Top Banner
Altered expression of mitochondrial 16S ribosomal RNA in p53-de¢cient mouse embryos revealed by di¡erential display Monier M. Ibrahim, Marjaneh Razmara, Diep Nguyen, Ronald J. Donahue, Judith A. Wubah, Thomas B. Knudsen * Department of Pathology, Anatomy and Cell Biology, Je¡erson Medical College, 1020 Locust Street, Philadelphia, PA 19107, USA Received 17 February 1998; accepted 21 May 1998 Abstract Inactivation of the tumor suppressor p53 is associated with neural tube defects and altered teratogenicity in early embryos. To gain insight into the function of p53 during early embryogenesis, RNA profiles of wild-type p53(+/+) and p53(3/3) null mutant mouse embryos were compared at the head-fold stage (day 8 post coitum) using HPLC-based mRNA differential display. The results of this screen revealed a deficiency of mitochondrial 16S ribosomal RNA in p53(3/3) embryos. RT-PCR showed abnormalities in 16S rRNA levels relative to some representative nuclear (COIV, L-actin) and mitochondrial (COIII) transcripts in p53(3/3) embryos, and that 16S rRNA expression increased with development of p53(+/+) embryos during neurulation. Embryos that lack p53 also displayed weakened cytochrome c oxidase staining and reduced ATP content. During neurulation, the mouse embryo switches from an anaerobic (glycolytic) to an aerobic (oxidative) metabolism. The preliminary results of the present study suggest that p53 may be involved, directly or indirectly, in this transition. ß 1998 Elsevier Science B.V. All rights reserved. Keywords : Tumor suppressor p53 ; Mitochondrial DNA ; Embryo development 1. Introduction Tumor suppressor p53 is a DNA binding protein that may arrest cell growth or trigger programmed cell death (apoptosis) in response to oncogenic sig- nals or DNA damage [1,2]. Loss of p53 function leads to genomic instability and tumor formation as indicated by the occurrence of mutant p53 pro- teins in a large proportion of human tumors [3] and strong cancer predisposition in mice with a homozy- gous null mutation at the p53 locus [4]. The association between p53 inactivation and tu- mor development has motivated studies to determine how p53 protein activity is controlled in the cell, and to identify critical target genes mediating p53 action (reviewed in [5]). Di¡erential screening revealed changes in several transcripts during p53-dependent apoptosis [6^9]. Altered gene expression is a likely basis of p53 activity in unstimulated cells because p53 can modulate other cellular processes at levels below those leading to apoptosis [10]. Neural cell di¡erentiation, for example, was a low-grade re- sponse to p53 whereas apoptosis occurred at higher levels [11]. This raises questions that pertain to the 0167-4889 / 98 / $19.00 ß 1998 Elsevier Science B.V. All rights reserved. PII:S0167-4889(98)00066-4 * Corresponding author. Fax: +1 (215) 923-3808; E-mail : knudsent@je£in.tju.edu Biochimica et Biophysica Acta 1403 (1998) 254^264
11

Altered expression of mitochondrial 16S ribosomal RNA in p53-deficient mouse embryos revealed by differential display

Jan 28, 2023

Download

Documents

Welcome message from author
This document is posted to help you gain knowledge. Please leave a comment to let me know what you think about it! Share it to your friends and learn new things together.
Transcript
Page 1: Altered expression of mitochondrial 16S ribosomal RNA in p53-deficient mouse embryos revealed by differential display

Altered expression of mitochondrial 16S ribosomal RNA in p53-de¢cientmouse embryos revealed by di¡erential display

Monier M. Ibrahim, Marjaneh Razmara, Diep Nguyen, Ronald J. Donahue,Judith A. Wubah, Thomas B. Knudsen *

Department of Pathology, Anatomy and Cell Biology, Je¡erson Medical College, 1020 Locust Street, Philadelphia, PA 19107, USA

Received 17 February 1998; accepted 21 May 1998

Abstract

Inactivation of the tumor suppressor p53 is associated with neural tube defects and altered teratogenicity in early embryos.To gain insight into the function of p53 during early embryogenesis, RNA profiles of wild-type p53(+/+) and p53(3/3) nullmutant mouse embryos were compared at the head-fold stage (day 8 post coitum) using HPLC-based mRNA differentialdisplay. The results of this screen revealed a deficiency of mitochondrial 16S ribosomal RNA in p53(3/3) embryos. RT-PCRshowed abnormalities in 16S rRNA levels relative to some representative nuclear (COIV, L-actin) and mitochondrial (COIII)transcripts in p53(3/3) embryos, and that 16S rRNA expression increased with development of p53(+/+) embryos duringneurulation. Embryos that lack p53 also displayed weakened cytochrome c oxidase staining and reduced ATP content.During neurulation, the mouse embryo switches from an anaerobic (glycolytic) to an aerobic (oxidative) metabolism. Thepreliminary results of the present study suggest that p53 may be involved, directly or indirectly, in this transition. ß 1998Elsevier Science B.V. All rights reserved.

Keywords: Tumor suppressor p53; Mitochondrial DNA; Embryo development

1. Introduction

Tumor suppressor p53 is a DNA binding proteinthat may arrest cell growth or trigger programmedcell death (apoptosis) in response to oncogenic sig-nals or DNA damage [1,2]. Loss of p53 functionleads to genomic instability and tumor formationas indicated by the occurrence of mutant p53 pro-teins in a large proportion of human tumors [3] and

strong cancer predisposition in mice with a homozy-gous null mutation at the p53 locus [4].

The association between p53 inactivation and tu-mor development has motivated studies to determinehow p53 protein activity is controlled in the cell, andto identify critical target genes mediating p53 action(reviewed in [5]). Di¡erential screening revealedchanges in several transcripts during p53-dependentapoptosis [6^9]. Altered gene expression is a likelybasis of p53 activity in unstimulated cells becausep53 can modulate other cellular processes at levelsbelow those leading to apoptosis [10]. Neural celldi¡erentiation, for example, was a low-grade re-sponse to p53 whereas apoptosis occurred at higherlevels [11]. This raises questions that pertain to the

0167-4889 / 98 / $19.00 ß 1998 Elsevier Science B.V. All rights reserved.PII: S 0 1 6 7 - 4 8 8 9 ( 9 8 ) 0 0 0 6 6 - 4

* Corresponding author. Fax: +1 (215) 923-3808;E-mail : knudsent@je£in.tju.edu

BBAMCR 14341 20-7-98

Biochimica et Biophysica Acta 1403 (1998) 254^264

Page 2: Altered expression of mitochondrial 16S ribosomal RNA in p53-deficient mouse embryos revealed by differential display

cell signaling pathways that have sensitivity to un-stressed p53 function.

The early mouse embryo provides a model tostudy the impact of p53 during developmentallyregulated changes in the steady-state. Most embry-onic tissues, with possible exception of the primitiveheart, strongly express p53 mRNA during neurula-tion and early organogenesis [12,13]. p53 protein,unlike the transcripts, was normally low in these tis-sues unless the embryo was subject to a teratogenicstress [14,15]. Between 8 and 16% of mouse embryoswith a homozygous null mutation at the p53 locusfail to neurulate properly [16,17]. Early manifestationof anterior neural tube defects (NTDs) among some,but not all, p53-de¢cient mouse embryos implies theabsence of p53 as a risk factor in some birth defects.Added support derives from experimental teratogen-esis, in which malformations induced by modelagents vary in incidence and severity depending onp53 genotype. This e¡ect on teratogenicity variedbetween agents. Sometimes the development of mal-formations was suppressed by wild-type p53 function[18] whereas other times the e¡ect of p53 was facil-itative [19]. Eye reduction defects (ERDs), for exam-ple, were induced when neurulation stage mouse em-bryos were exposed to 2-chloro-2P-deoxyadenosine(2-CdA). The incidence and severity of these malfor-mations were determined directly by the embryo'sp53 genotype, being high among p53(+/+) fetuses,low among p53(3/3) fetuses, and intermediateamong heterozygotes [19]. The positive associationbetween p53 function and ERDs was also observedin Xenopus embryos microinjected with p53 tran-scripts [20].

Because DNA binding is the likely basis of p53action [5], the present study was undertaken to iden-tify transcripts whose expression in neurulation stagemouse embryos was sensitive to the loss of p53 func-tion. RNA pro¢les were compared between wild-typep53(+/+) and null-mutant p53(3/3) embryos usingdi¡erential display [21,22]. Neurulation stage em-bryos that lack p53 have altered steady-state levelsof mitochondrial 16S ribosomal RNA transcripts,results that suggest a role for p53 in the developmen-tal transition from an anaerobic (glycolytic) to anaerobic (oxidative) metabolism.

2. Materials and methods

2.1. Animals

TSG-p53 mice [4] were purchased from TaconicFarms, Inc. (Germantown, NY). Outbred CD-1mice were purchased from Charles River BreedingLaboratories (Wilmington, MA). Both strains werekept on a 12 h photoperiod (07.00^19.00 h light).Timed pregnancies were generated by placing amale with nulliparous females at 08.30 h. Detectionof a vaginal plug at 13.30 h signi¢ed coitus (day 0).For di¡erential display, TSG-p53 embryos were har-vested from p53(+/+) damUp53(+/+) sire andp53(3/3) damUp53(3/3) sire crosses on day 8post coitum (pc). Pregnant dams were killed withcarbon dioxide. Embryos were submerged in ice-cold Hanks' balanced saline solution (HBSS) anddissected from the decidua and extraembryonic mem-branes. Six normal phenotypes [23] of each genotypewere selected at the 3^7 somite pair stage. For allother experiments involving TSG-p53 mice, the em-bryos were harvested from p53(+/3) damUp53(+/3)or p53(3/3) sire crosses and genotyped as described[19]. For developmental analysis, RNA was isolatedfrom six-pooled prosencephalic (optic) and cardiacregions of CD-1 (wild-type) embryos on days 8, 9,and 10 pc.

2.2. RNA isolation

Total cellular RNA was extracted by RNeasy(Qiagen, Chatsworth, CA). Tissues were lysed underRNase-free conditions in 0.35 ml lysis bu¡er RLTcontaining 1% (v/v) 2-mercaptoethanol. The sampleswere frozen at 370³C, thawed, and sonicated at 40W for 2 s. An equal volume of 70% ethanol wasadded to the lysate. The suspension was applied tothe micro-spin column, centrifuged for 15 s at8000Ug, and washed with 0.7 ml wash bu¡er RW1and twice with 0.5 ml wash bu¡er RPE. RNA waseluted from the column with 0.05 ml DEPC-treatedwater and centrifugation for 60 s. Residual DNAwas digested with MessageClean (GenHunter, Nash-ville, TN) or DNase I, ampli¢cation grade (Gibco-BRL, Gaithersburg, MD).

BBAMCR 14341 20-7-98

M.M. Ibrahim et al. / Biochimica et Biophysica Acta 1403 (1998) 254^264 255

Page 3: Altered expression of mitochondrial 16S ribosomal RNA in p53-deficient mouse embryos revealed by differential display

2.3. HPLC-based mRNA di¡erential display

RNA (0.2 Wg) was reverse transcribed into threedisplay pools using single-base 3P-anchored oligo(dT)primers (RNAimage, GenHunter). The reaction bu¡-er contained 20 WM dNTPs; and 0.2 WM single 3P-anchored oligo(dT) primers (H-T11M where M = A,G, or C) [22]. Reverse transcription (RT) was per-formed with 100 units of reverse transcriptase fromMoloney murine leukemia virus at 37³C for 50 min.PCR was carried out in 40 Wl reactions containing4 Wl of the RT-mix, 2 WM dNTPs; 1.5 mM MgCl2 ;50 mM KCl; 0.2 WM of the appropriate oligo(dT)primer; 0.2 WM of an arbitrary primer (H-AP1through H-AP8 from GenHunter); and 2 units ofTaq DNA Polymerase (Gibco-BRL) in 10 mMTris-HCl, pH 8.3, cycled for 40 rounds (94³C 30 s,40³C 2 min, and 72³C 30 s) followed by ¢nal exten-sion at 72³C for 5 min in a DNA thermal cycler(Perkin-Elmer, Norwalk, CT). Negative control reac-tions included omission of the reverse transcriptasestep, of either primer, or of Taq Polymerase.

Di¡erential display products were analyzed byhigh-performance liquid chromatography (HPLC).The HPLC column was a Gen-Pak FAX high-per-formance, anion-exchange column (4.6U100 mm)from Waters Associates (Millipore Corporation, Mil-ford, MA) protected with an in-line precolumn ¢lterand used at 50³C [24]. The HPLC system consistedof a model U6K sample injector, two model 501HPLC pumps, a model 680 automated gradient con-

troller and a model 990+ photodiode array detector(Waters Associates). The mobile phase was 25 mMTris-HCl, 1 mM EDTA, and 10% acetonitrile (pH8.0) at a constant £ow of 0.75 ml/min. DNA waseluted from the column by a superimposed gradientof 0.4^0.52 M NaCl (linear curve no. 6) for 0^0.01min and 0.52^0.67 M NaCl (convex curve no. 4) for0.01^25 min. This gradient resolved PCR products of150^600 bp in size. Absorbance was monitored byphotodiode array detection at A252ÿ272. After eachrun, the column was £ushed with 0.5 ml of 0.1 Nphosphoric acid and equilibrated to starting condi-tions for at least 12 min. The column was stored in25 mM Tris, 1 mM EDTA, and 10% acetonitrile (pH8.0) at 4³C and calibrated daily with a ladder ofrestriction fragments generated from digestion ofxX174 RF DNA with HaeIII. Eight of the 11 frag-ments (72, 118, 194, 234, 603, 872, 1078, and 1353bp) were separated with baseline resolution and three(271, 281, 310 bp) were resolved depending on theage of the column; progressive deterioration of col-umn performance occurred after about 200 sampleinjections. Integrated DNA peaks were directly com-pared between samples by the formula (A2+B2)/(2AB), where A and B represent the areas of corre-sponding peaks from p53(+/+) and p53(3/3) em-bryos, respectively. If single pass HPLC screeningproduced peaks di¡ering by at least two-fold, thedisplay reaction was repeated to verify reproducibil-ity. Where a reproducible di¡erence was observed inat least three separate trials with 2 WM dNTPs, the

Fig. 1. HPLC-based mRNA di¡erential display. Native unlabeled PCR products were resolved by HPLC using a GenPak FAX col-umn and convex NaCl gradient, 50³C. The NaCl gradient was optimized for separation of display products in the 150^600 bp range.Absorbance was monitored at 252^272 nm by photodiode array detection (only the region of the chromatogram corresponding to theNaCl gradient is shown). (A) HaeIII xX174 RF DNA ladder. Size of DNA fragments in bp were: 72 bp (peak 1), 118 bp (peak 2),194 bp (peak 3), 234 bp (peak 4), 271+281+310 bp (peak 5), 603 bp (peak 6), 872 bp (peak 7), 1078 bp (peak 8) and 1353 bp (peak9). Partial resolution of the three bands in peak 5 was lost as the HPLC column aged. (B,C) HPLC pro¢les displayed from p53(+/+)and p53(3/3) embryos, respectively in B and C, at the 3^7 somite pair stage. Primer pairs were H-AP7 and H-T11A, and dNTP con-centration was 2 WM. The arrow marks a 217 bp peak (W7A.614) which was markedly under-represented in the p53-null embryo. (D)Negative control reaction omitting the reverse-transcription step; the baseline £uctuation is caused by the convex NaCl gradient.(E,F) HPLC pro¢les displayed from p53(+/+) and p53(3/3) embryos, respectively in E and F, with primer set H-AP1 and H-T11A.Identical patterns were evident in the H-T11A display pools; dNTP concentration was 2 WM. (G,H) HPLC pro¢les displayed fromp53(+/+) and p53(3/3) embryos, respectively in G and H, with primer set H-AP7 and H-T11G. Identical patterns were evident in theH-T11G display pool ampli¢ed with H-AP7; dNTP concentration was 2 WM. (I,J) HPLC pro¢les of p53(+/+) embryos (I) and p53(3/3) embryos (J) ampli¢ed with primer pairs H-AP7 and H-T11A; dNTP concentration was 200 WM. (K) Preparative samples of p53(+/+) embryos used to purify W7A.614 for cloning and sequencing; the puri¢ed DNA was reampli¢ed with primers H-AP7 and H-T11Aand 20 WM dNTPs and re-chromatographed. Some contamination was evident in the foreshoulder of peak W7A.614. (L) Electropho-resis of puri¢ed W7A.614 (same as in K) on 1.5% agarose gel with ethidium bromide staining (lane 1); xX174 RF DNA cut withHaeIII (lane 2). The contaminating DNA was evident as a faint, smaller band.

C

BBAMCR 14341 20-7-98

M.M. Ibrahim et al. / Biochimica et Biophysica Acta 1403 (1998) 254^264256

Page 4: Altered expression of mitochondrial 16S ribosomal RNA in p53-deficient mouse embryos revealed by differential display

PCR reaction was repeated with higher (20 and 200WM) dNTP concentrations [25].

To isolate a di¡erential display product, the PCRreaction was scaled up by a factor of eight (320 Wltotal) and applied to the HPLC column. A 0.6 mlfraction containing the di¡erential peak was col-lected, reduced to 0.4 ml in a Speed-Vac concentrator(Savant Instruments, Farmingdale, NY), and dilutedto 1 ml with TTE bu¡er (0.1 M Tris-HCl, 10 mMtriethylamine, and 1 mM EDTA, pH 7.7). This sam-

ple was loaded onto a Nensorb 20 nucleic acid puri-¢cation cartridge (NEN Life Science, Boston, MA)primed with methanol and TTE bu¡er. After wash-ing the cartridge with 3 ml TTE bu¡er and water,DNA was eluted with 0.5 ml of 50% methanol inwater. The DNA was dried, resuspended in 15 Wlsterile water, and reampli¢ed with the appropriatePCR primers and 20 WM dNTPs. Reampli¢edDNA was cloned into pCR-TRAP and screenedwith Rgh and Lgh primers (GenHunter). Plasmids

BBAMCR 14341 20-7-98

M.M. Ibrahim et al. / Biochimica et Biophysica Acta 1403 (1998) 254^264 257

Page 5: Altered expression of mitochondrial 16S ribosomal RNA in p53-deficient mouse embryos revealed by differential display

were puri¢ed on Qiagen columns and extended withLseq and Rseq sequencing primers (Aidseq Kit C,GenHunter Corporation) in combination withPRISM Ready Reaction DyeDeoxy TerminatorCycle Sequencing Kit (Applied Biosystems, Foster,CA). Extension products were puri¢ed using QuickSpin columns (Boehringer-Mannheim, Indianapolis,IN) and sequenced with an ABI Model 373A DNAsequencer (Applied Biosystems) [26].

2.4. Semi-quantitative RT-PCR

Embryonic RNA (1^2 Wg) was annealed to ran-dom primers (Gibco-BRL) at 70³C and reverse-tran-scribed with SuperScript II RNase H3 reverse tran-scriptase (Gibco-BRL) at 42³C in the presence ofdithiothreitol and 200 WM of each dNTP. Negativecontrols omitted reverse transcriptase. Gene-speci¢coligo(d)nucleotide primers were designed from se-quence information in GenBank using OLIGO Pri-mer Analysis Software 5.0 (National Biosciences,Plymouth, MN). Each PCR cycle consisted of denat-uration at 94³C 1 min, annealing at 57³C 1 min, andextension at 72³C 1 min (10 min in last cycle). PCRwas initially performed with di¡erent cycle numbersto ¢nd the optimal number and sample dilution forquantitative ampli¢cation of target and controlgenes; subsequently, PCR was performed at 24cycles, which was optimal for a linear responseacross sample dilutions of 1:50, 1:100, and 1:500.Normalization to L-actin provided a control forsemi-quantitative measure of transcript abundance[27]. PCR products were resolved on a nondenatur-ing 8% polyacrylamide gel, electrophoresed, andstained with ethidium bromide. The gel was photo-graphed with Polaroid 665 reversal ¢lm, and neg-atives were scanned with an LKB Ultroscan XL

laser densitometer (Pharmacia Biotech, Piscataway,NJ).

2.5. Cytochrome c oxidase staining

Embryos were ¢xed 1 h at 4³C in 4% paraformal-dehyde and 2% glutaraldehyde in phosphate-bu¡eredsaline (PBS), rinsed in PBS for 3 h, in¢ltrated with10% sucrose in PBS at 4³C, and incubated in PBS-sucrose containing 0.05% diaminobenzidine (DAB)and 0.02% cytochrome c at 37³C [28]. Negative con-trols omitted cytochrome c or DAB.

2.6. Nucleotide analysis

Individual embryos were extracted in 0.1 ml 60%aqueous methanol at 320³C overnight [29]. The sam-ples were centrifuged for 5 min at 12 000Ug. Thepellet was used for protein determination (Bio-Rad,Hercules, CA); the supernatant was dried for 2 h in aSpeed-Vac and resuspended in 0.2 ml of 50 mM am-monium phosphate, pH 6.5, containing 2 mM tetra-butylammonium hydroxide and 5% acetonitrile (mo-bile phase). Samples were chromatographed on a C18

reversed-phase HPLC column at a £ow rate of 1.5ml/min. Peaks corresponding to ATP and ADP wereintegrated at A262.

3. Results

3.1. Di¡erential display peak analysis

RNA pro¢les of p53(+/+) and p53(3/3) embryoswere analyzed by HPLC-based di¡erential display atthe 3^7 somite pair stage of development. TheHPLC-based method a¡orded simple and reproduci-

Table 1Primers used for expression PCR analysis of murine respiratory subunits

Locusa Genome Upper primer Lower primer PCR product

16S rRNA mtDNA 5P-ACAGCTAGAAACCCCGAAAC-3P 5P-AAGATAAGAGACAGTTGGAC-3P 785 bpCOIII mtDNA 5P-AATCCAAGTCCATGACCATT-3P 5P-TGTGTTGGTACGAGGCTAGA-3P 296 bpND4L mtDNA 5P-ATGCCATCTACCTTCTTCAA-3P 5P-AAACTAAGGTGATGGGGATT-3P 193 bpCOIV nuDNA 5P-GCACCAATGAATGGAAGACA-3P 5P-CAGCGGGCTCTCACTTCTTC-3P 234 bpL-actin nuDNA 5P-TACCACAGGCATTGTGATGG-3P 5P-AATAGTGATGACCTGGCCGT-3P 310 bpaCOIII, cytochrome c oxidase, subunit III; ND4L, NADH:ubiquinone oxidoreductase, subunit 4L; COIV, cytochrome c oxidase, sub-unit IV.

BBAMCR 14341 20-7-98

M.M. Ibrahim et al. / Biochimica et Biophysica Acta 1403 (1998) 254^264258

Page 6: Altered expression of mitochondrial 16S ribosomal RNA in p53-deficient mouse embryos revealed by differential display

ble patterns of arbitrary PCR products in the 150^600 bp range (Fig. 1A). Di¡erences between samplescould be spotted quickly and were probably kept to aminimum by low peak complexity (5^10 peaks perprimer set). Since arbitrary PCR primers hybridize as8-mers [21,22], theoretical ampli¢cation of cDNAfragments in the 150^600 bp range should sampleone of every 146 cDNAs (48 divided by 450). Forthe typical mammalian cell expressing 15 000 di¡er-ent mRNA species [21] the eight arbitrary primerscreen should have ampli¢ed 822 di¡erential displaypeaks. A total of 167 (20.3% of the predicted num-ber) was observed. These probably represent only asmall subset of the transcripts expressed in an earlyembryo.

Comparison of the two genotypes indicated equalrepresentation for 165 (98.8%) of the di¡erential dis-play products. One di¡erence (W7A.614) was under-represented in p53(3/3) embryos (Fig. 1B,C) andthe other (N6G.610) was over-represented (notshown). Di¡erential display products were dependentupon reverse transcription (Fig. 1D), speci¢c for theparticular arbitrary primer (Fig. 1E,F), and uniqueto the corresponding display pool (Fig. 1G,H). Ofthe two, only W7A.614 remained di¡erent when thesamples were ampli¢ed with 20^200 WM dNTPs (Fig.1I,J). Neither was a¡ected by the p53-null conditionin neural display pools generated from the brain oreye of adult mice (not shown).

Both di¡erential display peaks were isolated (Fig.

Fig. 3. Developmental expression of 16S rRNA. RNA was iso-lated from the prosencephalon (pn) and heart (ht) of CD-1 em-bryos harvested at day 8 (4^6 somite pair stage), day 9 (16^18somite pair stage) and day 10 (28^30 somite pair stage). Expres-sion PCR was performed for 24 cycles using primers speci¢cfor 16S rRNA (top band) and L-actin (bottom band); reactionmixtures for test and control reactions were double-loaded ontothe gels. Photographic negative of 8% polyacrylamide gelstained with ethidium bromide; lane m: HaeIII digest ofxX174 RF DNA marker; two actin bands (320+310 bp) wereconsistently observed in heart samples only.

Table 2Relative expression of mitochondrial 16S rRNA in early embryosa

Genotype 16S rRNA 16S rRNA/ND4L 16S rRNA/COIII 16S rRNA/COIV 16S rRNA/L-actin 16S rRNA/mRNAsb

p53(+/3) 1.67 þ 0.25 7.61 þ 4.42 1.58 þ 0.17 8.25 þ 0.27 2.41 þ 0.54 0.75 þ 0.15p53(3/3) 0.34 þ 0.22 6.68 þ 3.90 0.46 þ 0.25 4.07 þ 1.25 0.55 þ 0.41 0.28 þ 0.20ratio (% reduction) 0.19 þ 0.10 (81%) 0.99 þ 0.11 (1%) 0.31 þ 0.19 (69%) 0.27 þ 0.07 (73%) 0.28 þ 0.25 (72%) 0.46 þ 0.18 (54%)aSignals for 16S rRNA relative to representative nuclear and mitochondrial mRNAs (mean þ S.E.M., n = 2) assayed at the early head-fold presomite stage.b16S rRNA relative to the four representative mRNAs (P = 0.037 by paired t-test).

Fig. 2. RT-PCR assay of respiratory transcripts. EmbryonicRNA samples were reverse-transcribed from p53(+/3) andp53(3/3) siblings at the early head-fold presomite stage on day8 pc. The gene-speci¢c primers are listed in Table 1. Ethidium-stained gels were photographed with Polaroid reversal ¢lm(negative shown). Lane m: HaeIII digest of xX174 RF DNAmarker; lanes 1^10: PCR products ampli¢ed from p53(+/3)embryos (lanes 1, 2, 3, 7 and 8) and p53(3/3) embryos (lanes4, 5, 6, 9 and 10) using primers speci¢c for ampli¢cation ofmurine L-actin (lanes 1 and 4), ND4L (lanes 2 and 5), COIII(lanes 3 and 6), COIV (lanes 7 and 9) and 16S rRNA (lanes 8and 10).

BBAMCR 14341 20-7-98

M.M. Ibrahim et al. / Biochimica et Biophysica Acta 1403 (1998) 254^264 259

Page 7: Altered expression of mitochondrial 16S ribosomal RNA in p53-deficient mouse embryos revealed by differential display

1K,L) and sequenced. N6G.610 corresponded to ananonymous cDNA (GenBank accession numberT03459) cloned from human infant-fetal brain [30].It was not pursued further. The W7A.614 sequencecorresponded to nucleotide positions (np) 2077^2275of the mouse mitochondrial genome (GenBank ac-cession number J01420). Nucleotide positions 2077^2275 of the mouse mitochondrial genome map to the16S ribosomal RNA gene (16S rRNA, np 1094^2675) [31]. Mature mitochondrial 16S rRNA tran-scripts are heterogeneous at the 3P-terminus butmost frequently terminate in `T' at np 2675, imme-diately preceding the tRNALeu�UUR� gene, in mice[32]. Annealing of 16S rRNA to the single-base 3P-anchored H-T11A primer used to generate W7A.614was consistent with this termination; however,W7A.614 mapped to an internal 5P-TA6 sequenceabout 400 nucleotides upstream from the predicted16S rRNA/tRNALeu�UUR� gene boundary. Hetero-geneity at the 3P-end was probably not responsible

for the di¡erences between p53(+/+) and p53(3/3)embryos. A more likely explanation was that p53(+/+) and p53(3/3) embryos di¡ered from one anotherwith respect to the abundance of 16S rRNA.

3.2. Semi-quantitative analysis of mitochondrialtranscripts

To con¢rm di¡erential display analysis, RT-PCRwas used to amplify 16S rRNA in relation to severalrepresentative mitochondrial and nuclear transcripts.This approach was selected over direct hybridizationmethods because of limitations in tissue volume.Gene-speci¢c PCR primers were designed to amplifythe 5P-end of 16S rRNA (np 1338^2103 of the mito-chondrial DNA genome); cytochrome c oxidase sub-unit III (COIII), a mitochondrial-encoded subunit ofcomplex IV of the respiratory chain; NADH:oxido-reductase subunit 4L (ND4L), a mitochondrial-en-coded subunit of complex I of the respiratory chain;

Table 3ATP and ADP levels of day 8 embryosa

Genotypeb n mg protein perembryo

ATP content(nmol/mg protein)c

ADP content(nmol/mg protein)

ATP/ADP ratioc

p53(+/+) 10 0.041 þ 0.012 8.55 þ 2.12 2.11 þ 0.64 4.18 þ 0.79p53(+/3) 13 0.044 þ 0.014 8.93 þ 2.66 2.44 þ 0.56 3.76 þ 1.14p53(3/3) 7 0.034 þ 0.014 6.56 þ 1.13a**,b** 2.55 þ 0.87 2.83 þ 0.98a**,b*

normal 5 0.036 þ 0.015 6.58 þ 1.35 2.41 þ 0.81 2.98 þ 1.07abnormal 2 0.028 þ 0.010 6.50 þ 0.52 2.88 þ 1.27 2.45 þ 0.90

aAverage developmental age was 8 somite pairs (range 4^12 somite pairs) ; data are given as mean þ S.D.bDetermined by PCR genotyping of yolk sac; p53(3/3) were also grouped as normal or abnormal based on their external morphol-ogy.cSigni¢cant di¡erences by unpaired t-test: (a) versus p53(+/+); and (b) versus p53(+/3) ; *P9 0.05; **P9 0.015.

Fig. 4. Cytochrome c oxidase staining of day 8 embryos. Embryos (7^8 somite pair stage) ¢xed and reacted with cytochrome c andDAB as whole mounts; genotype was determined by PCR analysis of yolk sac DNA. Light microscopy of: (A) p53(+/3) embryo rep-resenting the `dark' phenotype; (B) p53(3/3) littermate representing the `light' phenotype; and (C) negative control embryo wherebycytochrome c was omitted from the reaction medium. 35U magni¢cation (pn, prosencephalon; ht, heart).

BBAMCR 14341 20-7-98

M.M. Ibrahim et al. / Biochimica et Biophysica Acta 1403 (1998) 254^264260

Page 8: Altered expression of mitochondrial 16S ribosomal RNA in p53-deficient mouse embryos revealed by differential display

cytochrome c oxidase subunit IV (COIV), a nuclear-encoded subunit of complex IV of the respiratorychain; and L-actin, an internal control for inputRNA (Table 1). PCR signal, y, was linear for inputcDNA across sample dilutions of 1:50 (y = 0.63),1:100 (y = 0.30), and 1:500 (y = 0.12) (coe¤cient ofdetermination, R2 = 0.991).

RT-PCR ampli¢ed the predicted fragments fromembryonic RNA samples. Signals generated fromp53(3/3) embryos were generally weaker than werethose from p53(+/3) embryos relative to the L-actincontrol (Fig. 2). Signals for 16S rRNA and COIIIexceeded those for mitochondrial mRNAs andCOIV, respectively. The quantitative excess of ribo-somal over messenger RNAs, and of mitochondrialover nuclear mRNAs for respiratory complexes, wereseen in other developing tissues analyzed by directhybridization methods [33^35].

Comparison of p53(+/3) and p53(3/3) embryosat the early head-fold presomite stage con¢rmed thede¢ciency of 16S rRNA templates between siblingpairs. This was true whether PCR signals were nor-malized to L-actin or several representative mRNAs(Table 2). Overall, the reduction was 54% (P = 0.037,paired t-test). RT-PCR also revealed that steady-state levels of 16S rRNA increase as p53(+/+) em-bryos develop (Fig. 3). The p53(+/+) embryos wereobtained on day 8 (4^6 somite pairs), day 9 (16^18somite pairs), and day 10 (28^30 somite pairs) ofgestation. Analysis of the prosencephalon and heartindicated that 16S rRNA increased relative to L-actinin both developing structures. Just as these structuresdi¡er in their relative sensitivity to p53-dependentevents [15,19], so they may di¡er in developmentalexpression of 16S rRNA.

3.3. Bioenergy status of p53-de¢cient embryos

Cytochemical staining for cytochrome c oxidaserevealed `dark' and `light' phenotypes among em-bryos harvested from two crosses between p53(+/3)damUp53(3/3) sire (Fig. 4). All ¢ve p53(3/3) em-bryos displayed the light-staining phenotype, whereaseight of nine p53(+/3) embryos were dark-staining.Segregation of light-staining phenotype with p53(3/3) genotype was signi¢cant (P = 0.001, M2 analysis).

Another indicator of mitochondrial bioenergeticfunction is the ATP/ADP ratio. Total ATP and

ADP levels were measured in embryos of di¡erentp53 genotypes (Table 3). Embryos lacking p53 dis-played lower ATP per Wg protein or per pmol ADP.The ATP shortfall approached 33%. Two p53(3/3)embryos displayed an unusually wide gap betweencranial neural folds. These early manifestations ofanterior NTDs presented with a similar ATP short-fall as the phenotypically normal p53(3/3) subset(Table 3).

4. Discussion

About 80 subunits compose the ¢ve protein com-plexes of the mitochondrial respiratory chain. Thir-teen subunits are encoded in the mitochondrial DNA(mtDNA) genome along with small (12S) and large(16S) mitochondrial rRNAs and 22 transfer RNAsrequired for their translation [36]. The present studydetected under-expression of a mitochondrial en-coded gene, 16S rRNA, among neurulation stageembryos with a homozygous null mutation at thep53 locus. Partial (54%) loss of 16S rRNA expres-sion, together with weakened staining for cyto-chrome c oxidase activity and shortfall of ATP ap-proaching 33%, suggests a developmental connectionbetween tumor suppressor p53 and mitochondrialenergy transduction.

4.1. Mitochondrial translation

Mitochondrial 12S and 16S rRNA transcripts aredisproportionately expressed over mRNAs in ac-tively respiring cells [37,38]. The present study hasnot addressed the question of whether mitochondrialrRNA abundance in general is a¡ected by p53 or ifthe e¡ect is speci¢c for 16S rRNA. The alterations insteady-state levels of 16S rRNA could represent amore global deregulation, one that involves biogen-esis of mtDNA or the respiratory chain. Since thesmall subset (1%) of embryonic transcripts sampledby di¡erential display would probably have been toosmall to reveal a global e¡ect on respiratory chainmRNAs, additional studies will be needed to deter-mine if the e¡ect on 16S rRNA was general or spe-ci¢c.

Defects in mitochondrial translation can be man-ifested as cellular pathogenesis in neurulation stage

BBAMCR 14341 20-7-98

M.M. Ibrahim et al. / Biochimica et Biophysica Acta 1403 (1998) 254^264 261

Page 9: Altered expression of mitochondrial 16S ribosomal RNA in p53-deficient mouse embryos revealed by differential display

embryos. The supportive evidence comes from tera-togenicity studies with chloramphenicol, a speci¢cinhibitor of mitochondrial translation [39,40], andthe severe birth defects that can be associated withthe pathogenic A3243G MELAS mutation [41,42].Expansion of 16S rRNA transcript pools in the de-veloping mouse embryo suggests that mitochondrialprotein synthesis takes on a growing importance dur-ing neurulation stages. This is consistent with Xeno-pus, where large-scale ampli¢cation of the mitochon-drial ribosomal RNA transcript begins late ingastrulation and continues into neurulation [43].Failure of mouse embryos that lack p53 to achievenormal 16S rRNA levels might re£ect a function ofwild-type p53 as a signal to increase mitochondrialtranslational during neurulation, a scenario consis-tent with the known inhibitory e¡ects of chloram-phenicol on developing embryos.

4.2. Oxidative metabolism

For some time it has been known that mammalianembryos are initially adapted for anaerobic (glyco-lytic) bioenergy production and then switch to aero-bic (oxidative) metabolism [44,45]. Around the 8 so-mite stage, the rate at which lactate is produced bythe embryo drops from the previous steady state of3^7 nmol/mg protein/h, which is similar to a rapidlygrowing tumor; consequently, the embryo's demandfor oxygen grows [46^49]. Mitochondrial ultrastruc-ture becomes more characteristic of actively respiringcells as the embryo develops between the 8 and 30somite stages [44]. If expansion of 16S rRNA poolsre£ects this increase in mitochondrial respiratory ca-pacity, then control of this oxidative transition mayat least partly depend on p53 protein activity. It isinteresting to note that p53 protein activity is in-duced by hypoxia [50,51] or imbalances in ribonu-cleotide pools [52]. Perhaps p53 reacts to metabolicdemand to stimulate energy transduction in thegrowing embryo through cell signaling pathwaysthat control mitochondrial biogenesis or expressionof functional respiratory complexes.

Cranial neural folds are sensitive to alterations inboth p53 protein activity and bioenergetic metabo-lism. Dependence on p53 may be inferred from theassociation of anterior NTDs with a subset of p53-de¢cient embryos [17]. Closure of the anterior neuro-

pore normally takes place on days 8^9 pc [23].Hence, NTDs would be expected from loss of ap53-dependent bioenergetic conservation mechanismduring this period. Shortfall in ATP levels contributeto experimentally induced cranial NTDs [49], andan `energy gap' has been proposed to explain thehigh frequency of NTDs among diabetic mothers[53]. The dependence of bioenergetic conservationon p53 might also explain the di¡erential teratogen-icity of neurulation stage embryos to some terato-gens. For example, 2-CdA has recently been shownto alter patterns of energy metabolism [54]. The ca-pacity of 2-CdA to induce ERDs dependent on p53could be a re£ection of an early action on mitochon-dria.

Recent studies have demonstrated a connectionbetween p53 and oxidative metabolism in the controlof apoptosis. A cell's commitment to apoptosis maybe measured by the collapse of mitochondrial elec-trochemical potential, v8m [55]. Associated mito-chondrial changes include 16S rRNA degradation[56], suspension of mitoribosomal translation [57],and transcriptional shut-down of mtDNA [58]. Be-cause b0 cells that lack mtDNA still undergo apop-tosis, the changes in 16S rRNA and probably othermtDNA encoded genes are not primary for theapoptotic functions of mitochondria [59]. On theother hand, at least part of the control of p53-de-pendent apoptosis may be exerted at the level ofoxidative metabolism. Transactivation of cellular ox-idoreductases, and the subsequent generation of re-active oxygen intermediates, was causally linked top53-dependent apoptosis [8,60]. Perhaps p53-depend-ent alterations in 16S rRNA expression observed inthe present study re£ect a low-grade (subapoptotic)in£uence of p53 protein on oxidative metabolism atthe steady state, an in£uence which becomes over-shadowed during high-grade (apoptotic) p53 proteinactivity.

4.3. Conclusion

During neurulation the mammalian embryoswitches from primarily anaerobic (glycolytic) toaerobic (oxidative) metabolism. This is re£ected inup-regulation of mitochondrial 16S rRNA tran-scripts, a product of the mtDNA genome, and maydepend at least partly on p53 function. The novel

BBAMCR 14341 20-7-98

M.M. Ibrahim et al. / Biochimica et Biophysica Acta 1403 (1998) 254^264262

Page 10: Altered expression of mitochondrial 16S ribosomal RNA in p53-deficient mouse embryos revealed by differential display

connection between tumor suppressor p53 functionand activities encoded by the mtDNA genome hasbroader implications toward understanding the roleof p53-dependent events in oxidative metabolism andrelated mechanisms in apoptosis.

Acknowledgements

This research was supported by Grant RO1HD30302 from the National Institute of ChildHealth and Human Development (T.B.K.). M.M.I.,J.A.W. and R.J.D. were fellows on Training GrantT32 ES07282 from the National Institute of Environ-mental Health Sciences. Methods development wasassisted by Dr. William Warren of Waters Chroma-tography Division, and Dr. James Eberwine of theUniversity of Pennsylvania School of Medicine. Fortechnical assistance we are indebted to Je¡ Charlap.Helpful comments were o¡ered by Jan Hoek of Jef-ferson Medical College, Peter Liang of the CancerResearch Institute, Vanderbilt University, andCharles Bieberich of the University of Maryland inBaltimore.

References

[1] M.B. Kastan, O. Onyekwere, D. Sidransky, B. Vogelstein,R.W. Craig, Cancer Res. 51 (1991) 6304^6311.

[2] E. Yonish-Rouach, D. Resnitzky, J. Lotem, L. Sachs, A.Kimchi, M. Oren, Nature 352 (1993) 345^347.

[3] M. Hollstein, D. Sidransky, B. Vogelstein, C.C. Harris, Sci-ence 253 (1991) 49^53.

[4] L.A. Donehower, M. Harvey, B.L. Slagle, M.J. McArthur,C.A. Montgomery Jr., J.S. Butel, A. Bradley, Nature 356(1992) 215^224.

[5] L.J. Ko, C. Prives, Genes Dev. 10 (1996) 1054^1072.[6] S.M. Lehar, M. Nacht, T. Jacks, C.A. Vater, T. Chittenden,

B.C. Guild, Oncogene 12 (1996) 1181^1187.[7] D. Israeli, E. Tessler, Y. Haupt, A. Elkeles, S. Wilder, R.

Amson, A. Telerman, M. Oren, EMBO J. 16 (1997) 4384^4392.

[8] K. Polyak, Y. Xia, J.L. Zweler, K.W. Kinzler, B. Vogelstein,Nature 389 (1997) 300^305.

[9] G.S. Wu, T.F. Burns, E.R. McDonald III, W. Jiang, R.Meng, I.D. Krantz, G. Kao, D.-D. Gan, J.-Y. Zhou, R.Muschel, S.R. Hamilton, N.B. Spinner, S. Markowitz, G.Wu, W.S. El-Diery, Nature Genet. 17 (1997) 141^143.

[10] X. Chen, L.J. Ko, C. Prives, Genes Dev. 10 (1996) 2438^2451.

[11] O. Eizenberg, A. Faber-Elman, E. Gottlieb, M. Oren, V.Rotter, M. Schwartz, Mol. Cell. Biol. 16 (1996) 5178^5185.

[12] M. Rogel, M. Popliker, C.G. Webb, M. Oren, Mol. Cell.Biol. 5 (1985) 2851^2855.

[13] P. Schmid, A. Lorenz, H. Hameister, M. Montenarh, Devel-opment 113 (1991) 857^865.

[14] D.E. MacCallum, T.R. Hupp, C.A. Midgley, D. Stuart, S.J.Campell, A. Harper, F.S. Walsh, E.G. Wright, A. Balmain,D.P. Lane, P.A. Hall, Oncogene 13 (1996) 2575^2587.

[15] E. Gottlieb, R. Ha¡ner, A. King, G. Asher, P. Gruss, P.Lonai, M. Oren, EMBO J. 16 (1997) 1381^1390.

[16] J.F. Armstrong, M.A. Kaufman, D.J. Harrison, A.R.Clarke, Curr. Biol. 5 (1995) 931^936.

[17] V.P. Sah, L.D. Attardi, G.J. Mulligan, B.O. Williams, R.T.Bronson, T. Jacks, Nature Genet. 10 (1995) 175^180.

[18] C.J. Nicol, M.L. Harrison, R.R. Laposa, I.L. Gimelshtein,P.G. Wells, Nature Genet. 10 (1995) 181^187.

[19] J.A. Wubah, M.M. Ibrahim, X. Gao, D. Nguyen, M.M.Pisano, T.B. Knudsen, Curr. Biol. 6 (1996) 60^69.

[20] M. Hoever, J.H. Clement, D. Wedlich, M. Montenarh, W.Knochel, Oncogene 9 (1994) 109^120.

[21] P. Liang, A.B. Pardee, Science 257 (1992) 967^971.[22] P. Liang, W. Zhu, X. Zhang, Z. Guo, R.P. O'Connell, L.

Averboukh, F. Wang, A.B. Pardee, Nucleic Acids Res. 22(1994) 5763^5764.

[23] M.H. Kaufman, The Atlas of Mouse Development, Aca-demic Press, San Diego, CA, 1992.

[24] W. Warren, J. Doniger, BioChromatography 10 (1991) 216^219.

[25] K. Miyashiro, M. Dichter, J. Eberwine, Proc. Natl. Acad.Sci. USA 91 (1994) 10800^10804.

[26] M.M. Ibrahim, I.T. Weber, T.B. Knudsen, Biochem. Bio-phys. Res. Commun. 209 (1995) 407^416.

[27] W.G. Cance, R.J. Craven, T.M. Weiner, E.T. Liu, Surg.Oncol. 1 (1992) 309^314.

[28] S. Liu, M. Wong-Riley, J. Neurosci. 14 (1994) 5338^5351.[29] X. Gao, M.R. Blackburn, T.B. Knudsen, Teratology 49

(1994) 1^12.[30] A.S. Khan, A.S. Wilcox, M.H. Polymeropoulos, J.A. Hop-

kins, T.J. Stevens, M. Robinson, A.K. Orpana, J.M. Sikela,Nature Genet. 2 (1992) 180^185.

[31] M.J. Bibb, R.A. Van Etten, C.T. Wright, M.W. Walberg,D.A. Clayton, Cell 26 (1981) 167^180.

[32] R.A. Van Etten, J.W. Bird, D.A. Clayton, J. Biol. Chem.258 (1983) 10104^10110.

[33] L. Piko, K.D. Taylor, Dev. Biol. 123 (1987) 364^374.[34] M. Renis, P. Cantatore, P. Loguercia Polosa, F. Fracasso,

M.N. Gadaleta, J. Neurochem. 52 (1989) 750^754.[35] K.D. Taylor, L. Piko, Mol. Reprod. Dev. 40 (1995) 29^35.[36] G. Attardi, G. Schatz, Annu. Rev. Cell Biol. 4 (1988) 289^

333.[37] B. Kruse, N. Narasimhan, G. Attardi, Cell 58 (1989) 391^

397.[38] J.R. Valverde, R. Marco, R. Garesse, Proc. Natl. Acad. Sci.

USA 91 (1994) 5368^5371.[39] L. Piko, D.G. Chase, J. Cell Biol. 58 (1973) 357^378.

BBAMCR 14341 20-7-98

M.M. Ibrahim et al. / Biochimica et Biophysica Acta 1403 (1998) 254^264 263

Page 11: Altered expression of mitochondrial 16S ribosomal RNA in p53-deficient mouse embryos revealed by differential display

[40] D. Oerter, R. Bass, Naunyn-Schmiedeberg Arch. Pharmacol.290 (1975) 175^189.

[41] M.S. Damian, P. Seibel, W. Scachenmayr, H. Reichmann,W. Dorndorf, Am. J. Med. Genet. 62 (1996) 398^403.

[42] A. Feigenbaum, D. Chitayat, B. Robinson, D. MacGregor,T. Myint, G. Arbus, J.M. Nowaczyk, Am. J. Med. Genet. 62(1996) 404^409.

[43] J.W. Chase, I.B. Dawid, Dev. Biol. 27 (1972) 504^518.[44] B. Mackler, R. Grace, H.M. Duncan, Arch. Biochem. Bio-

phys. 144 (1971) 603^610.[45] T. Tanimura, T.H. Shepard, Proc. Soc. Exp. Biol. Med. 135

(1970) 51^54.[46] G.M. Morriss, D.A.T. New, J. Embryol. Exp. Morphol. 54

(1979) 17^35.[47] J. Clough, D.G. Whittingham, J. Embryol. Exp. Morphol.

74 (1983) 133^142.[48] A. Miki, E. Fujimoto, T. Ohsaki, H. Mizoguti, Anat. Em-

bryol. 178 (1988) 337^343.[49] E.S. Hunter III, J.A. Tugman, Teratology 52 (1995) 317^

323.[50] T.G. Graeber, M. Peterson, M. Tsai, K. Monica, A.J.

Fornace Jr., A.J. Giaccia, Mol. Cell. Biol. 14 (1994) 6264^6277.

[51] T.G. Graeber, C. Osmanian, T. Jacks, D.E. Housman, C.J.Koch, S.W. Lowe, A.J. Giaccia, Nature 379 (1996) 88^91.

[52] S.P. Linke, K.C. Clarkin, A. Di Leonardo, A. Tsou, G.M.Wahl, Genes Dev. 10 (1996) 934^947.

[53] B.E. Finley, S. Norton, Am. J. Obstet. Gynecol. 165 (1991)1661^1666.

[54] P. Hentosh, M. Tibudan, Mol. Pharmacol. 51 (1997) 613^619.

[55] M. Castedo, A. Macho, N. Zamzami, T. Hirsch, P. Marche-tti, J. Uriel, G. Kroemer, Eur. J. Immunol. 25 (1995) 3277^3284.

[56] D.R. Crawford, R.J. Lauzon, Y. Wang, J.E. Mazurkiewicz,G.P. Schools, K.J.A. Davies, Free Radical Biol. Med. 22(1997) 1295^1300.

[57] J.-L. Vayssiere, P.X. Petit, Y. Risler, B. Mignotte, Proc.Natl. Acad. Sci. USA 91 (1994) 11752^11756.

[58] B.A. Osborne, S.W. Smith, Z.G. Liu, K.A. McLaughlin, L.Grimm, L.M. Schwartz, Immunol. Rev. 142 (1994) 301^320.

[59] P. Marchetti, S.A. Susin, D. Decaudin, S. Gamen, M. Cas-tedo, T. Hirsch, N. Zamzami, J. Naval, A. Senik, G.Kroemer, Cancer Res. 56 (1996) 2033^2038.

[60] T.M. Johnson, Z.-X. Yu, V.J. Ferrans, R.A. Lowenstein, T.Finkel, Proc. Natl. Acad. Sci. USA 93 (1996) 11848^11852.

BBAMCR 14341 20-7-98

M.M. Ibrahim et al. / Biochimica et Biophysica Acta 1403 (1998) 254^264264