakibaei M, Schulze-Tanzil G, de Souza P, John T, Rahmanzadeh M, Rahmanzadeh R, and Merker HJ. Inhibition of mitogen-activated protein kinase kinase induces apoptosis of human chondrocytes. J Biol Chem 276: 13289-13294, 2001.
8 j/ b6 C' ?6 k# o; L. Q% \# i) ^- C! l- f' H6 `6 j6 W

* k4 n; @: z: p- N  v
( q! }2 v! L' L! z5 oSheikh-Hamad D, Cacini W, Buckley AR, Isaac J, Truong LD, Tsao CC, and Kishore BK. Cellular and molecular studies on cisplatin-induced apoptotic cell death in rat kidney. Arch Toxicol 78: 147-155, 2003.
8 n6 G8 A4 I) J+ X; u: M
# ^, s" @& o( U+ h4 O$ F
) b% {1 I+ w# U- A* z0 T5 @* n8 ~% e6 s% }) w- E
Shimada K, Nakamura M, Ishida E, Kishi M, Yonehara S, and Konishi N. Contributions of mitogen-activated protein kinase and nuclear factor B to N -(4-hydroxyphenyl)retinamide-induced apoptosis in prostate cancer cells. Mol Carcinog 35: 127-137, 2002.
# q# H. G0 f' n: \
6 Q# Q. y8 S# w9 T2 U
7 T8 z  c+ v9 L! J. P" C1 [$ ~8 q! Y: N' e
Thomas SM and Grandis JR. Pharmacokinetic and pharmacodynamic properties of EGFR inhibitors under clinical investigation. Cancer Treat Rev 30: 255-268, 2004.2 Z3 Y& t' \; F
' b6 g% S; L: e& q5 [" d

! E& l7 w4 h& Y. A) A, ^; h, z+ a9 C- y$ G: H9 Z8 M9 q" u
Varghese J, Khandre NS, and Sarin A. Caspase-3 activation is an early event and initiates apoptotic damage in a human leukemia cell line. Apoptosis 8: 363-370, 2003.
% B* b0 k2 t4 r: M' U, p; p
% C9 |* J4 c6 i8 t4 r
2 u( e' j) s5 g$ W# H' h6 ?) ^! R0 r# ~% e4 S
Wang X, Martindale JL, and Holbrook NJ. Requirement for ERK activation in cisplatin-induced apoptosis. J Biol Chem 275: 39435-39443, 2000.9 Y+ ]9 ]* U+ W5 C& V+ z

# n2 i/ \% z, A) z7 _- y. D, A) W, f5 _  D

9 c  q& \$ W; B6 QWang Y, Su B, Sah VP, Brown JH, Han J, and Chien KR. Cardiac hypertrophy induced by mitogen-activated protein kinase kinase 7, a specific activator for c-Jun NH 2 -terminal kinase in ventricular muscle cells. J Biol Chem 273: 5423-5426, 1998.
+ u) r; I, @$ g! M' @) [# i/ J: X# k9 w" C9 L! {5 u
' B& b3 ?( u) D; b9 A+ B
- g, Q# h9 }9 H$ X3 D
Xia Z, Dickens M, Raingeaud J, Davis RJ, and Greenberg ME. Opposing effects of ERK and JNK-p38 MAP kinases on apoptosis. Science 270: 1326-1331, 1995.( o+ p5 Z- o. {& q% J+ F

7 H/ R% ]& g' d- ~. A' w
+ Q+ _3 l* I) Q
  X5 r) c4 C! Z1 @, s, f2 K. eZanke BW, Lee C, Arab S, and Tannock IF. Death of tumor cells after intracellular acidification is dependent on stress-activated protein kinases (SAPK/JNK) pathway activation and cannot be inhibited by Bcl-2 expression or interleukin 1 -converting enzyme inhibition. Cancer Res 58: 2801-2808, 1998.

Rank: 2

积分
161 
威望
161  
包包
1862  
沙发
发表于 2015-5-23 11:08 |只看该作者
我来看看!谢谢  

Rank: 2

积分
56 
威望
56  
包包
1853  
藤椅
发表于 2015-5-27 21:38 |只看该作者
干细胞行业  

Rank: 2

积分
64 
威望
64  
包包
1769  
板凳
发表于 2015-6-12 13:52 |只看该作者
干细胞之家微信公众号
不错不错,我喜欢看  

Rank: 2

积分
72 
威望
72  
包包
1859  
报纸
发表于 2015-7-13 13:54 |只看该作者
经过你的指点 我还是没找到在哪 ~~~  

Rank: 2

积分
61 
威望
61  
包包
1757  
地板
发表于 2015-7-19 09:43 |只看该作者
加油啊!!!!顶哦!!!!!  

Rank: 2

积分
163 
威望
163  
包包
1852  
7
发表于 2015-7-23 21:51 |只看该作者
羊水干细胞

Rank: 2

积分
73 
威望
73  
包包
1833  
8
发表于 2015-8-19 21:59 |只看该作者
真的有么  

Rank: 2

积分
79 
威望
79  
包包
1769  
9
发表于 2015-8-28 18:00 |只看该作者
每天早上起床都要看一遍“福布斯”富翁排行榜,如果上面没有我的名字,我就去上班……  

Rank: 2

积分
61 
威望
61  
包包
1757  
10
发表于 2015-8-30 11:38 |只看该作者
我毫不犹豫地把楼主的这个帖子收藏了  
‹ 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, 2024-4-25 12:56

Powered by Discuz! X1.5

© 2001-2010 Comsenz Inc.