文档库 最新最全的文档下载
当前位置:文档库 › 持续性细胞皱缩在人上皮细胞凋亡中的必要性

持续性细胞皱缩在人上皮细胞凋亡中的必要性

512

Acta Physiologica Sinica , August 25, 2007, 59 (4): 512-516

https://www.wendangku.net/doc/438092464.html,

Review

Received 2007-04-19 Accepted 2007-07-03

This work was supported by Grants-in Aid for Scientific Research from MEXT and JSPS. *Corresponding author. Tel: +81-564-557731; Fax: +81-564-557735; E-mail: okada@nips.ac.jp

Prerequisite role of persistent cell shrinkage in apoptosis of human epithelial cells

SHIMIZU Takahiro, MAENO Emi, OKADA Yasunobu *

Department of Cell Physiology, National Institute for Physiological Sciences, Okazaki 444-8585, Japan

Abstract: Persistent cell volume reduction is a major hallmark of apoptosis. Recent studies have demonstrated that cell volume reduction is not a passive, secondary event of the apoptotic cell death process. Whole-cell shrinkage, termed apoptotic volume decrease (A VD), takes place soon after stimulation with apoptogen and precedes caspase activation, DNA and cell fragmentation in a variety of cell types including human epithelial cells. The A VD induction is the result of KCl efflux attained by activation of K + and Cl - channels.Inhibition of A VD induction leads to rescue of the cells from apoptosis. Since the A VD process is coupled to dysfunction of the regulatory volume increase (RVI), apoptotic cells undergo persistent cell shrinkage in human epithelial HeLa cells. When the RVI mechanism was impaired, hypertonic stress itself induced not only persistent cell shrinkage but also apoptotic cell death in HeLa cells.Even under normotonic apoptogen-free conditions, exposure of HeLa cells to Na +- or Cl --deficient solution alone can bring about persistent cell shrinkage and thereafter apoptotic cell death. Thus, it is concluded that persistent cell shrinkage, which comprises A VD induction and RVI dysfunction, is a prerequisite to apoptosis induction in human epithelial cells.

Key words: apoptosis; apoptotic volume decrease; persistent cell shrinkage; cell volume regulation; human epithelial cells

持续性细胞皱缩在人上皮细胞凋亡过程中的必要性

SHIMIZU Takahiro ,MAENO Emi ,OKADA Yasunobu *

国立生理学研究所细胞生理学部,冈崎 444-8585,日本

摘 要:持续性细胞皱缩是凋亡发生的一个主要标志。近期研究发现细胞皱缩在细胞凋亡过程中并不是一个被动的次要事件。在各种细胞中,包括人上皮细胞,凋亡因子(apoptogen)刺激后马上发生全细胞皱缩,又称为凋亡性容积减小(apoptotic volume decrease, AVD),继而发生caspase 激活、DNA 片段化、细胞破裂死亡。K +和Cl -通道的激活导致KCl 外流,诱导AVD 发生。抑制AVD 发生可以抑制细胞凋亡。AVD 与调节性容积增加(regulatory volume increase, RVI)异常相伴发生时,人上皮性HeLa 细胞发生持续性细胞皱缩。RVI 功能受损时,高渗本身就能诱导HeLa 细胞持续性细胞皱缩,继而凋亡。即使在正常渗透压、无凋亡因子刺激的情况下,将HeLa 细胞置于缺乏Na + 或Cl - 的溶液也会导致细胞持续性皱缩,继而凋亡。因此,AVD 诱导和RVI 异常所导致的持续性细胞皱缩是人上皮细胞发生凋亡的首要条件。关键词:凋亡;凋亡性容积减小;持续性细胞皱缩;细胞容积调节;人上皮细胞中图分类号:Q 255;Q 256

1 Introduction

Apoptosis, also called programmed cell death, is essential to maintaining somatic cell turnover and tissue homeostasis.

This type of cell death is morphologically and biochemi-cally characterized by cell shrinkage, caspase activation,cytochrome c release, nuclear condensation, DNA laddering, and cell fragmentation into apoptotic bodies. In

513

SHIMIZU Takahiro et al : Persistent Cell Shrinkage in Apoptosis

the last ten years, scientific interest in apoptotic cell shrin-kage has upsurged [1-3], not only because it is a common phenomenon in apoptotic cell death [4] but also because block-ing of cell shrinkage was found to rescue cells from apoptosis [2,5]. Previously, it was believed that apoptotic cell shrinkage is observed only after biochemical changes such as caspase activation, proteolytic events and DNA cleavage [6].However, it is now noted that apoptotic cell shrinkage must be divided into two phases: the early-phase whole-cell vo-lume decrease, termed apoptotic volume decrease (A VD)[2,5]and the late-phase cell fragmentation into apoptotic bodies.Our recent studies demonstrated that the early-phase A VD is independent of caspase activation whereas the late-phase cell fragmentation is dependent on caspase activation [2,7].A VD takes place within 0.5-1 h after stimulation either with a mitochondrion-mediated apoptosis inducer (stauros-porine) or with a death receptor-mediated apoptogen (TNF α,Fas ligand)[2,5,7]. The A VD event precedes caspase activation,cytochrome c release, DNA fragmentation and apoptotic body formation [2,7]. These facts indicate that A VD is one of the requisite or causative processes of apoptosis, but not a secondary feature or the result of executive apoptotic events.

2 A VD induction and RVI dysfunction

It is well known that animal cell volume is elaborately con-trolled and the cell volume regulation is underlying a variety of cell functions [8]. Osmotically swollen cells regulate their volume mainly by inducing KCl efflux which drives out-flow of osmotically obliged water, whereas shrunken cells restore their volume mainly by inducing NaCl uptake and water inflow. These mechanisms are called regulatory volume decrease (RVD) and regulatory volume increase (R VI), respectively, and are accompanied by activation of

a number of ion channels and transporters existing on the plasma membrane [9,10].

Hazama and Okada, through collaboration with Ishizaki,accidentally found that the RVD process is facilitated in human epithelial HeLa and lymphoid U937 cells undergoing A VD [2]. Thus, it was hypothesized that A VD shares a simi-lar induction mechanism with R VD which is known to be accomplished by parallel operation of Ca 2+-activated K +channels [11,12] and swelling-activated, volume-sensitive out-wardly rectifying (VSOR) Cl - channels [11,13]. In fact, Shimizu and Okada recently demonstrated that even though HeLa cells are not undergoing swelling but rather shrinking, the VSOR Cl - channel is activated by staurosporine, TNF α or Fas ligand [14]. So far, apoptotic stimuli have been shown to activate a variety of K + channels [15] including voltage-gated K + channels [16,17], Ca 2+-activated K + channels [17-19], two-pore domain K + channels [20], ROMK [21], HERG [22] and Kv2.1[23].It is now finally established that the A VD induction is brought about by activation of K + and Cl - channels which results in KCl efflux and exit of osmotically obliged water (Fig.1).

When the A VD induction was prevented by blocking K +or Cl - channels, apoptotic biochemical events and cell death were found to be abolished in human epithelial HeLa, lym-phoid U937, rat pheochromocytoma PC12, and mouse neuroblastoma×rat glioma hybrid NG108-15 cells stimu-lated with staurosporine or TNF α[2] as well as mouse cardiomyocytes stimulated with staurosporine [24,25]. Also,we showed that preventing the A VD induction by anion channel blockers inhibited apoptosis in rat cardiomyocytes stimulated with staurosporine [26], mouse cardiomyocytes subjected to ischemia-reperfusion [27], human adenocarci-noma KB cells challenged with an anti-cancer drug cisplatin [28],and mouse hippocampal CA1 neurons subjected to tran-sient forebrain ischemia [29]. Thus, it appears that the A VD

Fig. 1. Schematic illustration of roles of persistent cell shrinkage in apoptosis of cells stimulated with a mitochondrion-mediated apoptogen (STS) or a death receptor-mediated apoptogen (FasL or TNF α). Activation of volume-regulatory K + channels and Cl - channels (VSOR) is involved in the A

VD induction, and inhibition of volume-regulatory cation channels (HICC) and transporters (NHE and AE) may be involved

in the RVI dysfunction in apoptotic cells (see text for details).

Acta Physiologica Sinica, August 25, 2007, 59 (4): 512-516 514

event is an essential process of apoptosis in a large variety

of non-excitable and excitable cells, including epithelial cells.

In apoptotic cells, persistent whole-cell shrinkage takes

place. However, most types of normal cells possess the

ability to regulate their volume even after osmotic shrinkage,

a process called R VI which involves not only the operation

of Na+/H+ exchanger (NHE) and anion exchanger (AE)

but also activation of hypertonicity-induced cation channel

(HICC) in HeLa cells[30]. In apoptotic cells, thus, the RVI

process must be overridden by the A VD process or the

RVI mechanism must be impaired[1]. Our recent study[31]

clearly demonstrated that the latter is the case in HeLa cells

stimulated with a Fas ligand, TNFα or staurosporine.

Therefore, it appears that persistent cell shrinkage com-

prises A VD induction and R VD dysfunction (Fig.1).

3 Persistent cell shrinkage as a prerequisite to

apoptosis

It has been reported that hypertonicity-induced cell shrink-

age often induces apoptotic cell death. Bortner and

Cidlowski[1], for the first time, showed that hypertonicity-

induced cell shrinkage leads to apoptotic death in lymphoid

cells that lack the R VI ability but not in several other R VI-

exhibiting cell types including HeLa cells. We have con-

firmed their findings, as presented in Fig.2. When human

lymphoid U937 cells that lack the R VI ability (Fig.2A, filled

triangles) were exposed to hypertonic solution prepared

by adding either 300 mmol/L mannitol or 150 mmol/L NaCl,

activation of caspase-3 was induced (Fig.2B, filled

symbols). However, hypertonic stimulation failed to in-duce capase-3 activation in human epithelial HeLa cells (Fig. 2B, open symbols) that can exhibit the R VI response (Fig. 2A, open triangles).

In addition, we have recently shown that hypertonic stress induced apoptosis even in HeLa cells when the R VI was inhibited by combined application of blockers for NHE and AE[31] or by application of HICC blocker[32]. Furthermore, we found that hypertonicity-induced apoptosis in NHE1-deficient PS120 fibroblasts which lack the RVI response, but hypertonicity-induced apoptosis was completely pre-vented when R VI was restored by transfection with NHE1[31]. Thus, it is evident that hypertonicity-induced cell shrin-kage causes apoptosis, if persisted due to dysfunction of RVI. However, there remains a possibility that hypertonic stress may have activated some intracellular signals that are essentially related to apoptosis induction.

To test whether persistent shrinkage per se is a causative factor for apoptosis induction under normotonic conditions,we have examined effects of deprivation of extracellular Cl-, because Na+-K+-2Cl- cotransporters (NKCC) and K+-Cl- cotransporter (KCC) are known to be involved in cell volume maintenance in many cell types[33,34]. When an elec-trochemical driving force for facilitation of KCC-mediated Cl- efflux and for inhibition of NKCC-mediated Cl- influx was imposed by reducing the extracellular Cl- concentra-tion (Fig.3), normotonic cell shrinkage was actually ob-served in HeLa cells[35]. Under low Cl- conditions, cell shrinkage should persist because of inhibition of RVI due to inability to accomplish volume-regulatory Cl- influx via AE and background Cl- channels (Fig.3). Also reduction of cell viability coupled to caspase-3 activation was found to be induced after exposure to low Cl- solution for over 2 h[35]. Next, we examined effects of deprivation of extracellular Na+, because this maneuver is supposed to induce not only inhibition of forward-mode operation of NKCC and Na+/ Ca2+ exchanger (NCX) but also induction of reverse-mode operation of NKCC and NCX (Fig.3). Also, Na+ deprivation Fig. 2. Hypertonicity-induced apoptosis in RVI-lacking U937 cells but not in RVI-exhibiting HeLa cells. Each symbol stands for the means±SEM (vertical bar) of 5 observations. A: Changes in mean cell volume upon hypertonic stress (300 → 600 mosM) produced by adding 300 mmol/L mannitol in HeLa cells (open triangles) and U937 cells (filled triangles). B: Changes in caspase-3 activity upon hypertonic stress (300 →600 mosM) produced by adding 300 mmol/L mannitol

(triangles) or 150 mmol/L NaCl (circles) in HeLa cells (open) and

U937 cells (filled symbols).

515

SHIMIZU Takahiro et al : Persistent Cell Shrinkage in Apoptosis

may inhibit the R VI mechanism by inhibiting Na + influx via HICC [30] and via NHE (Fig.3). In fact, extracellular Na +deprivation brought about persistent shrinkage and apoptosis in HeLa cells [36]. When Na + deprivation-induced cell shrin-kage was prevented by a blocker of NKCC or NCX, HeLa cells were found to be rescued from apoptosis under Na +-deficient conditions [36].

4 Concluding remark

Considering all the data provided, it is concluded that per-sistent cell shrinkage per se provides a sufficient condi-tions for apoptosis induction in human epithelial cells. Fur-ther studies are required to elucidate how apoptosis indu-cers inhibit RVI and how persistent cell shrinkage per se triggers apoptosis cell death.

* * *

ACKNOWLEDGEMENTS: The authors thank K.Shigemoto, C. Kondo, M. Yamasawa and M. Ohara for technical assistance, and T. Okayasu for secretarial assistance.

REFERENCES

1

Bortner CD, Cidlowski JA. Absence of volume regulatory mecha-nisms contributes to the rapid activation of apoptosis in thymocytes. Am J Physiol Cell Physiol 1996; 271: C950-C961.2

Maeno E, Ishizaki Y, Kanaseki T, Hazama A, Okada Y.Normotonic cell shrinkage because of disordered volume regula-tion is an early prerequisite to apoptosis. Proc Natl Acad Sci USA 2000; 97: 9487-9492.

3Friis MB, Friborg CR, Schneider L, Nielsen MB, Lambert IH,

Christensen ST, Hoffmann EK. Cell shrinkage as a signal to apoptosis in NIH 3T3 fibroblasts. J Physiol 2005; 567: 427-443.

4Wyllie AH, Kerr JFR, Currie AR. Cell death: the significance of apoptosis. Int Rev Cytol 1980; 68: 251-306.

5

Okada Y, Maeno E, Shimizu T, Dezaki K, Wang J, Morishima S.Receptor-mediated control of regulatory volume decrease (RVD)and apoptotic volume decrease (AVD). J Physiol 2001; 532: 3-16.6

Saraste A, Pulkki K. Morphologic and biochemical hallmarks of apoptosis. Cardiovasc Res 2000; 45: 528-537.

7

Okada Y, Maeno E. Apoptosis, cell volume regulation and vo-lume-regulatory chloride channels. Comp Biochem Physiol Part A 130: 377-383.

8

Lang F, Busch GL, Ritter M, Volkl H, Waldegger S, Gulbins E,Haussinger D. Functional significance of cell volume regulatory mechanisms. Physiol Rev 1998; 78: 247-306.

9

Okada Y. Ion channels and transporters involved in cell volume regulation and sensor mechanisms. Cell Biochem Biophys 2004;41: 233-258.

10

Wehner F, Olsen H, Tinel H, Kinne-Saffran E, Kinne RK. Cell volume regulation: osmolytes, osmolyte transport, and signal transduction. Rev Physiol Biochem Pharmacol, 2003; 148: 1-80.11

Hazama A, Okada Y. Ca 2+ sensitivity of volume-regulatory K +and Cl - channels in cultured human epithelial cells. J Physiol (London) 1988; 402: 687-702

12

Wang J, Morishima S, Okada Y. I K channels are involved in the regulatory volume decrease in human epithelial cells. Am J Physiol Cell Physiol 2003; 284: C77-C84.

13

Okada Y. Volume expansion-sensing outward-rectifier Cl -channel: fresh start to the molecular identity and volume sensor.Am J Physiol Cell Physiol 1997; 273: C755-C789.

14

Shimizu T, Numata T, Okada Y. A role of reactive oxygen spe-cies in apoptotic activation of volume-sensitive Cl - channel. Proc Natl Acad Sci USA 2004; 101: 6770-6773.

15

Wang Z. Roles of K + channels in regulating tumour cell prolifera-

Fig. 3. Schematic illustration of the mechanisms of persistent cell shrinkage causing apoptosis under normotonic Na +-free and low Cl -conditions. The process of persistent cell shrinkage comprises two phases, A VD induction and RVI dysfunction, both involving function/dysfunction of ion transporters and channels (see text for details). NKCC, Na +-K +-2Cl - cotransporter; NCX, Na +/Ca 2+ exchanger; KCC, K +-Cl -cotransporter; HICC, hypertonicity-induced cation channel; NHE, Na +/H +

exchanger; AE, anion exchanger.

Acta Physiologica Sinica, August 25, 2007, 59 (4): 512-516 516

tion and apoptosis. Pflugers Arch 2004; 448: 274-286.

16Yu SP, Yeh CH, Sensi SL, Gwag BJ, Canzoniero LM, Farhangrazi ZS, Ying HS, Tian M, Dugan LL, Choi DW. Mediation of neu-ronal apoptosis by enhancement of outward potassium current.

Science 1997; 278: 114-117.

17Nietsch HH, Roe MW, Fiekers JF, Moore AL, Lidofsky SD.

Activation of potassium and chloride channels by tumor necro-sis factor alpha. Role in liver cell death. J Biol Chem 2000; 275: 20556-20561.

18Krick S, Platoshyn O, Sweeney M, Kim H, Yuan JX. Activation of K+ channels induces apoptosis in vascular smooth muscle cells.

Am J Physiol Cell Physiol 2001; 280: C970-C979.

19Elliott JI, Higgins CF. IKCa1 activity is required for cell shrinkage, phosphatidylserine translocation and death in T lymphocyte apoptosis. EMBO Rep 2003; 4: 189-194.

20Trimarchi JR, Liu L, Smith PJ, Keefe DL. Apoptosis recruits two-pore domain potassium channels used for homeostatic vo-lume regulation. Am J Physiol Cell Physiol 2002; 282: C588-C594.

21Nadeau H, McKinney S, Anderson DJ, Lester HA. ROMK1 (Kir1.1) causes apoptosis and chronic silencing of hippocampal neurons. J Neurophysiol 2000; 84: 1062-1075.

22Wang H, Zhang Y, Cao L, Han H, Wang J, Yang B, Nattel S, Wang Z. HERG K+ channel, a regulator of tumor cell apoptosis and proliferation. Cancer Res 2002; 62: 4843-4848.

23Redman PT, He K, Hartnett KA, Fefferson BS, Hu L, Rosenberg PA, Levitan ES, Aizenman E. Apoptotic surge of potassium currents is mediated by p38 phosphorylation of Kv2.1. Proc Natl Acad Sci USA 2007; 104: 3568-3573.

24Takahashi N, Wang XM, Tanabe S, Uramoto H, Jishage K, Uchida S, Sasaki S, Okada Y. ClC-3-independent sensitivity of apoptosis to Cl- channel blockers in mouse cardiomyocytes. Cell Physiol Biochem 2005; 15: 263-270.

25Tanabe S, Wang XM, Takahashi N, Uramoto H, Okada Y. HCO

3

--independent rescue from apoptosis by stilbene derivatives in rat cardiomyocytes. FEBS Lett 2005; 579: 517-522.26Okada Y, Shimizu T, Maeno E, Tanabe S, Wang X, Takahashi N.

Volume-sensitive chloride channels involved in apoptotic vo-lume decrease and cell death. J Membr Biol 2006; 209: 21-29. 27Wang X, Takahashi N, Uramoto H, Okada Y. Chloride channel inhibition prevents ROS-dependent apoptosis induced by is-chemia-reperfusion in mouse cardiomyocytes. Cell Physiol Biochem 2005; 16: 147-154.

28Ise T, Shimizu T, Lee EL, Inoue H, Kohno K, Okada Y. Roles of volume-sensitive Cl- channel in cisplatin-induced apoptosis in human epidermoid cancer cells. J Membr Biol 2005; 205: 139-145.

29Inoue H, Ohtaki H, Nakamachi T, Shioda S, Okada Y. Anion channel blockers attenuate delayed neuronal cell death induced by transient forebrain ischemia. J Neurosci Res 2007; 85 (7): 1427-1435.

30Wehner W, Shimizu T, Sabirov R, Okada Y. Hypertonic activa-tion of a non-selective cation conductance in HeLa cells and its contribution to cell volume regulation. FEBS Lett 2003; 551: 20-

24.

31Maeno E, Takahashi N, Okada Y. Dysfunction of regulatory volume increase is a key component of apoptosis. FEBS Lett 2006; 580: 6513-6517.

32Shimizu T, Wehner F, Okada Y. Inhibition of hypertonicity-induced cation channels sensitizes HeLa cells to shrinkage-induced apoptosis. Cell Physiol Biochem 2006; 18: 295-302. 33Russell JM. Sodium-potassium-chloride cotransport. Physiol Rev 2000; 80: 211-276.

34Lytle C, McManus T. Coordinate modulation of Na-K-2Cl cotransport and K-Cl cotransport by cell volume and chloride.

Am J Physiol Cell Physiol 2002; 283: C1422-C1431.

35Maeno E, Shimizu T, Okada Y. Normotonic cell shrinkage in-duces apoptosis under extracellular low Cl- conditions in human lymphoid and epithelial cells. Acta Physiol 2006; 187: 217-222. 36Nukui M, Shimizu T, Okada Y. Normotonic cell shrinkage in-duced by Na+ deprivation results in apoptotic cell death in human epithelial HeLa cells. J Physiol Sci 2006; 56: 335-339.

* * * * *

Yasunobu Okada, M.D., Ph.D.

Yasunobu Okada is the Director-General, National Institute for Physiological Sciences (NIPS); the Vice President, Na-tional Institutes of Natural Sciences (NINS); and the Professor, School of Life Science, Graduate University for Ad-vanced Studies (SOKENDAI); the President of The Physiological Society of Japan (PSJ); the President of The Federa-tion of Asian and Oceanian Physiological Societies (FAOPS); as well as the Vice President and the Secretary General of the Organizing Committee of The 36th International Congress of Physiological Sciences (IUPS2009). Born in 1943, graduated at Kyoto University Faculty of Medicine in 1970, obtained a medical doctor license in 1970, and studied Physiology at Department Physiology, Kyoto University Faculty of Medicine as Research Fellow (~1974), Research Associate (~1981) and Assistant Professor (~1992). Awarded a scholarship from Japan Government to study Biophysics at Montreal University (1976-1977) and received a Ph.D. in 1981 at Kyoto University Faculty of Medicine. Moved in 1992 to National Institute for Physiological Sciences in Okazaki as Professor, and took the present position in April, 2007.

相关文档