Corrections Proc. Natl. Acad. Sci. USA 96 (1999) 6571 › content › pnas › 96 › 6 ›...

9
Immunology. In the article “Absence of tumor necrosis factor rescues RelA-deficient mice from embryonic lethality” by Taka- hiro S. Doi, Michael W. Marino, Toshitada Takahashi, Toshimichi Yoshida, Teruyo Sakakura, Lloyd J. Old, and Yuichi Obata, which appeared in number 6, March 16, 1999, of Proc. Natl. Acad. Sci. USA (96, 2994–2999), the following corrections should be noted: In Table 1, page 2995, line 1 of the title, the second sentence, “Results of matings of TNF1y1 or TNF2y2 mice with relA1y2 mice,” should be removed. In Table 2, page 2997, line 2, right column, the genotype of the male parent which currently reads “TNF1y2relA1y1should read “TNF2y2relA1y2.” In Table 2, page 2997, line 3, right-most column, the genotype of the offspring which currently reads 21y 2relA1y1” should read “TNF1y2relA1y1.” In line 2 of Discussion, page 2997, the sentence that reads “TNF mediates this abnormality and the associated lethality, as viable RelA-deficient progeny without liver damage result from matings of TNF-deficient RelA-heterozygous mice with RelA-deficient mice” should read “TNF mediates this abnor- mality and the associated lethality, as viable RelA-deficient progeny without liver damage result from matings of TNF- deficient RelA-heterozygous mice.” Medical Sciences. In the article “RB-mediated suppression of spontaneous multiple neuroendocrine neoplasia and lung me- tastases in Rb 1y2 mice” by Alexander Yu. Nikitin, Marı ´a I. Jua ´rez-Pe ´rez, Song Li, Leaf Huang, and Wen-Hwa Lee, which appeared in number 7, March 30, 1999, of Proc. Natl. Acad. Sci. USA (96, 3916–3921), the following correction should be noted. The labeling of the gel in Fig. 1O was erroneously moved down and to the right. The corrected figure and its legend are shown below. Immunology. In the article “Ubiquitin-dependent degradation of IkBa is mediated by a ubiquitin ligase Skp1yCul1yF-box protein FWD1” by Shigetsugu Hatakeyama, Masatoshi Kita- gawa, Keiko Nakayama, Michiko Shirane, Masaki Matsumoto, Kimihiko Hattori, Hideaki Higashi, Hiroyasu Nakano, Ko Okumura, Kazunori Onoe ´, Robert A. Good, and Kei-ichi Nakayama, which appeared in number 7, March 30, 1999, of Proc. Natl. Acad. Sci. USA (96, 3859–3863), due to printer’s error, the following changes should be noted. On page 3859 in the abstract, text, and footnote, the word “Cull 1” should be “Cul1.” Biophysics. In the article “High-resolution NMR of encapsu- lated proteins dissolved in low-viscosity fluids” by A. Joshua Wand, Mark R. Ehrhardt, and Peter F. Flynn, which appeared in number 26, December 22, 1998, of Proc. Natl. Acad. Sci. USA (95, 15299–15302), the term “high pressure NMR” was incorrectly printed as “HPLC NMR,” due to a printer’s error. FIG. 1. Multiple neuroendocrine neoplasia in Rb 1/2 mice. (A and B) Gross compound pituitary tumor on P394. The tumor consists of two histologically distinct components that substitute for the pituitary anterior (AL) and intermediate (IL) lobes, which are demarcated by remnants of Rathke’s cleft (arrows). AL tumor cells contain a-GSU (A) and are positioned loosely around sinusoid-like vessels. IL tumor cells contain a-melanocyte-stimulating hormone (B) and form poorly vascularized epithelioid fields with central necrotic and hemorrhagic areas. (C and D) EAP in the anterior pituitary lobe on P90 before (C) and after (D) microdissection for genotype analysis by the PCR. (E) C-cell carcinoma on P340 showing the typical arrangement of poly- hedral tumor cells in solid nests and rough hyalinized collagen with calcification (arrow). (F and G) C-cell EAP (arrow) on P60 before (F) and after (G) microdissection. The atypical cells show a parafollicular location. (H) Medullary thyroid carcinoma invading surrounding tissues on P469. Accumulation of calcitonin (brown color) is evident in the cytoplasm of tumor cells (arrow). CR, cartilage. (I) Calcitonin- containing metastatic cells in the lung on P463. The metastatic cells exhibit intraalveolar spreading (arrows). (J) A well-vascularized para- thyroid tumor (PG) and a neighboring solid C-cell tumor of the thyroid gland (TG) on P370. (K) Parathyroid hormone expression in para- thyroid tumor cells. (L) Pheochromocytoma of the adrenal medulla (AM) compressing the adrenal cortex (AC). (M and N) EAP in the adrenal medulla (AM) on P60 before (M) and after (N) microdissec- tion. The arrow indicates multiple apoptotic figures. Staining: avidin- biotin-peroxidase immunostaining for a-GSU (A), a-melanocyte- stimulating hormone (B), calcitonin (H and I), or parathyroid hor- mone (K) with hematoxylin counterstaining; (CG, J, and LN) hematoxylin-eosin staining. [Bar: 160 mm(A and B), 40 mm(C, D, and I), 110 mm(E), 60 mm(F and G), 50 mm(H), 150 mm(J), and 20 mm (K), 390 mm(L), and 70 mm(M and N).] (O) Absence of the wild-type Rb allele (151-bp PCR product) in gross tumors (T; lanes 3, 4, 7, 10, 11, 17, and 18) and EAPs (E; lanes 1, 2, 6, 8, 9, 14, and 16) of the pituitary anterior lobe (AL, lanes 1–4), the parathyroid gland (PG, lanes 6 and 7), thyroid C cells (TG, lanes 8–11), lung metastases (L, lanes 12 and 13), or adrenal medulla (lanes 14 and 16–18). N, Rb 1/2 normal tissue (lanes 5 and 15). Nondenaturing 12% polyacrylamide gel stained with silver. The 236-bp band corresponds to the mutant Rb allele (11). Corrections Proc. Natl. Acad. Sci. USA 96 (1999) 6571 Downloaded by guest on July 29, 2020 Downloaded by guest on July 29, 2020 Downloaded by guest on July 29, 2020 Downloaded by guest on July 29, 2020 Downloaded by guest on July 29, 2020 Downloaded by guest on July 29, 2020 Downloaded by guest on July 29, 2020 Downloaded by guest on July 29, 2020

Transcript of Corrections Proc. Natl. Acad. Sci. USA 96 (1999) 6571 › content › pnas › 96 › 6 ›...

Page 1: Corrections Proc. Natl. Acad. Sci. USA 96 (1999) 6571 › content › pnas › 96 › 6 › 2994.full.pdf · killing, in contrast to the resistance of normal fibroblasts (8). In addition,

Immunology. In the article “Absence of tumor necrosis factorrescues RelA-deficient mice from embryonic lethality” by Taka-hiro S. Doi, Michael W. Marino, Toshitada Takahashi, ToshimichiYoshida, Teruyo Sakakura, Lloyd J. Old, and Yuichi Obata, whichappeared in number 6, March 16, 1999, of Proc. Natl. Acad. Sci.USA (96, 2994–2999), the following corrections should be noted:

In Table 1, page 2995, line 1 of the title, the second sentence,“Results of matings of TNF1y1 or TNF2y2 mice withrelA1y2 mice,” should be removed.

In Table 2, page 2997, line 2, right column, the genotype ofthe male parent which currently reads “TNF1y2relA1y1”should read “TNF2y2relA1y2.”

In Table 2, page 2997, line 3, right-most column, thegenotype of the offspring which currently reads “21y2relA1y1” should read “TNF1y2relA1y1.”

In line 2 of Discussion, page 2997, the sentence that reads“TNF mediates this abnormality and the associated lethality,as viable RelA-deficient progeny without liver damage resultfrom matings of TNF-deficient RelA-heterozygous mice withRelA-deficient mice” should read “TNF mediates this abnor-mality and the associated lethality, as viable RelA-deficientprogeny without liver damage result from matings of TNF-deficient RelA-heterozygous mice.”

Medical Sciences. In the article “RB-mediated suppression ofspontaneous multiple neuroendocrine neoplasia and lung me-

tastases in Rb1y2 mice” by Alexander Yu. Nikitin, Marıa I.Juarez-Perez, Song Li, Leaf Huang, and Wen-Hwa Lee, whichappeared in number 7, March 30, 1999, of Proc. Natl. Acad. Sci.USA (96, 3916–3921), the following correction should benoted. The labeling of the gel in Fig. 1O was erroneouslymoved down and to the right. The corrected figure and itslegend are shown below.

Immunology. In the article “Ubiquitin-dependent degradationof IkBa is mediated by a ubiquitin ligase Skp1yCul1yF-boxprotein FWD1” by Shigetsugu Hatakeyama, Masatoshi Kita-gawa, Keiko Nakayama, Michiko Shirane, Masaki Matsumoto,Kimihiko Hattori, Hideaki Higashi, Hiroyasu Nakano, KoOkumura, Kazunori Onoe, Robert A. Good, and Kei-ichiNakayama, which appeared in number 7, March 30, 1999, ofProc. Natl. Acad. Sci. USA (96, 3859–3863), due to printer’serror, the following changes should be noted. On page 3859 inthe abstract, text, and footnote, the word “Cull 1” should be“Cul1.”

Biophysics. In the article “High-resolution NMR of encapsu-lated proteins dissolved in low-viscosity f luids” by A. JoshuaWand, Mark R. Ehrhardt, and Peter F. Flynn, which appearedin number 26, December 22, 1998, of Proc. Natl. Acad. Sci.USA (95, 15299–15302), the term “high pressure NMR” wasincorrectly printed as “HPLC NMR,” due to a printer’s error.

FIG. 1. Multiple neuroendocrine neoplasia in Rb1/2 mice. (A andB) Gross compound pituitary tumor on P394. The tumor consists oftwo histologically distinct components that substitute for the pituitaryanterior (AL) and intermediate (IL) lobes, which are demarcated byremnants of Rathke’s cleft (arrows). AL tumor cells contain a-GSU(A) and are positioned loosely around sinusoid-like vessels. IL tumorcells contain a-melanocyte-stimulating hormone (B) and form poorlyvascularized epithelioid fields with central necrotic and hemorrhagicareas. (C and D) EAP in the anterior pituitary lobe on P90 before (C)and after (D) microdissection for genotype analysis by the PCR. (E)C-cell carcinoma on P340 showing the typical arrangement of poly-hedral tumor cells in solid nests and rough hyalinized collagen withcalcification (arrow). (F and G) C-cell EAP (arrow) on P60 before (F)and after (G) microdissection. The atypical cells show a parafollicularlocation. (H) Medullary thyroid carcinoma invading surroundingtissues on P469. Accumulation of calcitonin (brown color) is evidentin the cytoplasm of tumor cells (arrow). CR, cartilage. (I) Calcitonin-containing metastatic cells in the lung on P463. The metastatic cellsexhibit intraalveolar spreading (arrows). (J) A well-vascularized para-thyroid tumor (PG) and a neighboring solid C-cell tumor of the thyroidgland (TG) on P370. (K) Parathyroid hormone expression in para-thyroid tumor cells. (L) Pheochromocytoma of the adrenal medulla(AM) compressing the adrenal cortex (AC). (M and N) EAP in theadrenal medulla (AM) on P60 before (M) and after (N) microdissec-tion. The arrow indicates multiple apoptotic figures. Staining: avidin-biotin-peroxidase immunostaining for a-GSU (A), a-melanocyte-stimulating hormone (B), calcitonin (H and I), or parathyroid hor-mone (K) with hematoxylin counterstaining; (C–G, J, and L–N)hematoxylin-eosin staining. [Bar: 160 mm (A and B), 40 mm (C, D, andI), 110 mm (E), 60 mm (F and G), 50 mm (H), 150 mm (J), and 20 mm(K), 390 mm (L), and 70 mm (M and N).] (O) Absence of the wild-typeRb allele (151-bp PCR product) in gross tumors (T; lanes 3, 4, 7, 10,11, 17, and 18) and EAPs (E; lanes 1, 2, 6, 8, 9, 14, and 16) of thepituitary anterior lobe (AL, lanes 1–4), the parathyroid gland (PG,lanes 6 and 7), thyroid C cells (TG, lanes 8–11), lung metastases (L,lanes 12 and 13), or adrenal medulla (lanes 14 and 16–18). N, Rb1/2

normal tissue (lanes 5 and 15). Nondenaturing 12% polyacrylamide gelstained with silver. The 236-bp band corresponds to the mutant Rballele (11).

Corrections Proc. Natl. Acad. Sci. USA 96 (1999) 6571

Dow

nloa

ded

by g

uest

on

July

29,

202

0 D

ownl

oade

d by

gue

st o

n Ju

ly 2

9, 2

020

Dow

nloa

ded

by g

uest

on

July

29,

202

0 D

ownl

oade

d by

gue

st o

n Ju

ly 2

9, 2

020

Dow

nloa

ded

by g

uest

on

July

29,

202

0 D

ownl

oade

d by

gue

st o

n Ju

ly 2

9, 2

020

Dow

nloa

ded

by g

uest

on

July

29,

202

0 D

ownl

oade

d by

gue

st o

n Ju

ly 2

9, 2

020

Page 2: Corrections Proc. Natl. Acad. Sci. USA 96 (1999) 6571 › content › pnas › 96 › 6 › 2994.full.pdf · killing, in contrast to the resistance of normal fibroblasts (8). In addition,

Pharmacology. In the article “Nuclear localization of prosta-glandin E2 receptors” by Mousumi Bhattacharya, Krishna G.Peri, Guillermina Almazan, Alfredo Ribeiro-da-Silva, HitoshiShichi, Yves Durocher, Mark Abramovitz, Xin Hou, Daya R.Varma, and Sylvain Chemtob, which appeared in number 26,December 22, 1998, of Proc. Natl. Acad. Sci. USA (95, 15792–15797), the paragraph on page 15793, left column, should readas follows (change indicated in italic type): “Indirect Immu-

nofluorescence of EP1 Receptors. For examining the immu-nolocalization of EP1 receptors, immunocytochemistry wasperformed as described (21) on Swiss 3T3, HEK293 (EBNA),or endothelial cells with rabbit anti-EP1 antibodies (22) andFITC-conjugated or Texas red-conjugated anti-rabbit IgG(BioyCan Scientific, Mississauga, ON), diluted 1:50.” In ad-dition, because Figs. 6 and 7 were printed with markedly poorquality, they and their legends are reproduced below.

FIG. 7. Immunoperoxidase localization of EP1 in adult rat braincortex by electron microscopy (arrows). Specific immunostainingobserved in plasma membrane and inner and outer nuclear mem-branes of capillary endothelial cell (a) and nuclear membranes ofneurons (b). Note the luminal space of capillary on right in a. (Bars 50.5 mm.)

FIG. 6. Immunoperoxidase and immunogold localization of EP1 inporcine endothelial cells detected by electron microscopy (arrows). (a)Immunoperoxidase-IgG alone; note absence of immunostaining whenprimary antibody is omitted. (b) A low magnification showing immuno-staining in plasma membrane and nuclear envelope. (c) A higher mag-nification showing immunostaining in the nuclear envelope. (d) Immu-nogold-IgG alone; note absence of immunostaining. Specific immuno-staining can be observed in the plasma membrane in e, nuclear envelopein f, and Golgi apparatus in g. (Bar 5 0.5 mm, except in b 5 2 mm.)

6572 Corrections Proc. Natl. Acad. Sci. USA 96 (1999)

Dow

nloa

ded

by g

uest

on

July

29,

202

0

Page 3: Corrections Proc. Natl. Acad. Sci. USA 96 (1999) 6571 › content › pnas › 96 › 6 › 2994.full.pdf · killing, in contrast to the resistance of normal fibroblasts (8). In addition,

Microbiology. In the article “Molecular and biophysical char-acterization of TT virus: Evidence for a new virus familyinfecting humans” by Isa K. Mushahwar, James C. Erker, A.Scott Muerhoff, Thomas P. Leary, John N. Simons, Larry G.Birkenmeyer, Michelle L. Chalmers, Tami J. Pilot-Matias, andSuresh M. Dexai, which appeared in number 6, March 16, 1999,of Proc. Natl. Acad. Sci. USA (96, 3177–3182), the followingcorrection should be noted. The last author’s name has beenmisspelled, and it should be amended as Suresh M. Desai andnot Suresh M. Dexai.

Physiology. In the article “Parathyroid hormone leads to thelysosomal degradation of the renal type II NayPi cotrans-porter” by Markus F. Pfister, Isabelle Ruf, Gerti Stange, UrsZiegler, Eleanor Lederer, Jurg Biber, and Heini Murer, whichappeared in number 4, February 17, 1998, of Proc. Natl. Acad.Sci. USA (95, 1909–1914), the authors wish to note that theAcknowledgment section should have included the followinggrant information: “Dr. Lederer is an employee of and herwork is supported by a grant from the U.S. Department ofVeterans Affairs.”

Corrections Proc. Natl. Acad. Sci. USA 96 (1999) 6573

Dow

nloa

ded

by g

uest

on

July

29,

202

0

Page 4: Corrections Proc. Natl. Acad. Sci. USA 96 (1999) 6571 › content › pnas › 96 › 6 › 2994.full.pdf · killing, in contrast to the resistance of normal fibroblasts (8). In addition,

Proc. Natl. Acad. Sci. USAVol. 96, pp. 2994–2999, March 1999Immunology

Absence of tumor necrosis factor rescues RelA-deficient micefrom embryonic lethality

TAKAHIRO S. DOI*†‡, MICHAEL W. MARINO†§, TOSHITADA TAKAHASHI*, TOSHIMICHI YOSHIDA¶,TERUYO SAKAKURA¶, LLOYD J. OLD§i, AND YUICHI OBATA*i

*Laboratory of Immunology, Aichi Cancer Center Research Institute, Nagoya 464, Japan; §Ludwig Institute for Cancer Research, New York Branch at MemorialSloan-Kettering Cancer Center, 1275 York Avenue, New York, NY 10021; and ¶Department of Pathology, Mie University School of Medicine, Tsu 514, Japan

Contributed by Lloyd J. Old, December 30, 1998

ABSTRACT Mice lacking the RelA (p65) subunit ofNF-kB die between days 14 and 15 of embryogenesis becauseof massive liver destruction. Fibroblasts and macrophagesisolated from relA2y2 embryos were found to be highlysensitive to tumor necrosis factor (TNF) cytotoxicity, raisingthe possibility that endogenous TNF is the cause of liver cellapoptosis. To test this idea, we generated mice lacking bothTNF and RelA. Embryogenesis proceeds normally in suchmice, and TNFyRelA double-deficient mice are viable and havenormal livers. Thus, the RelA-mediated antiapoptotic signalthat protects normal cells from TNF injury in vitro can beshown to be operative in vivo.

One of the defining characteristics of tumor necrosis factor(TNF) is its cytostaticycytotoxic activity in vitro for certainmouse and human transformed cells (1, 2). Early studiesshowed that cytotoxicity could be enhanced by inhibitors ofprotein or RNA synthesis (3) and that cell killing had thefeatures of an apoptotic process (4, 5). Although suspicionswere raised that TNF cytotoxicity might be an epiphenomenonnot relevant to the biological functions of TNF, the discoveryof a growing family of ligands and receptors related to TNF(6)—most notably FasyFas ligand with potent apoptosis in-ducing activity—provided support for the idea that TNF-mediated cytotoxicity has physiological significance. Furthersupport comes from recent findings implicating NF-kB in TNFcytotoxicity. Although activation of NF-kB was known tomediate several activities of TNF (7), recent studies indicatethat NF-kB activation by TNF protects cells from TNF cyto-toxicity (8–11). Thus, fibroblasts from mice with a deletion ofthe RelA component of NF-kB are highly sensitive to TNFkilling, in contrast to the resistance of normal fibroblasts (8).In addition, inhibition of NF-kB activity by overexpression ofIkB greatly increases the sensitivity of cells to TNF cytotoxicity(9–11). Mice lacking the RelA component of NF-kB die inutero at days 14–15, and the major morphological abnormalityin these mice at the time of death is massive liver degenerationassociated with apoptosis of hepatocytes (12, 13). Because ofthe sensitivity of fibroblasts and macrophages from RelA-deficient mice to TNF-mediated cytotoxicity, Beg and Balti-more (8) suggested that endogenous production of TNF wasresponsible for the killing of hepatocytes in these mice. Wetested this hypothesis by mating TNF-deficient and RelA-deficient mice to determine what effect absence of TNF has onthe RelA-deficient phenotype.

MATERIALS AND METHODS

Derivation and Maintenance of Knockout Mice. TNF-deficient mice and RelA-deficient mice have been described in

detail elsewhere (13, 14). relA1y2 mice backcrossed to C57BLy6 mice for 10 successive generations and TNF2y2 micebackcrossed to C57BLy6 mice for 5 generations were used inthis study. Mice were maintained in a specific pathogen-freemouse facility at Aichi Cancer Center Research Institute.

DNA and RNA Blot Analyses. DNA and total RNA wereisolated by using proteinase KySDS and guanidium thiocya-nateyCsCl procedures, respectively (15, 16). For genotyping,DNA from developing embryos and limbs and tails of post-natal mice was digested with restriction enzymes and separatedby agarose gel electrophoresis and then transferred to nitro-cellulose filters (Schleicher & Schuell). To confirm disruptionof the TNF gene, DNA digested with BamHI was examinedwith a 1,218-bp SmaI fragment from the lymphotoxin-b gene(nucleotides 3,181–4,399, GenBank accession no. U06950), akind gift of Sergei Nedospasov. For analyzing relA disruption,DNA digested with PstI was analyzed with an 850-bp PstI–SmaI fragment of the 59-untranslated region of the relA gene(13). Total RNA was separated by 2.2 M formaldehyde agarosegel electrophoresis and transferred to nitrocellulose filters.RNA blots were analyzed by using cDNA probes for relA(codons 185–277, ref. 17), c-rel (codons 144–277, ref. 18), andrelB (codons 458–580, ref. 19).

Immunohistochemistry and Histology. Five-micrometerfrozen sections of whole embryos were air-dried, fixed withcold acetone, and reacted with hamster anti-murine TNF mAb(ref. 20; kindly provided by Dr. Robert Schreiber) or apolyclonal rabbit anti-mouse TNF antibody (Genzyme). Bind-ing was visualized by an avidin–biotin enzyme complexmethod (Vectastain Elite ABC kit; Vector Laboratories), withsubsequent counterstaining with hematoxylin. For histologicalexamination, tissues were fixed in 4% paraformaldehyde andembedded in paraffin. The sections (5 mm) were stained withhematoxylinyeosin.

Reverse Transcription–PCR (RT-PCR) Analysis of the Ex-pression of TNF, TNF Receptor (TNFR)1, and TNFR2. Theexpression of TNF, TNFR1, and TNFR2 was determined byusing RT-PCR analysis. Total RNA (10 mg) isolated fromtissues or cultured cells of 14.5-day postcoitum (dpc) embryosor from cultured cells of 3-day postnatal mice was used forcDNA synthesis using oligo(dT) primers and 200 units ofSuperScript II (Life Technologies, Gaithersburg, MD). PCRwas performed in a final volume of 50 ml containing all fourdNTPs, 2 mM MgCl2, 2.5 units of Taq polymerase (TakaraShuzo, Otsu, Japan), and each primer at 0.2 mM by using aProgram Control System PC-700 (Astec, Fukuoka, Japan).Each amplification cycle consisted of 93°C for 1 min, 60°C for1 min, and 72°C for 1 min. After 30 cycles, PCR products were

The publication costs of this article were defrayed in part by page chargepayment. This article must therefore be hereby marked ‘‘advertisement’’ inaccordance with 18 U.S.C. §1734 solely to indicate this fact.

PNAS is available online at www.pnas.org.

Abbreviations: TNF, tumor necrosis factor; TNFR, TNF receptor;RT-PCR, reverse transcription–PCR; dpc, days postcoitum.†T.S.D. and M.W.M. contributed equally to this work.‡Present address: Ludwig Institute for Cancer Research, New YorkBranch at Memorial Sloan-Kettering Cancer Center, 1275 YorkAvenue, New York, NY 10021.

iTo whom reprint requests should be addressed.

2994

Page 5: Corrections Proc. Natl. Acad. Sci. USA 96 (1999) 6571 › content › pnas › 96 › 6 › 2994.full.pdf · killing, in contrast to the resistance of normal fibroblasts (8). In addition,

separated by electrophoresis on a 1.5% agarose gel andvisualized by ethidium bromide staining. The primers used inthese experiments were designed to distinguish authenticRT-PCR products from PCR products generated from con-taminating genomic DNA as described (21).

TNF Cytotoxicity. Cells from liver and brain and fibroblastsof 14.5-dpc embryos and cells from spleen, kidney and heart of3-day old mice were prepared according to methods describedpreviously (22). Briefly, liver, kidney, and heart cells wereplated on collagen type I-coated plastic dishes (Becton Dick-inson Labware) and cultured in DMEM (Life Technologies,Gaithersburg, MD) with 10% fetal calf serum. Fibroblasts andcells from brain were plated on plastic dishes in DMEM with10% fetal calf serum. Spleen cells were cultured in RPMImedium 1640 (Life Technologies, Gaithersburg, MD) with10% fetal calf serum. Cultured cells from embryonic liverexpressed a-fetoprotein, and cells from heart continued toshow pulsation in vitro. After 24 hours in culture, recombinantmurine TNF (Sigma) was added at a final concentration of 1,10, or 100 ngyml. Viability was examined by using trypan blueexclusion.

Injection of TNF. Newborn mice from the mating ofTNF2y2relA1y2 mice were injected intravenously with 1 mgor 0.2 mg of recombinant murine TNF (Sigma). Mice weremonitored for 24 hours after injection and then genotyped bySouthern blot analysis.

RESULTS

TNFyRelA Double-Deficient Mice. Mice lacking relA die inutero around 14.5 days postcoitum (dpc), as shown in Table 1,confirming previous results (12, 13). To determine the role ofTNF in the embryonic death of RelA-deficient mice, mice

heterozygous for the relA gene deletion were crossed toTNF-deficient mice for two successive generations. From thesecond generation, TNF2y2relA1y2 mice were selected andintercrossed. As shown in Table 1, matings of TNF2y2relA1y2gave rise to viable TNF2y2relA2y2 progeny in the predictedMendelian ratio. This is in contrast to the embryonic lethality ofmice with the TNF1y1relA2y2 genotype (Table 1). In a broadsurvey of tissues from double-deficient mice, no morphologicalabnormalities were found, either in liver or other tissues, incontrast to the liver degeneration in mice withTNF1y1 relA2y2 genotype (Fig. 1). Thus, elimination of TNFrescues RelA-deficient mice from embryonic lethality by abol-ishing TNF-mediated liver degeneration. Furthermore, in theabsence of TNF, RelA is dispensable for embryonic and postnataldevelopment.

TNF2y2relA2y2 mice are healthy and active during thefirst 4–5 weeks of life. After approximately 40 days of age,however, the mice become sick and die. At 40 days of age,infiltration of lymphocytes was found in both portal areas andlobules of the liver (Fig. 2 A–C). In the lung, thickening ofalveolar walls with inflammatory cell infiltration was observed(Fig. 2E). Inflammatory infiltrates also were seen around thebronchi and the pulmonary blood vessels. In the kidney,glomerular abnormalities with expansion of mesangial areasand thickening of capillary walls were seen (Fig. 2G). No suchabnormalities were observed in TNF2y2relA1y1 mice (Fig.2 D, F, and H). The inflammatory changes are most likelyrelated to the compromised immune system of TNFyRelAdouble-deficient mice. As mice deficient in TNF (14, 23) ormice reconstituted with relA2y2 lymphocytes (13, 24) haveimpaired immune systems, mice lacking both TNF and RelAwould be expected to be highly susceptible to microbial andviral infections.

FIG. 1. Normal development of the liver in TNF2y2relA2y2 mice. (A) TNF1y1relA2y2 14.5-dpc embryo. No viable hepatocytes can beseen. (B) TNF2y2relA2y2 14.5-dpc embryo. (C) TNF2y2relA2y2 newborn mouse. (D) TNF2y2relA1y1 newborn mouse. Sections arehematoxylinyeosin-stained. (3400.)

Table 1. Influence of TNF genotype on survival of RelA-deficient mice. Results of matings of TNF1y1 or TNF2y2 mice withrelA1y2 mice

Mouse age

TNF1y1relA1y2 (/) 3 TNF1y1relA1y2 (?) TNF2y2relA1y2 (/) 3 TNF2y2relA1y2 (?)

TNF1y1relA2y2 TNF1y1relA1y2 TNF1y1relA1y1 TNF2y2relA2y2 TNF2y2relA1y2 TNF2y2relA1y1

14.5 dpc 11 31 12 7 13 615.5 dpc 3 24 6 — — —16.5 dpc 0 25 11 10 22 12Newborn 0 18 9 25 62 2921 Days old 0 146 77 9 22 10

Data are given as number of mice generated.

Immunology: Doi et al. Proc. Natl. Acad. Sci. USA 96 (1999) 2995

Page 6: Corrections Proc. Natl. Acad. Sci. USA 96 (1999) 6571 › content › pnas › 96 › 6 › 2994.full.pdf · killing, in contrast to the resistance of normal fibroblasts (8). In addition,

Origin of TNF in Embryos. To test whether TNF present inembryos is produced by the embryos themselves or producedby mothers and transmitted to the embryos through theplacenta, two sets of crosses (female TNF2y2relA1y2 matedwith male TNF1y1relA1y2 and female TNF1y1relA1y2with male TNF2y2relA1y2) were set up (Table 2). MaternalTNF was not the cause of death of TNF1y2relA2y2 progeny,

as the timing of embryonic lethality did not differ in theTNF1y2relA2y2 progeny from both crosses. Thus, TNF isproduced by the embryos themselves. During this study, it wasnoted that TNF1y2relA2y2 mice survived longer thanTNF1y1relA 2y2 mice, as some of the former were aliveeven up to birth whereas the latter died between 14.5 and 15.5dpc (Tables 1 and 2). This gene dosage effect of TNF on thesurvival of relA2y2 embryos is consistent with previousobservations showing less TNF production in TNF1y2 miceas compared with TNF1y1 mice (14).

To examine TNF production during development, embryosof 11.5- to 13.5-dpc mice were examined by immunohistochem-istry using a hamster anti-TNF mAb (20) and a polyclonalrabbit anti-mouse TNF antibody. The specificity of thesereagents for TNF was confirmed by their complete lack ofreactivity with normal tissues from TNF2y2 mice. Stainingwith these antibodies showed that immunoreactive TNF ispresent in most tissues of TNF1y1 embryos, including liver.Although staining tended to be diffuse and difficult to ascribeto discrete cell populations, there were a small number of cellsin the liver that were strongly stained (Fig. 3). By usingRT-PCR, TNF transcripts were detected in all embryonictissues tested and especially strongly in liver, spleen, andthymus (Fig. 4).

Expression of TNF Receptors and NF-kB Subunits inTissues of Embryonic Mice. Although TNF appears to bepresent ubiquitously in the embryo, tissue destruction wasrestricted to the liver at the time of death of TNF1y1 relA2y2mice. To explore the reason for this tissue specific destructionby TNF, the expression of TNF receptors and NF-kB familymembers was examined. By using RT-PCR, transcripts ofTNFR1 and TNFR2 were detected in all embryonic tissuestested (Fig. 4). Among NF-kB family members, relA mRNAwas ubiquitously expressed at high levels. relB mRNA was alsowidely expressed, but showed particularly high expression inplacenta, thymus, and liver. Transcripts of the c-rel gene werebelow the level of detection in these embryonic tissues exceptfor trace amounts in spleen. This was also the case in RelA-deficient mice, where no compensatory increase in expressionof other NF-kB members was found (13).

TNF Sensitivity of Cultured Cells from RelA-DeficientMice. To analyze cell type differences in TNF sensitivity,cultured cells derived from various tissues ofTNF2y2relA2y2 or TNF2y2relA1y1 mice were examinedfor their sensitivity to TNF. As shown Fig. 5, cells fromrelA2y2 liver and kidney, as well as embryonic fibroblasts,were sensitive to TNF, whereas cells from relA2y2 spleen,heart, and brain were not. All cell types from relA1y1 micewere resistant to TNF treatment. Transcripts of TNFR1 and 2were detectable in all these cultured cells (Fig. 6). Theexpression of NF-kB family members in cultured cells fromrelA wild-type mice was examined by using Northern blotanalysis. Transcripts of relA and relB were present in all cellstested. Transcripts of c-rel were detected in cells derived fromspleen, heart, and brain, but not in cells from kidney orembryonic fibroblasts. Low levels of c-rel expression weredetected in cultured cells from the liver, but this may beaccounted for by residual hematopoietic cells derived from theembryonic liver. The presence of c-rel expression generallycorrelates well with resistance of cells to TNF. Thus, c-rel,when it is expressed, may also mediate antiapoptotic signalsthat protect cells from TNF cytotoxicity.

Sensitivity of RelA-Deficient Mice to Exogenous TNF. TNFwas injected intravenously into newborn mice from inter-crosses between TNF2y2relA1y2 mice. With 1 mg of TNF,all TNF2y2relA2y2 mice died within 6 hours, whereasTNF2y2relA1y2 and TNF2y2relA1y1 mice survivedlonger than 12 hours (Fig. 7). Influence of relA gene dosage onTNF sensitivity was observed, with longer survival in relA1y1than relA1y2 mice. Similar results were obtained with 0.2 mg

FIG. 2. Morphological abnormalities in TNF2y2relA2y2 40-day-old mice. (A–C) Liver from TNF2y2 relA2y2 40-day-old mouse. (A)Portal areas and parenchyma showing marked cellular infiltrates.(3150). (B) Predominantly lymphocytic infiltrate in portal areas withbile ducts showing regenerative changes. (3450). (C) Intrasinusoidalaggregates of inflammatory cells. Megakaryocytes were present, butother hematopoietic cells were not observed. (3450). (D) Liver fromTNF2y2relA1y1 40-day-old mouse. No abnormalities observed.(3150.) (E) Lung from TNF2y2relA2y2 40-day-old mouse. Thick-ened alveolar walls associated with lymphocytic infiltration, activatedpneumocytes, and increased mucous cells in bronchi. (3150) (F) Lungfrom TNF2y2relA1y1 40-day-old mouse. No abnormalities ob-served. (3150.) (G) Kidney from TNF2y2relA2y2 40-day-oldmouse. Expanded mesangial areas in glomeruli with increased mes-angial matrix and numbers of cells. Marked thickening of capillarywalls with narrowing of the capillary lumen. (3300.) (H) Kidney fromTNF2y2relA1y1 40-day-old mouse. No abnormalities observed.(3300.)

2996 Immunology: Doi et al. Proc. Natl. Acad. Sci. USA 96 (1999)

Page 7: Corrections Proc. Natl. Acad. Sci. USA 96 (1999) 6571 › content › pnas › 96 › 6 › 2994.full.pdf · killing, in contrast to the resistance of normal fibroblasts (8). In addition,

of TNF. In moribund double-deficient mice, no parenchymaldamage was observed in liver or other organs. However,capillary damage was seen in liver, kidney, and lung, suggestingthat endothelial cells are the main target of TNF injury in thesemice. More extensive capillary damage was observed in TNF-treated TNF2y2relA2y2 mice than in TNF2y2relA1y2and TNF2y2relA1y1 mice.

DISCUSSION

Liver degeneration with absence of hepatocytes is the majorpathology found in RelA-deficient 14.5-day embryos. TNFmediates this abnormality and the associated lethality, asviable RelA-deficient progeny without liver damage resultfrom matings of TNF-deficient RelA-heterozygous mice withRelA-deficient mice. The hepatocytes derived from double-deficient mice are sensitive to the cytotoxic action of TNF invitro, suggesting that loss of hepatocytes in RelA-deficient miceis caused by a direct action of TNF on hepatocytes. Anotherpossibility is that the death of hepatocytes is secondary toendothelial cell apoptosis, and the capillary damage observedin the liver and other organs of TNF2y2relA2y2 miceinjected with TNF would support an indirect mechanismleading to hepatocyte loss. Whether caused by a direct or anindirect action of TNF on hepatocytes, the organ specificity(liver) and timing of TNF action in RelA deficient mice needexplanation, especially because (i) TNF is widely expressed inembryonic tissues and is detectable before 14.5 days, (ii)organs other than liver (e.g., kidney) contain cells that aresensitive to TNF cytotoxicity, and (iii) capillary damage in-duced by TNF in double-deficient mice is not restricted to liver.Possibly, a surge of TNF production occurs around 14–15 daysof embryonic life, and liver is the target of damage because aparticularly high concentration of TNF is produced by Kuppfercells in the developing liver.

In view of the cytotoxic action of TNF, we and others havelong speculated that TNF is involved in the cell death thataccompanies normal morphogenesis and organogenesis. How-ever, no developmental abnormalities have been detected inTNF2y2 mice, indicating that TNF does not have an irre-placeable role in this process (14, 23). Where TNF may be

essential is in the elimination of cells damaged by chemical orphysical agents or by infectious agents and their products. Ifthis were the case, there would be a critical need for amechanism, e.g., the NF-kB antiapoptotic signal, to protectnormal cells. The increased TNF sensitivity of cells treated invitro with protein or RNA synthesis inhibitors (3) may wellhave its counterpart in mice treated with these agents in vivo,and there is a speculation that TNF-mediated apoptosis isinvolved in the antitumor effects of irradiation and chemo-therapeutic agents used in clinical practice. As a model for thissurveillance role of TNF in eliminating damaged cells, miceinjected with galactosamine (an agent that specifically inhibitstranscriptional activity in hepatocytes) become exquisitelysensitive to bacterial lipopolysaccharide (25), with the liver ofgalactosamineylipopolysaccharide-treated mice showing mas-sive necrosis. This action of lipopolysaccharide and galac-tosamine is mediated by TNF (14, 23, 26, 27). In addition, cellsinfected with bacteria (28) or with a number of differentviruses, including herpesviruses (29) and HIV (30), becomesensitive to TNF-mediated cytotoxicity. Exotoxins, a productof Gram-positive organisms, also greatly enhance TNF cyto-toxicity (31). In fact, the lethality of bacterial sepsis may wellbe due to the sensitization of cells in key organs by bacterialproducts to the apoptotic damage of TNF rather than to thecurrently held view that TNF-induced inflammatory damageand circulatory collapse are the primary pathogenic factors inseptic shock, and recent evidence for TNF-mediated apoptosisof endothelial cells in lipopolysaccharide-treated mice (32) isconsistent with this idea. It will be important to determinewhether regulation of the NF-kB antiapoptotic signal is theunifying element in these examples of selective cytotoxiceffects of TNF on damaged or infected cells. With the rapidexpansion of the TNF ligandyTNFR superfamily, a number ofnew cytotoxic factors and new mechanisms for resistance tocytotoxicity have been identified, including expression of thedecoy receptors, DcR1 and DcR2, in the TRAIL (Apo2)yDR5

FIG. 3. Immunostaining of TNF in the liver of 14.5 dpc embryowith hamster anti-mouse TNF antibody. Shown are TNF1y1 (A) andTNF2y2 (B) embryos. (3400)

FIG. 4. Expression of TNF, TNFR1, TNFR2, relA, c-rel, and relB.RNA was isolated from various tissues and cultured fibroblasts ofTNF1y1RelA1y1 14.5 dpc embryos. Lanes: (1) fibroblasts, (2) liver,(3) kidney, (4) spleen, (5) heart, (6) brain, (7) thymus, (8) lung, (9)intestine, and (10) placenta. The expression of TNF, TNFR1, andTNFR2 was analyzed by using RT-PCR. The expression of relA, c-rel,and relB was examined by using Northern blot analysis.

Table 2. Influence of parental TNF genotype and TNF gene dosage on embryonic lethality of relA2y2 mice

Mouse age

TNF2y2relA1y2 (/) 3 TNF1y1relA1y2 (?) TNF1y1relA1y2 (/) 3 TNF1y2relA1y1 (?)

TNF1y2relA2y2 TNF1y2relA1y2 TNF1y2relA1y1 TNF1y2relA2y2 TNF1y2relA1y2 21y2relA1y1

16.5 dpc 5 10 7 7 19 818.5 dpc 3 17 8 4 22 12Newborn 2 24 16 1 39 1921 Days old 0 18 8 0 25 13

Data are given as number of mice generated.

Immunology: Doi et al. Proc. Natl. Acad. Sci. USA 96 (1999) 2997

Page 8: Corrections Proc. Natl. Acad. Sci. USA 96 (1999) 6571 › content › pnas › 96 › 6 › 2994.full.pdf · killing, in contrast to the resistance of normal fibroblasts (8). In addition,

and DR4 pathways (33–35), and tissue-restricted receptorexpression in the Apo3LyDR3 (Apo3yWsl1) system (36).However, recent evidence that NF-kB may have an importantantiapoptotic role in the Apo3LyDR3 pathways (36, 37)suggests that activation of NF-kB may be a more generalprotective mechanism, and further studies of NF-kB-mediatedantiapoptotic signals in other TNF ligandyTNFR superfamilyare needed. TNF also is known to induce reactive oxygenspecies, and reactive oxygen species can mediate cytotoxicityand NF-kB activation (38). Defining the precise role of NF-kBin TNF-induced reactive oxygen species production, action,and protection of cells against reactive oxygen species cyto-toxicity will be facilitated by availability of NF-kB-deficientmice. Similarly, the role of NF-kB in ceramide generation andapoptosis induction through the sphingomyelin pathway initi-ated by TNF can also be clarified in this way (39).

Cultured cells derived from several tissues of TNFyRelAdouble-deficient mice showed differential sensitivity to TNF;hepatocytes, kidney cells, and fibroblasts are sensitive to TNFcytotoxicity in vitro whereas those from heart, brain, andspleen are resistant. Given the capacity of high level c-rel

expression to overcome the TNF sensitivity of cell linesexpressing the IkB mutant (9), the expression of c-rel is apossible reason for the resistance of these cells to TNF.However, the contribution of c-rel to protection against TNF-mediated apoptosis or other cytotoxic agents has been difficultto determine because of the ubiquitous expression of RelA.TNFyRelAyc-rel triple-deficient and TNFyc-rel double-deficient offspring produced by a cross between TNFyRelAdouble-deficient mice and c-rel-deficient mice (40) shouldfacilitate definition of the role of c-rel in NF-kB-dependentresistance to TNF.

The search for TNF-mediatedyNF-kB-dependent ‘‘surviv-al’’ genes has become a major objective in recent years. Severalcandidate ‘‘survival’’ genes have been identified, includingIEX-1L (41), TRAF1 (TNFR-associated factor 1), TRAF2,c-IAP (cellular inhibitor of apoptosis)-1 and c-IAP-2 (42), andA20 (43). Transcription of IEX-1L is induced by TNF, andthere is decreased IEX-1L transcription in NF-kB-compro-mised cells. Furthermore, IEX-1L expression in transfectedrelA2y2 cells or in mutant IkBa cells renders these cellsresistant to TNF treatment (41). The simultaneous expressionof TRAF1, TRAF2, c-IAP-1, and c-IAP-2, but not the indi-vidual expression of these genes, was reported to protectrelA2y2 cells and mutant IkBa-transfected cells from TNF-mediated cytotoxicity (42). Although previous reports haveestablished that TRAF2, c-IAP-1, and c-IAP-2 mediateNF-kB activation by TNF, recent evidence indicates that, likeIEX-1L, all four of these genes appear to be rapidly up-regulated by NF-kB activation, (9, 44). Collectively, it seemslikely that upon TNF receptor activation, the NF-kByTRAF1,TRAF2, c-IAP-1, and c-IAP-2 pathways operate as a positivefeedback system to amplify the survival signal to protect cellsfrom TNF injury. With regard to the A20 zinc finger proteinin TNF-mediated cytotoxicity, some studies show a protectiverole in TNF cytotoxicity (43), whereas others indicate that A20inhibits NF-kB activation (45). Such contradictions might beresolved by the discovery of NF-kB-‘‘independent’’ pathways,suggested already by studies on TRAF2-deficient mice and ontransgenic mice with a dominant negative form of TRAF2 (46,47). TRAF2-compromised cells showed increased sensitivityto TNF cytotoxicity despite normal activation of NF-kB byTNF. In TRAF2-compromised cells, Jun N-terminal kinaseactivation induced by TNF treatment was severely reduced,and genes downstream of Jun N-terminal kinase may also beinvolved in antiapoptosis. TNFyTRAF2 double- and TNFyTRAF2yRelA triple-deficient mice should facilitate testing ofthis hypothesis. How these candidate survival genes exert theirantiapoptotic action is not resolved at present. A recent reportindicated that the expression of TRAF1, TRAF2, c-IAP-1, andc-IAP-2 suppresses apoptosis by blocking activation of theinitiator caspase, caspase-8 (42). Other studies, however, sug-gested that IAPs inhibit apoptosis by blocking activities of

FIG. 5. TNF cytotoxicity in vitro. Cultured cells were prepared asdescribed in Materials and Methods and were treated with TNF for 8,24, or 48 hours. TNF2y2relA2y2 cells with no TNF (h);TNF2y2relA2y2 cells with 10 ngyml TNF (‚); TNF2y2relA2y2cells with 100 ngyml TNF (E) and TNF2y2relA1y1 cells with 100ngyml TNF (F). Viability was examined by using trypan blue exclusion.

FIG. 6. The expression of TNFR1, TNFR 2, relA, c-rel, and relB incultured cells. RNA was isolated from cultured cells of TNF2y2relA1y1 embryos or postnatal mice (see Fig. 5). Lanes: (1)hepatocytes (14.5 dpc) (2) fibroblasts (14.5 dpc), (3) kidney cells(3-day-postnatal), (4) heart cells (3-day-postnatal), (5) spleen cells(3-day-postnatal), and (6) brain cells (14.5 dpc). The expression ofTNFR1 and TNFR2 was analyzed by using RT-PCR. The expression ofrelA, c-rel, and relB was examined by using Northern blot analysis.

FIG. 7. Survival of newborn mice after injection of TNF. TNF2y2relA2y2 (solid line; n 5 8), TNF2y2relA1y2 (dashed line; n 5 15),TNF 2y2relA1y1 (broken line; n 5 5). Mice were injected with 1 mgof TNF intravenously.

2998 Immunology: Doi et al. Proc. Natl. Acad. Sci. USA 96 (1999)

Page 9: Corrections Proc. Natl. Acad. Sci. USA 96 (1999) 6571 › content › pnas › 96 › 6 › 2994.full.pdf · killing, in contrast to the resistance of normal fibroblasts (8). In addition,

effector caspases (48). Furthermore, the NF-kB-dependentsurvival pathway has been thought to operate independently ofBcl-2, the other major antiapoptotic pathway. However, arecent study showed that Bcl-2 regulates NF-kB activationthrough induction of IkB degradation (49).

Clearly, a comprehensive picture of TNF-mediatedyNF-kB-dependent antiapoptotic signaling has not yet emerged. De-fining the nature of the NF-kB dependent antiapoptotic signaland identifying its site of action in the apoptotic cascadeinitiated by TNF are critical to understanding why normal cellsare usually resistant to the cytotoxic actions of TNF, and whythis resistance is sometimes compromised in injured cells.Furthermore, it will be important to know whether abnormal-ities in NF-kB signaling are central to the high sensitivity ofcertain transformed cells of mouse and human origin to TNFcytotoxicity that occurs in the absence of any added protein orRNA synthesis inhibitors.

1. Carswell, E. A., Old, L. J., Kassel, R. L., Green, S., Fiore, N. &Williamson, B. (1975) Proc. Natl. Acad. Sci. USA 72, 3666–3670.

2. Williamson, B. D., Carswell, E. A., Rubin, B. Y., Prendergast,J. S. & Old, L. J. (1983) Proc. Natl. Acad. Sci. USA 80, 5397–5401.

3. Ruff, M. R. & Gifford, G. E. (1981) Lymphokines 2, 235–272.4. Dealtry, G. B., Naylor, M. S., Fiers, W. & Balkwill, F. R. (1987)

Eur. J. Immunol. 17, 689–693.5. Rubin, B. Y., Smith. L. J., Hellermann, G. R., Lunn, R. M.,

Richardson, N. K. & Anderson, S. L. (1988) Cancer Res. 48,6006– 6010.

6. Ashkenazi, A. & Dixit, V. M. (1998) Science 281, 1305–1308.7. Baeuerle, P. A. & Baichwal, V. R. (1997) Adv. Immunol. 65, 111–

137.8. Beg, A. A. & Baltimore, D. (1996) Science 274, 782–784.9. Liu, Z., Hsu, H., Goeddel, D. V. & Karin, M. (1996) Cell 87,

565–576.10. Wang, C.-Y., Mayo, M. W. & Baldwin, A. S., Jr. (1996) Science

274, 784–787.11. Van Antwerp, D. J., Martin, S. J., Kafri, T., Green, D. R. &

Verma, I. M. (1996) Science 274, 787–789.12. Beg, A. A., Sha, W. C., Bronson, R. T., Ghosh, S. & Baltimore,

D. (1995) Nature (London) 376, 167–170.13. Doi, T. S., Takahashi, T., Taguchi, O., Azuma, T. & Obata, Y.

(1997) J. Exp. Med. 185, 953–961.14. Marino, M. W., Dunn, A., Grail, D., Inglese, M., Noguchi, Y.,

Richards, E., Jungbluth, A., Wada, H., Moore, M., Williamson,B., Basu, S. & Old, L. J. (1997) Proc. Natl. Acad. Sci. USA 94,8093–8098.

15. Blin, N. & Stafford, D. W. (1976) Nucleic Acids Res. 3, 2303–2308.16. Chirgwin, J. M., Przybyla, A. E., MacDonald, R. J. & Rutter,

W. J. (1979) Biochemistry 18, 5294–5299.17. Nolan, G. P., Ghosh, S., Liou, H.-C., Tempst, P. & Baltimore, D.

(1991) Cell 64, 961–969.18. Bull, P., Morley, K. L., Hoekstra, M. F., Hunter, T. & Verma,

I. M. (1990) Mol. Cell. Biol. 10, 5473–5485.19. Ryseck, R.-P., Bull, P., Takamiya, M., Bours, V., Siebenlist, U.,

Dobrzanski, P. & Bravo, R. (1992) Mol. Cell. Biol. 12, 674–684.20. Sheehan, K. C. F., Pinckard, J. K., Arthur, C. D., Dehner, L. P.,

Goeddel, D. V. & Schreiber, R. D. (1995) J. Exp. Med. 181, 607–617.

21. Kohchi, C., Noguchi, K., Tanabe, Y., Mizuno, D. & Soma, G.(1994) Int. J. Biochem. 26, 111–119.

22. Pollard, J. W. & Walker, J. M., eds. (1990) Methods in MolecularBiology (Humana, Clifton, NJ), Vol. 5.

23. Pasparakis, M., Alexopoulou, L., Episkopou, V. & Kollias, G.(1996) J. Exp. Med. 184, 1397–1411.

24. Horwitz, B. H., Scott, M. L., Cherry, S. R., Bronson, R. T. &Baltimore, D. (1997) Immunity 6, 765–772.

25. Galanos, C., Freudenberg, M. A. & Reutter, W. (1979) Proc.Natl. Acad. Sci. USA 76, 5939–5943.

26. Pfeffer, K., Matsuyama, T., Kundig, T. M., Wakeham, A., Kishi-hara, K., Shahinian, A., Wiegmann, K., Ohashi, P. S., Kronke, M.& Mak, T. W. (1993) Cell 73, 457–467.

27. Rothe, J., Lesslauer, W., Lotscher, H., Lang, Y., Koebel, P.,Kontgen, F., Althage, A., Zinkernagel, R., Steinmetz, M. &Bluethmann, H. (1993) Nature (London) 364, 798–802.

28. Klimpel, G. R., Shaban, R. & Niesel, D. W. (1990) J. Immunol.145, 711–717.

29. Koff, W. C. & Fann, A. V. (1986) Lymphokine Res. 5, 215–221.30. Wong, G. H. W., Krowka, J. F., Stites, D. P. & Goeddel, D. V.

(1988) J. Immunol. 140, 120–124.31. Morimoto, H., Safrit, J. T. & Bonavida, B. (1991) J. Immunol.

147, 2609–2616.32. Haimovitz-Friedman, A., Cordon-Cardo, C., Bayoumy, S., Gar-

zotto, M., McLoughlin, M., Gallily, R., Edwards, C. K., III,Schuchman, E. H., Fuks, Z. & Kolesnick, R. (1997) J. Exp. Med.186, 1831–1841.

33. Pan, G., Ni, J., Wei, Y.-F., Yu, G.-L., Gentz, R. & Dixit, V. M.(1997) Science 277, 815–818.

34. Sheridan, J. P., Marsters, S. A., Pitti, R. M., Gurney, A.,Skubatch, M., Baldwin, D., Ramakrishnan, L., Gray, C. L., Baker,K., Wood, W. I., et al. (1997) Science 277, 818–821.

35. Mongkolsapaya, J., Cowper, A. E., Xu, X.-N., Morris, G., Mc-Michael, A. J., Bell, J. I. & Screaton, G. R. (1998) J. Immunol.160, 3–6.

36. Chinnaiyan, A. M., O’Rourke, K., Yu, G.-L., Lyons, R. H., Garg,M., Duan, D. R., Xing, L., Gentz, R., Ni, J. & Dixit, V. M. (1996)Science 274, 990–992.

37. Marsters, S. A., Sheridan, J. P., Donahue, C. J., Pitti, R. M., Gray,C. L., Goddard, A. D., Bauer, K. D. & Ashkenazi, A. (1996) Curr.Biol. 6, 1669–1676.

38. McGowan, A. J., Bowie, A. G., O’Neill, L. A. J. & Cotter, T. G.(1998) Exp. Cell. Res. 238, 248–256 .

39. Hannun, Y. A. (1996) Science 274, 1855–1859.40. Kontgen, F., Grumont, R. J., Strasser, A., Metcalf, D., Li, R.,

Tarlinton, D. & Gerondakis, S. (1995) Genes Dev. 9, 1965–1977.41. Wu, M. X., Ao, Z., Prasad, K. V. S., Wu, R. & Schlossman, S. F.

(1998) Science 281, 998–1001.42. Wang, C.-Y., Mayo, M. W., Korneluk, R. G., Goeddel, D. V. &

Baldwin, A. S., Jr. (1998) Science 281, 1680–1683.43. Opipari, A. W., Jr., Hu, H. M., Yabkowitz, R. & Dixit, V. M.

(1992) J. Biol. Chem. 267, 12424–12427.44. Chu, Z.-L., McKinsey, T. A., Liu, L., Gentry, J. J., Malim, M. H.

& Ballard, D. W. (1997) Proc. Natl. Acad. Sci. USA 94, 10057–10062.

45. Song, H. Y., Rothe, M. & Goeddel, D. V. (1996) Proc. Natl. Acad.Sci. USA 93, 6721–6725.

46. Lee, S. Y., Reichlin, A., Santana, A., Sokol, K. A., Nussenzweig,M. C. & Choi, Y. (1997) Immunity 7, 703–713.

47. Yeh, W.-C., Shahinian, A., Speiser, D., Kraunus, J., Billia, F.,Wakeham, A., de la Pompa, J. L., Ferrick, D., Hum, B., Iscove,N., et al. (1997) Immunity 7, 715–725.

48. Roy, N., Deveraux, Q. L., Takahashi, R., Salvesen, G. S. & Reed,J. C. (1997) EMBO J. 16, 6914–6925.

49. De Moissac, D., Mustapha, S., Greenberg, A. H. & Kirshenbaum,L. A. (1998) J. Biol. Chem. 273, 23946–23951.

Immunology: Doi et al. Proc. Natl. Acad. Sci. USA 96 (1999) 2999