a beginner's guide to nf-κb signaling pathways

13
1 Ann. N.Y. Acad. Sci. 1030: 1–13 (2004). © 2004 New York Academy of Sciences. doi: 10.1196/annals.1329.002 A Beginner’s Guide to NF-kB Signaling Pathways SYLVIE DELHALLE, ROMAIN BLASIUS, MARIO DICATO, AND MARC DIEDERICH a Laboratoire de Biologie Moléculaire et Cellulaire du Cancer, Hôpital Kirchberg, Luxembourg ABSTRACT: Nuclear factor kB (NF-kB) belongs to a family of heterodimeric transcription factors that play a key role in inflammatory and stress re- sponses as well as in tumor cell resistance to apoptosis. These effects are due to the NF-kB-dependent transcription of many proinflammatory and antiapo- ptotic genes, whose products ensure various cell responses to environmental conditions. The signal transduction pathways leading to NF-kB activation are well characterized, and the different steps implicated in these pathways involve proteins that could constitute targets for NF-kB inhibition. Several inhibitors aiming to prevent NF-kB activity and thus the transcription of target genes are studied, and a few compounds seem particularly promising. We try here to summarize the advantages that can issue from various studies on NF-kB. KEYWORDS: NF-kB; apoptosis; inflammation; cancer INTRODUCTION The nuclear factor κB (NF-κB) transcription factor is a dimeric complex of vari- ous subunits that belong to the Rel family (p105/50, p100/52, p65 [RelA], RelB, and c-Rel). NF-κB proteins all share a 300-amino acid domain, the Rel homology do- main (RHD), allowing DNA binding, dimerization, and nuclear localization. NF-κB proteins reside normally in the cytoplasm, where they are sequestered by a family of inhibitors of κB (IκB) proteins through interactions between inhibitors’ ankyrin re- peats and the RHD. Upon stimulation of the cells by various activators such as cytokines, lipopolysaccharide, growth factors, stress inducers, chemotherapeutic agents, and other stimuli, IκB is phosphorylated on two serine residues, which trig- gers its ubiquitinylation and degradation by the 26S proteasome. NF-κB is then free to enter the nucleus and to activate the transcription of target genes by binding to its cognate decameric DNA sequence 5-GGGRNNYYCC-3, where R indicates A or G, Y indicates C or T, and N can be any base. NF-κB is involved in the transcription of many proinflammatory as well as antiapoptotic genes and is thus a key player in the progression of carcinogenesis and inflammatory diseases such as rheumatoid arthritis, inflammatory bowel disease, and asthma. 1 Therapies aiming to suppress a Address for correspondence: Marc Diederich, Laboratoire de Biologie Moléculaire et Cellu- laire du Cancer, Hôpital Kirchberg, L-2540 Luxembourg, Luxembourg. Voice: + 352-2468-4040; fax: + 352-2468-4060. e-mail: [email protected]

Upload: sylvie-delhalle

Post on 02-Aug-2016

213 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: A Beginner's Guide to NF-κB Signaling Pathways

1

Ann. N.Y. Acad. Sci. 1030: 1–13 (2004). © 2004 New York Academy of Sciences.doi: 10.1196/annals.1329.002

A Beginner’s Guide to NF-

k

B Signaling Pathways

SYLVIE DELHALLE, ROMAIN BLASIUS, MARIO DICATO,AND MARC DIEDERICH

a

Laboratoire de Biologie Moléculaire et Cellulaire du Cancer, Hôpital Kirchberg, Luxembourg

A

BSTRACT

: Nuclear factor

k

B (NF-

k

B) belongs to a family of heterodimerictranscription factors that play a key role in inflammatory and stress re-sponses as well as in tumor cell resistance to apoptosis. These effects are dueto the NF-

k

B-dependent transcription of many proinflammatory and antiapo-ptotic genes, whose products ensure various cell responses to environmentalconditions. The signal transduction pathways leading to NF-

k

B activation arewell characterized, and the different steps implicated in these pathways involveproteins that could constitute targets for NF-

k

B inhibition. Several inhibitorsaiming to prevent NF-

k

B activity and thus the transcription of target genes arestudied, and a few compounds seem particularly promising. We try here tosummarize the advantages that can issue from various studies on NF-

k

B.

K

EYWORDS

: NF-

k

B; apoptosis; inflammation; cancer

INTRODUCTION

The nuclear factor

κ

B (NF-

κ

B) transcription factor is a dimeric complex of vari-ous subunits that belong to the Rel family (p105/50, p100/52, p65 [RelA], RelB, andc-Rel). NF-

κ

B proteins all share a 300-amino acid domain, the Rel homology do-main (RHD), allowing DNA binding, dimerization, and nuclear localization. NF-

κ

Bproteins reside normally in the cytoplasm, where they are sequestered by a family ofinhibitors of

κ

B (I

κ

B) proteins through interactions between inhibitors’ ankyrin re-peats and the RHD. Upon stimulation of the cells by various activators such ascytokines, lipopolysaccharide, growth factors, stress inducers, chemotherapeuticagents, and other stimuli, I

κ

B is phosphorylated on two serine residues, which trig-gers its ubiquitinylation and degradation by the 26S proteasome. NF-

κ

B is then freeto enter the nucleus and to activate the transcription of target genes by binding to itscognate decameric DNA sequence 5

-GGGRNNYYCC-3

, where R indicates A orG, Y indicates C or T, and N can be any base. NF-

κ

B is involved in the transcriptionof many proinflammatory as well as antiapoptotic genes and is thus a key player inthe progression of carcinogenesis and inflammatory diseases such as rheumatoidarthritis, inflammatory bowel disease, and asthma.

1

Therapies aiming to suppress

a

Address for correspondence: Marc Diederich, Laboratoire de Biologie Moléculaire et Cellu-laire du Cancer, Hôpital Kirchberg, L-2540 Luxembourg, Luxembourg. Voice:

+

352-2468-4040;fax:

+

352-2468-4060.e-mail: [email protected]

Page 2: A Beginner's Guide to NF-κB Signaling Pathways

2 ANNALS NEW YORK ACADEMY OF SCIENCES

NF-

κ

B-induced survival genes are thus an interesting approach to fight and curethese diseases. The purpose of this brief review is to summarize the role of NF-

κ

Bin cell proliferation and the different ways of inhibiting its activity.

NF-

k

B ACTIVATION PATHWAY

Activation of NF-

κ

B through I

κ

B phosphorylation and degradation depends onthe activation of I

κ

B kinases (IKKs). The IKK complex is composed of three sub-units, the catalytic subunits IKK

α

and IKK

β

and the regulatory subunit IKK

γ

(or NEMO,NF-

κ

B essential modulator), and was originally identified as a high-molecular-weightkinase complex able to phosphorylate serines 32 and 36 of I

κ

B

α

.

2

The differentcomponents of this complex have been further identified and characterized,

3–7

andknock-out experiments have determined their role in NF-

κ

B activation. From thesestudies, it appears that IKK

γ

is absolutely required for IKK activity and classical NF-

κ

B activation through the I

κ

B phosphorylation and degradation pathways

8,9

but notfor the alternative pathway leading to p52/RelB dimer translocation.

10

IKK

β

genedisruption in mice leads to death between days 12.5 and 14.5 after gestation frommassive liver apoptosis, whereas IKK

α

gene suppression results in defective kerati-nocyte differentiation.

IKK

β

and IKK

γ

are required for the canonical pathway of I

κ

B phosphorylationand degradation in response to proinflammatory stimuli, whereas IKK

α

is dispens-able for I

κ

B phosphorylation but is involved and necessary for inducible p100 pro-cessing. RelB–p100 forms an inactive dimer, and the IKK

α

-dependent degradationof the C-terminal part of p100 allows nuclear translocation of the formed RelB–p52complex. Activation of this dimer is important for lymphoid organ development andadaptive immune response but not for other NF-

κ

B-dependent functions such asapoptosis inhibition and innate immunity.

11,12

NF-

k

B AND APOPTOSIS

Apoptosis is a kind of cell death characterized by morphological changes and in-volves the activation of a family of cysteine aspartate proteases called caspases.These proteases preexist in unstimulated cells as inactive zymogens and must be ac-tivated by proteolytic cleavage to form a heterotetramer consisting of two large andtwo small subunits. Two major ways of caspase activation are well characterized todate: the death receptors (DR) or extrinsic pathway, and the mitochondrial or intrin-sic pathway.

13

Apoptotic signaling through the extrinsic pathway is triggered by the engagementof members of the tumor necrosis factor (TNF) receptor family (TNFR1, Fas/CD95,DR3, DR4, DR5, and DR6) by their ligands (TNF

α

, lymphotoxin

α

, FasL, Apo3L,and TNF-related apoptosis-inducing ligand), resulting in receptor trimerization andactivation of procaspase-8 and -10. DNA damage or cell stress leading to the activa-tion of p53 or the Bcl-2 family proapoptotic proteins such as Bax and Bak initiate the in-trinsic pathway and induce the mitochondrial release of apoptogenic molecules suchas cytochrome

c

, apoptosis-inducing factor, or Smac/DIABLO (second mitochondria-derived activator of caspases/direct inhibitors of apoptosis [IAP]-binding protein

Page 3: A Beginner's Guide to NF-κB Signaling Pathways

3DELHALLE

et al.

: BEGINNER’S GUIDE TO NF-

k

B SIGNALING PATHWAYS

with low pI). Cytochrome

c

binds to the apoptotic protease-activating factor-1, and thisdimer forms the apoptosome complex with procaspase-9.

14

Interconnection existsbetween the extrinsic and intrinsic pathways, since caspase-8 proteolysis through theextrinsic pathway leads to cleavage and activation of Bid, a proapoptotic member ofthe Bcl-2 family. Once cleaved, truncated Bid incorporates into the mitochondrialmembrane and induces cytochrome c release, which in turns leads to apoptosomeformation and thus the intrinsic apoptotic pathway.

15

Antiapoptotic Role

Transcription factors of the NF-

κ

B family play a role in the regulation of the apo-ptotic program in different cell lines.

16

The first evidence of the NF-

κ

B cytoprotec-tive role came from the analysis of p65

/

mice dying on embryonic day 15 frommassive liver apoptosis, which could be corrected by p65 reexpression.

17,18

NF-

κ

Btranscriptional activity leads to the expression of many antiapoptotic genes. Amongthose gene products, some are directly implicated in repressing and blocking theapoptotic cascade, such as IAPs, c-FLIP (caspase-8/FADD-like-IL-1

β

-convertingenzyme inhibitory protein), or Bcl-2 family proteins.

IAPs are a family of antiapoptotic proteins highly conserved throughout evolu-tion that have the ability to inhibit the activation of various caspases by direct bind-ing and interaction through their baculoviral inhibitory repeat.

19

NF-

κ

B inhibitsTNF-

α

-induced apoptosis through transcriptional activation of IAP1, IAP2, and TNFreceptor-associated factor (TRAF) 1 and TRAF2, thus inhibiting caspase-8 activity.

20

c-FLIP is a proteolytically inactive analog of caspase-8 and competes with pro-caspase-8 and -10 for binding to FADD, thus preventing their activation and subse-quent apoptosis.

21,22

Bcl-2 family proteins such as Bfl-1/A1, Bcl-XL and Bcl-2 prevent apoptosis byinhibiting cytochrome

c

leakage from the mitochondria, thus avoiding apoptosomeformation and the apoptotic cascade.

NF-

κ

B activation also leads to repression of several proapoptotic genes. For exam-ple, two genes coding for the transcription factors Forkhead and Growth Arrest and DNADamage-inducible gene 153/C/EBP-homologous protein

23,24

as well as the proapoptoticgene

bax25,26 are repressed by the activity of NF-κB. NF-κB activation also interfereswith p53 proapoptotic function, since both transcription factors can inhibit each other’stranscriptional activity by competing for a limiting pool of CBP/p300 complexes.26

Proapoptotic Role

NF-κB may have proapoptotic activity in some cell types under certain condi-tions. It regulates the expression of genes whose products can induce apoptosis, suchas the TNF receptor superfamily members DR4, DR5, DR6, Fas, and the FasLligand.27–30 Moreover, NF-κB activity can sometimes be correlated to apoptosis.NF-κB is activated during serum starvation-induced human embryonic kidney cellapoptosis, and its inhibition partially protects cells from this death.31 Glutamate-induced neuronal toxicity is avoided by treatment with the NF-κB inhibitor sodiumsalicylate,32,33 and NF-κB is also involved in the apoptosis occurring during aviandevelopment.34 More recently, NF-κB activity has been shown essential todaunomycin-induced apoptosis.35

Page 4: A Beginner's Guide to NF-κB Signaling Pathways

4 ANNALS NEW YORK ACADEMY OF SCIENCES

NF-kB AND DRUG RESISTANCE

NF-κB activity confers an increased resistance of cancer cells to chemotherapeu-tic drugs. For instance, overexpression of a mutant IκBα protein sensitizes colorectalcancer lineage LOVO to irinotecan (camptothecin-11), a topoisomerase I poison,and induces apoptosis in HT1080 fibrosarcoma cells treated with irinotecan, dauno-rubicin, or ionizing radiation.36,37 NF-κB also regulates genes that modulate cellulardrug uptake or elimination, such as multidrug resistance-1, encoding the P-glycopro-tein membrane transporter38,39 and glutathione S-transferase p1-1.40

NF-kB AND CARCINOGENESIS

Several studies suggest that NF-κB family members are involved in cancer devel-opment.41–43 The first evidence establishing a role for NF-κB in carcinogenesiscame from the fact that c-Rel is the human counterpart of v-Rel, a viral oncogenecausing leukemia and lymphoma in chickens, mice, and transgenic animals.44,45

Moreover, many cancers are associated with gene amplification or chromosomictranslocation of genes such as p65, c-rel, nf-κb1, nf-κb2, bcl-3, iκbα, and iκb«.46–50

Constitutive activity of NF-κB is also observed in various tumors and hematologicalmalignancies. For example, high levels of constitutively active NF-κB are found inHodgkin’s lymphoma Reed–Sternberg cells, and functional inhibition of this tran-scription factor facilitates apoptosis.51–53 This constitutive activation in Reed–Stern-berg cells is linked to mutations of the genes coding for IκBα and IκB« inhibitors,54–56

although ligand-independent activation of CD30 receptors leading to TRAF aggre-gation and NF-κB stimulation has recently been described.57,58 Elevated NF-κBactivities were detected in a variety of tumors, including hormone-dependent and-independent breast cancers as well as chemically induced breast cancers in ro-dents.48,59–62 Several viral oncoproteins, such as Tax from the human T-cell leuke-mia virus type 1, hepatitis virus protein HBx, and latent membrane protein 1 fromEpstein-Barr virus, activate NF-κB during the cell transformation process.63–66

NF-kB AND INFLAMMATION

Numerous proinflammatory mediators, such as interleukin (IL)-1 and TNFα, in-duce nuclear translocation of NF-κB, which in turn activates the transcription ofgene products involved in the inflammatory response, angiogenesis, and cell adhe-sion. NF-κB promotes the expression of cell adhesion molecules (intercellular ad-hesion molecule [ICAM]-1, vascular cell adhesion molecule [VCAM]-1, E selectin,tenascin C), vascular endothelial growth factor, and matrix metalloprotease-2 and-9, which are responsible for degradation of the extracellular matrix.67 This transcrip-tion factor is also involved in the transcription of genes coding for enzymes such asinducible nitric oxide synthase, cyclooxygenase (COX)-2, 5- and 12-lipooxygenase,chemokines, and cytokines (IL-1 and TNFα). COX-2 is upregulated in aggressivecolorectal cancers and has been found to promote angiogenesis. Mediators such asnitric oxide, prostaglandins, IL-1, and TNFα are involved in the regulation of bloodpressure, platelet aggregation, and body temperature. Moreover, NF-κB is also impor-tant in the transcription of genes coding for several acute-phase proteins.

Page 5: A Beginner's Guide to NF-κB Signaling Pathways

5DELHALLE et al.: BEGINNER’S GUIDE TO NF- kB SIGNALING PATHWAYS

NF-κB constitutive activity seems to play a role in chronic inflammatory diseasessuch as inflammatory bowel disease, Crohn’s disease, ulcerative colitis, rheumatoid ar-thritis, and asthma.1 NF-κB activation in chronic inflammation may participate in tumorinitiation, since the antiapoptotic genes activated by this transcription factor may contrib-ute to the survival of altered cells that would otherwise be committed to apoptosis andthereby allow the formation of precancerous lesions. For instance, Helicobacter pyloriinfections or constitutive activation of the IL-1 gene, both triggering NF-κB activation,are risk factors for the development of gastric cancer.68,69 Constitutive NF-κB activationmay also be responsible for the development of colitis-associated cancer in patients suf-fering from inflammatory bowel disease, ulcerative colitis, or Crohn’s disease.70–72

NF-kB INHIBITION

The identification of NF-κB as a key player in apoptosis resistance and inflam-matory diseases underscores the potential benefits of its inhibition. Several steps ofthe NF-κB signal transduction pathway, such as IKK activation, IκB phosphoryla-tion and degradation, NF-κB nuclear translocation, and transcriptional activity, canbe targeted by various inhibitors (FIG. 1).

IKK Inhibitors

The IKK kinases constitute the apical complex responsible for NF-κB activation,and the effects of many inhibitory compounds on this complex have thus been studied.

Nonsteroidal Anti-Inflammatory Drugs

Anti-inflammatory agents such as acetylsalicylic acid and sodium salicylate exerttheir effect at least partially through NF-κB inhibition, mediated by competitive in-hibition of the ATP-binding site of IKKβ.73,74 Sulfasalazine has also been shown toinhibit NF-κB through binding to IKKβ and repressing its catalytic activity.75 Sulin-dac and its metabolites, sulindac sulfide and sulindac sulfone, also interfere with theNF-κB activation pathway by inhibiting IKK activity.76

Cyclopentenone Prostaglandins

Cyclopentenone prostaglandins such as prostaglandin A1 and 15-deoxy-∆12-14-PGJ2 are irreversible inhibitors of IKKβ by covalent modifications of cysteine resi-dues.77 Moreover, covalent changes in NF-κB subunits leading to binding impair-ment have also been observed with the same compounds.78,79

Natural Compounds

Flavonoids are naturally occurring phenolic compounds present in plants that ex-hibit anti-inflammatory and cancer-preventing properties. Different studies haveshown that flavonoids exert some of these effects by inhibiting NF-κB and subse-quent transcription of proinflammatory or antiapoptotic genes. For example, curcumin(a yellow extract from turmeric), capsaicin (extract from red pepper), resveratrol (redwine polyphenol), and green tea polyphenols are potent inhibitors of NF-κB activityby preventing IKK activity.80–87

Page 6: A Beginner's Guide to NF-κB Signaling Pathways

6 ANNALS NEW YORK ACADEMY OF SCIENCES

Glucocorticoids

Glucocorticoids (GCs) are widely used anti-inflammatory compounds known toinhibit proinflammatory gene products such as COX-2 and TNF. GCs exert their ef-fects by inducing the binding of the glucocorticoid receptor (GR) family of ligand-dependent transcription factors to their DNA glucocorticoid response elements. GRsinduce the synthesis of IκBα, and this newly synthesized inhibitor is able to complexNF-κB and thus prevent its activity.88,89 However, this mechanism is not the only

FIGURE 1. Different approaches to inhibit NF-κB activation according to the molecu-lar target of the pathway. Several steps involving different molecular intermediates are pos-sible targets for NF-κB inhibitors. Peptides such as NEMO binding domain (NBD) peptideprevent IKK complex formation, whereas cyclopentenone prostaglandins (CyPGs), nonste-roidal anti-inflammatory drugs (NSAIDs), and natural compounds inhibit IKK activation.Proteasome inhibitors block IκB degradation, and glucocorticoids induce its neosynthesis.Finally, nuclear localization sequence (NLS)-based peptides inhibit NF-κB nuclear translo-cation, whereas antisense or decoy oligonucleotides prevent the transcription of target genes.

Page 7: A Beginner's Guide to NF-κB Signaling Pathways

7DELHALLE et al.: BEGINNER’S GUIDE TO NF- kB SIGNALING PATHWAYS

one by which GRs repress NF-κB-dependent transactivation. GCs are also involvedin the inhibition of RNA polymerase II phosphorylation,90 the inhibition of p65-mediated histone acetyltransferase activity,91 and the competition with p65 for thecatalytic subunit of protein kinase A.92

Proteasome Inhibitors

The 26S proteasome is a large (1,500–2,000 kDa) multicatalytic enzyme complexpresent in the nucleus and cytoplasm of eukaryotic cells, whose essential function isto degrade proteins.93,94 It mediates the degradation of several key cell cycle regu-lators and apoptosis-inducing factors such as p53, Bcl-2, p21, p27, and cyclins A, B,D, and E.95–101

The 26S proteasome regulates NF-κB activation in that it is responsible for IκBαdegradation after its phosphorylation and ubiquitinylation. Therapies aiming to sup-press proteasome activity are thus worthy of interest, as they attempt to inhibit theNF-κB-driven expression of antiapoptotic and proinflammatory gene products. Theactivities of several proteasome inhibitors, such as peptide aldehydes (MG132and PSI),102 lactacystine,103 peptide boronic acids,104 vinyl sulfone tripeptides,105,106

and natural products such as eponemycin and epoxomycin,107,108 have thus been thor-oughly examined. Among those substances, the most promising are peptide boronicacids, with the compound bortezomib (Velcade, PS-341) presenting death-inducingproperties in multiple myeloma, lung cancer,109 human prostate cancer, and squa-mous cell carcinoma xenografts in mice as well as in grafted mouse mammarytumors.104,110 Antiproliferative and antiproteolytic properties of Bortezomib wereassessed against 60 cell lines derived from various human tumors in a National Can-cer Institute study.104 Moreover, Bortezomib overcomes in vitro multicellular drugresistance and sensitizes cancer cells to tumoricidal drugs.111,112 Bortezomib hassuccessfully entered clinical trials and seems efficient in solid tumors as well as inhematologic malignancies.

Mesalamine

Mesalamine is an aminosalicylate compound inhibiting IL-1-induced NF-κBtranscription without modifying either IκBα degradation or NF-κB nuclear translo-cation and DNA binding. In this case, prevention of gene expression is due to theinhibition of IL-1-driven p65 phosphorylation.113

Oligonucleotides

Two types of oligonucleotides have been studied to prevent NF-κB activation: (a)antisense oligonucleotides directed against the p50 subunit reduce IgM and IgG syn-thesis in B cells,114 whereas use of the same method with p65 leads to in vitro inhi-bition of cell adhesion molecules or in vivo blockade of tumor growth in nude miceand prolongs allograft and xenograft survival;115–117 (b) decoy oligonucleotidesbinding to transcription factors and thereby preventing promoter activation impairthe expression of several NF-κB target genes such as IL-1, IL-6, ICAM-1, andVCAM.118–121 This approach leads to in vivo diminution of inflammatory signs in arat model of rheumatoid arthritis.122

Page 8: A Beginner's Guide to NF-κB Signaling Pathways

8 ANNALS NEW YORK ACADEMY OF SCIENCES

Peptides

Cell-penetrating peptides interfering with protein–protein interactions also leadto NF-κB function impairment. Peptides constructed with the NF-κB nuclear local-ization sequence (NLS), such as SN50, inhibit NF-κB nuclear translocation trig-gered by lipopolysaccharide and TNF-α.123,124 However, this effect does not seemto be specific to NF-κB, because a peptide based on p50 NLS impairs the nuclearimport of AP-1, NFAT, and STAT-1, whose NLS are different from the p50 NLS.125

Development of a peptide containing the IKKγ-binding motif (NEMO bindingdomain [NBD]) of IKKα and IKKβ has also been reported, and this peptide inhibitsTNF-α-induced NF-κB activation.126 This peptide has been tested in various animalmodels of inflammation and seems to be a potent anti-inflammatory agent.

CONCLUSIONS

NF-κB and its target genes are potent antiapoptotic and proinflammatory media-tors and are thus involved in various pathologies such as chronic inflammatory dis-eases and cancers. Inhibition of NF-κB activation is a very promising therapeuticapproach to fight those pathologies. Some inhibitors are already potent NF-κB mod-ulators in clinical use, and others remain perfectible benchtop compounds. Never-theless, a better understanding of the different steps leading to NF-κB transcriptionalactivation in different cancer types will permit increased accuracy in the synthesis ofvarious inhibitors, allowing precise targeting of the chosen step. So, the best is yetto come.

ACKNOWLEDGMENTS

Research at the laboratory of M.D. is supported by the Fondation de RechercheCancer et Sang, by the Recherches Scientifiques Luxembourg association, byTélévie grants, and by the Een Häerz fir kriibskrank Kanner association.

REFERENCES

1. BARNES, P.J. & M. KARIN. 1997. Nuclear factor-κB: a pivotal transcription factor inchronic inflammatory diseases. N. Engl. J. Med. 336: 1066–1071.

2. CHEN, Z.J., L. PARENT & T. MANIATIS. 1996. Site-specific phosphorylation of IκBα by anovel ubiquitination-dependent protein kinase activity. Cell 84: 853–862.

3. DI DONATO, J.A., et al. 1997. A cytokine-responsive IκB kinase that activates the tran-scription factor NF-κB. Nature 388: 548–554.

4. MERCURIO, F., et al. 1997. IKK-1 and IKK-2: cytokine-activated IκB kinases essentialfor NF-κB activation. Science 278: 860–866.

5. ZANDI , E., et al. 1997. The IκB kinase complex (IKK) contains two kinase subunits,IKK α and IKKβ, necessary for IκB phosphorylation and NF-κB activation. Cell 91:243–252.

6. WORONICZ, J.D., et al. 1997. IκB kinase-β: NF-κB activation and complex formationwith IκB kinase-α and NIK. Science 278: 866–869.

7. REGNIER, C.H., et al. 1997. Identification and characterization of an IκB kinase. Cell 90:373–383.

Page 9: A Beginner's Guide to NF-κB Signaling Pathways

9DELHALLE et al.: BEGINNER’S GUIDE TO NF- kB SIGNALING PATHWAYS

8. ROTHWARF, D.M., et al. 1998. IKK-γ is an essential regulatory subunit of the IκBkinase complex. Nature 395: 297–300.

9. MAKRIS, C., et al. 2000. Female mice heterozygous for IKK γ/NEMO deficienciesdevelop a dermatopathy similar to the human X-linked disorder incontinentia pig-menti. Mol. Cell 5: 969–979.

10. DEJARDIN, E., et al. 2002. The lymphotoxin-β receptor induces different patterns ofgene expression via two NF-κB pathways. Immunity 17: 525–535.

11. SENFTLEBEN, U., et al. 2001. Activation by IKKα of a second, evolutionarily con-served, NF-κB signaling pathway. Science 293: 1495–1499.

12. GHOSH, S. & M. KARIN. 2002. Missing pieces in the NF-κB puzzle. Cell 109(Suppl.):S81–S96.

13. EARNSHAW, W.C., L.M. MARTINS & S.H. KAUFMANN. 1999. Mammalian caspases:structure, activation, substrates, and functions during apoptosis. Annu. Rev. Bio-chem. 68: 383–424.

14. STRASSER, A., L. O’CONNOR & V.M. DIXIT . 2000. Apoptosis signaling. Annu. Rev. Bio-chem. 69: 217–245.

15. GROSS, A., J.M. MCDONNELL & S.J. KORSMEYER. 1999. BCL-2 family members andthe mitochondria in apoptosis. Genes Dev. 13: 1899–1911.

16. BARKETT, M. & T.D. GILMORE. 1999. Control of apoptosis by Rel/NF-κB transcriptionfactors. Oncogene 18: 6910–6924.

17. BEG, A.A., et al. 1995. Embryonic lethality and liver degeneration in mice lacking theRelA component of NF-κB. Nature 376: 167–170.

18. BEG, A.A. & D. BALTIMORE. 1996. An essential role for NF-κB in preventing TNF-α-induced cell death. Science 274: 782–784.

19. DEVERAUX, Q.L. & J.C. REED. 1999. IAP family proteins: suppressors of apoptosis.Genes Dev. 13: 239–252.

20. WANG, C.Y., et al. 1998. NF-κB antiapoptosis: induction of TRAF1 and TRAF2and c-IAP1 and c-IAP2 to suppress caspase-8 activation. Science 281: 1680–1683.

21. IRMLER, M., et al. 1997. Inhibition of death receptor signals by cellular FLIP. Nature388: 190–195.

22. GOLTSEV, Y.V., et al. 1997. CASH, a novel caspase homologue with death effectordomains. J. Biol. Chem. 272: 19641–19644.

23. TRAN, H., et al. 2003. The many forks in FOXO’s road. Sci. STKE 2003: RE5.24. NOZAKI , S., G.W. SLEDGE, Jr. & H. NAKSHATRI. 2001. Repression of GADD153/CHOP

by NF-κB: a possible cellular defense against endoplasmic reticulum stress-inducedcell death. Oncogene 20: 2178–2185.

25. BENTIRES-ALJ, M., et al. 2001. Inhibition of the NF-κB transcription factor increasesBax expression in cancer cell lines. Oncogene 20: 2805–2813.

26. WEBSTER, G.A. & N.D. PERKINS. 1999. Transcriptional cross talk between NF-κB andp53. Mol. Cell. Biol. 19: 3485–3495.

27. MATSUI, K., et al. 1998. Identification of two NF-κB sites in mouse CD95 ligand (Fasligand) promoter: functional analysis in T cell hybridoma. J. Immunol. 161: 3469–3473.

28. RAVI , R., et al. 2001. Regulation of death receptor expression and TRAIL/Apo2L-induced apoptosis by NF-κB. Nat. Cell Biol. 3: 409–416.

29. ZHENG, Y., et al. 2001. NF-κB RelA (p65) is essential for TNF-α-induced fas expres-sion but dispensable for both TCR-induced expression and activation-induced celldeath. J. Immunol. 166: 4949–4957.

30. KASOF, G.M., et al. 2001. Tumor necrosis factor-α induces the expression of DR6, amember of the TNF receptor family, through activation of NF-κB. Oncogene 20:7965–7975.

31. GRIMM, S., et al. 1996. Bcl-2 down-regulates the activity of transcription factor NF-κBinduced upon apoptosis. J. Cell Biol. 134: 13–23.

32. GRILLI , M., et al. 1996. Neuroprotection by aspirin and sodium salicylate throughblockade of NF-κB activation. Science 274: 1383–1385.

33. LIPTON, S.A. 1997. Janus faces of NF-κB: neurodestruction versus neuroprotection.Nat. Med. 3: 20–22.

Page 10: A Beginner's Guide to NF-κB Signaling Pathways

10 ANNALS NEW YORK ACADEMY OF SCIENCES

34. ABBADIE, C., et al. 1993. High levels of c-rel expression are associated with pro-grammed cell death in the developing avian embryo and in bone marrow cells invitro. Cell 75: 899–912.

35. ASHIKAWA, K., et al. 2004. Evidence that activation of nuclear factor-κB is essentialfor the cytotoxic effects of doxorubicin and its analogues. Biochem. Pharmacol. 67:353–364.

36. WANG, C.Y., M.W. MAYO & A.S. BALDWIN , Jr. 1996. TNF- and cancer therapy-inducedapoptosis: potentiation by inhibition of NF-κB. Science 274: 784–787.

37. WANG, C.Y., et al. 1999. Control of inducible chemoresistance: enhanced anti-tumortherapy through increased apoptosis by inhibition of NF-κB. Nat. Med. 5: 412–417.

38. ZHOU, G. & M.T. KUO. 1997. NF-κB-mediated induction of mdr1b expression by insu-lin in rat hepatoma cells. J. Biol. Chem. 272: 15174–15183.

39. BENTIRES-ALJ, M., et al. 2003. NF-κB transcription factor induces drug resistancethrough MDR1 expression in cancer cells. Oncogene 22: 90–97.

40. MORCEAU, F., et al. 2004. Regulation of glutathione S-transferase P1-1 gene expres-sion by NF-κB in tumor necrosis factor α-treated K562 leukemia cells. Biochem.Pharmacol. 67: 1227–1238.

41. SYLLA , B.S. & H.M. TEMIN. 1986. Activation of oncogenicity of the c-rel proto-onco-gene. Mol. Cell. Biol. 6: 4709–4716.

42. MOORE, B.E. & H.R. BOSE, Jr. 1988. Expression of the v-rel oncogene in reticuloen-dotheliosis virus-transformed fibroblasts. Virology 162: 377–387.

43. GILMORE, T.D., et al. 1996. Rel/NF-κB/IκB proteins and cancer. Oncogene 13: 1367–1378.

44. CAPOBIANCO, A.J., D.L. SIMMONS & T.D. GILMORE. 1990. Cloning and expression ofa chicken c-rel cDNA: unlike p59v-rel, p68c-rel is a cytoplasmic protein in chickenembryo fibroblasts. Oncogene 5: 257–265.

45. GILMORE, T.D. 2003. The Re1/NF-κB/IκB signal transduction pathway and cancer.Cancer Treat. Res. 115: 241–265.

46. NERI, A., et al. 1991. B cell lymphoma-associated chromosomal translocation involvescandidate oncogene lyt-10, homologous to NF-κB p50. Cell 67: 1075–1087.

47. LU, D., et al. 1991. Alterations at the rel locus in human lymphoma. Oncogene 6:1235–1241.

48. DEJARDIN, E., et al. 1995. Highly-expressed p100/p52 (NFKB2) sequesters otherNF-κB-related proteins in the cytoplasm of human breast cancer cells. Oncogene 11:1835–1841.

49. KITAJIMA , I., et al. 1992. Ablation of transplanted HTLV-I Tax-transformed tumors inmice by antisense inhibition of NF-κB. Science 258: 1792–1795.

50. OHNO, H., G. TAKIMOTO & T.W. MCKEITHAN. 1990. The candidate proto-oncogene bcl-3is related to genes implicated in cell lineage determination and cell cycle control.Cell 60: 991–997.

51. BARGOU, R.C., et al. 1997. Constitutive nuclear factor-κB-RelA activation is requiredfor proliferation and survival of Hodgkin’s disease tumor cells. J. Clin. Invest. 100:2961–2969.

52. Hinz, M., et al. 2001. Constitutive NF-κB maintains high expression of a characteristicgene network, including CD40, CD86, and a set of antiapoptotic genes in Hodgkin/Reed-Sternberg cells. Blood 97: 2798–2807.

53. IZBAN, K.F., et al. 2001. Characterization of NF-κB expression in Hodgkin’s disease:inhibition of constitutively expressed NF-κB results in spontaneous caspase-independent apoptosis in Hodgkin and Reed-Sternberg cells. Mod. Pathol. 14:297–310.

54. CABANNES, E., et al. 1999. Mutations in the IkBa gene in Hodgkin’s disease suggest atumour suppressor role for IκBα. Oncogene 18: 3063–3070.

55. KRAPPMANN, D., et al. 1999. Molecular mechanisms of constitutive NF-κB/Rel activa-tion in Hodgkin/Reed-Sternberg cells. Oncogene 18: 943–953.

56. WOOD, K.M., M. ROFF & R.T. HAY. 1998. Defective IκBα in Hodgkin cell lines withconstitutively active NF-κB. Oncogene 16: 2131–2139.

57. HORIE, R., et al. 2002. Cytoplasmic aggregation of TRAF2 and TRAF5 proteins in theHodgkin-Reed-Sternberg cells. Am. J. Pathol. 160: 1647–1654.

Page 11: A Beginner's Guide to NF-κB Signaling Pathways

11DELHALLE et al.: BEGINNER’S GUIDE TO NF- kB SIGNALING PATHWAYS

58. HORIE, R., et al. 2002. Ligand-independent signaling by overexpressed CD30 drivesNF-κB activation in Hodgkin-Reed-Sternberg cells. Oncogene 21: 2493–2503.

59. COGSWELL, P.C., et al. 2000. Selective activation of NF-κB subunits in human breastcancer: potential roles for NF-κB2/p52 and for Bcl-3. Oncogene 19: 1123–1131.

60. NAKSHATRI, H., et al. 1997. Constitutive activation of NF-κB during progression ofbreast cancer to hormone-independent growth. Mol. Cell. Biol. 17: 3629–3639.

61. RAYET, B. & C. GELINAS. 1999. Aberrant rel/nfkb genes and activity in human cancer.Oncogene 18: 6938–6947.

62. SOVAK, M.A., et al. 1997. Aberrant nuclear factor-κB/Rel expression and the patho-genesis of breast cancer. J. Clin. Invest. 100: 2952–2960.

63. YAMAOKA , S., et al. 1996. Constitutive activation of NF-κ B is essential for transfor-mation of rat fibroblasts by the human T-cell leukemia virus type I Tax protein.EMBO J. 15: 873–887.

64. XIAO, G., E.W. HARHAJ & S.C. SUN. 2000. Domain-specific interaction with the I κ Bkinase (IKK) regulatory subunit IKK γ is an essential step in tax-mediated activationof IKK. J. Biol. Chem. 275: 34060–34067.

65. KNECHT, H., et al. 2001. The role of Epstein-Barr virus in neoplastic transformation.Oncology 60: 289–302.

66. KOWALIK , T.F., et al. 1993. Multiple mechanisms are implicated in the regulation ofNF-κ B activity during human cytomegalovirus infection. Proc. Natl. Acad. Sci. USA90: 1107–1111.

67. PAHL, H.L. 1999. Activators and target genes of Rel/NF-κB transcription factors.Oncogene 18: 6853–6866.

68. EL-OMAR, E.M., et al. 2000. Interleukin-1 polymorphisms associated with increasedrisk of gastric cancer. Nature 404: 398–402.

69. PEEK, R.M., Jr. & M.J. BLASER. 2002. Helicobacter pylori and gastrointestinal tractadenocarcinomas. Nat. Rev. Cancer 2: 28–37.

70. NEURATH, M.F., et al. 1996. Local administration of antisense phosphorothioate oligo-nucleotides to the p65 subunit of NF-κ B abrogates established experimental colitisin mice. Nat. Med. 2: 998–1004.

71. ROGLER, G., et al. 1998. Nuclear factor κB is activated in macrophages and epithelialcells of inflamed intestinal mucosa. Gastroenterology 115: 357–369.

72. EKBOM, A., et al. 1990. Ulcerative colitis and colorectal cancer: a population-basedstudy. N. Engl. J. Med. 323: 1228–1233.

73. KOPP, E. & S. GHOSH. 1994. Inhibition of NF-κ B by sodium salicylate and aspirin.Science 265: 956–959.

74. YIN, M.J., Y. YAMAMOTO & R.B. GAYNOR. 1998. The anti-inflammatory agents aspirinand salicylate inhibit the activity of I(κ)B kinase-β. Nature 396: 77–80.

75. WAHL, C., et al. 1998. Sulfasalazine: a potent and specific inhibitor of nuclear factor κB. J. Clin. Invest. 101: 1163–1174.

76. YAMAMOTO, Y., et al. 1999. Sulindac inhibits activation of the NF-κB pathway. J. Biol.Chem. 274: 27307–27314.

77. ROSSI, A., et al. 2000. Anti-inflammatory cyclopentenone prostaglandins are directinhibitors of IκB kinase. Nature 403: 103–108.

78. STRAUS, D.S., et al. 2000. 15-Deoxy-delta 12,14-prostaglandin J2 inhibits multiplesteps in the NF-κ B signaling pathway. Proc. Natl. Acad. Sci. USA 97: 4844–4849.

79. CERNUDA-MOROLLON, E., et al. 2001. 15-Deoxy-delta 12,14-prostaglandin J2 inhibi-tion of NF-κB-DNA binding through covalent modification of the p50 subunit. J.Biol. Chem. 276: 35530–35536.

80. HOLMES-MCNARY, M. & A.S. BALDWIN , Jr. 2000. Chemopreventive properties of trans-resveratrol are associated with inhibition of activation of the IκB kinase. Cancer Res.60: 3477–3483.

81. YANG, F., et al. 2001. The green tea polyphenol (−)-epigallocatechin-3-gallate blocksnuclear factor-κ B activation by inhibiting I κ B kinase activity in the intestinal epi-thelial cell line IEC-6. Mol. Pharmacol. 60: 528–533.

82. PAN, M.H., et al. 2000. Suppression of lipopolysaccharide-induced nuclear factor-κBactivity by theaflavin-3,3′-digallate from black tea and other polyphenols through

Page 12: A Beginner's Guide to NF-κB Signaling Pathways

12 ANNALS NEW YORK ACADEMY OF SCIENCES

down-regulation of IκB kinase activity in macrophages. Biochem. Pharmacol. 59:357–367.

83. JOBIN, C., et al. 1999. Curcumin blocks cytokine-mediated NF-κ B activation andproinflammatory gene expression by inhibiting inhibitory factor I-κ B kinase activ-ity. J. Immunol. 163: 3474–3483.

84. PLUMMER, S.M., et al. 1999. Inhibition of cyclo-oxygenase 2 expression in colon cellsby the chemopreventive agent curcumin involves inhibition of NF-κB activation viathe NIK/IKK signalling complex. Oncogene 18: 6013–6020.

85. KUMAR, A., et al. 1998. Curcumin (diferuloylmethane) inhibition of tumor necrosisfactor (TNF)-mediated adhesion of monocytes to endothelial cells by suppressionof cell surface expression of adhesion molecules and of nuclear factor-κB activa-tion. Biochem. Pharmacol. 55: 775–783.

86. CHAN, M.M. 1995. Inhibition of tumor necrosis factor by curcumin, a phytochemical.Biochem. Pharmacol. 49: 1551–1556.

87. DUVOIX, A., et al. 2003. Induction of apoptosis by curcumin: mediation by glu-tathione S-transferase P1-1 inhibition. Biochem. Pharmacol. 66: 1475–1483.

88. AUPHAN, N., et al. 1995. Immunosuppression by glucocorticoids: inhibition of NF-κB activity through induction of I κ B synthesis. Science 270: 286–290.

89. SCHEINMAN, R.I., et al. 1995. Role of transcriptional activation of I κ B α in media-tion of immunosuppression by glucocorticoids. Science 270: 283–286.

90. NISSEN, R.M. & K.R. YAMAMOTO. 2000. The glucocorticoid receptor inhibits NFκBby interfering with serine-2 phosphorylation of the RNA polymerase II carboxy-terminal domain. Genes Dev. 14: 2314–2329.

91. ITO, K., et al. 2001. p65-activated histone acetyltransferase activity is repressed byglucocorticoids: mifepristone fails to recruit HDAC2 to the p65-HAT complex. J.Biol. Chem. 276: 30208–30215.

92. DOUCAS, V., et al. 2000. Cytoplasmic catalytic subunit of protein kinase A mediatescross-repression by NF-κ B and the glucocorticoid receptor. Proc. Natl. Acad. Sci.USA 97: 11893–11898.

93. CIECHANOVER, A. 1994. The ubiquitin-proteasome proteolytic pathway. Cell 79: 13–21.

94. BROOKS, P., et al. 2000. Subcellular localization of proteasomes and their regulatorycomplexes in mammalian cells. Biochem. J. 346(Pt 1): 155–161.

95. MAKI , C.G., J.M. HUIBREGTSE & P.M. HOWLEY. 1996. In vivo ubiquitination and pro-teasome-mediated degradation of p53(1). Cancer Res. 56: 2649–2654.

96. CHADEBECH, P., et al. 1999. Phosphorylation and proteasome-dependent degradationof Bcl-2 in mitotic-arrested cells after microtubule damage. Biochem. Biophys.Res. Commun. 262: 823–827.

97. CAYROL, C. & B. DUCOMMUN. 1998. Interaction with cyclin-dependent kinases andPCNA modulates proteasome-dependent degradation of p21. Oncogene 17: 2437–2444.

98. CLURMAN, B.E., et al. 1996. Turnover of cyclin E by the ubiquitin-proteasome path-way is regulated by cdk2 binding and cyclin phosphorylation. Genes Dev. 10:1979–1990.

99. DIEHL, J.A., F. ZINDY & C.J. SHERR. 1997. Inhibition of cyclin D1 phosphorylation onthreonine-286 prevents its rapid degradation via the ubiquitin-proteasome pathway.Genes Dev. 11: 957–972.

100. PAGANO, M., et al. 1995. Role of the ubiquitin-proteasome pathway in regulatingabundance of the cyclin-dependent kinase inhibitor p27. Science 269: 682–685.

101. SUDAKIN , V., et al. 1995. The cyclosome, a large complex containing cyclin-selectiveubiquitin ligase activity, targets cyclins for destruction at the end of mitosis. Mol.Biol. Cell 6: 185–197.

102. ADAMS, J., et al. 1998. Potent and selective inhibitors of the proteasome: dipeptidylboronic acids. Bioorg. Med. Chem. Lett. 8: 333–338.

103. DICK, T.P., et al. 1998. Contribution of proteasomal β-subunits to the cleavage of pep-tide substrates analyzed with yeast mutants. J. Biol. Chem. 273: 25637–25646.

104. ADAMS, J., et al. 1999. Proteasome inhibitors: a novel class of potent and effectiveantitumor agents. Cancer Res. 59: 2615–2622.

Page 13: A Beginner's Guide to NF-κB Signaling Pathways

13DELHALLE et al.: BEGINNER’S GUIDE TO NF- kB SIGNALING PATHWAYS

105. ADAMS, J., V.J. PALOMBELLA & P.J. ELLIOTT. 2000. Proteasome inhibition: a newstrategy in cancer treatment. Invest. New Drugs 18: 109–121.

106. LEE, D.H. & A.L. GOLDBERG. 1998. Proteasome inhibitors: valuable new tools for cellbiologists. Trends Cell Biol. 8: 397–403.

107. MENG, L., et al. 1999. Eponemycin exerts its antitumor effect through the inhibitionof proteasome function. Cancer Res. 59: 2798–2801.

108. MENG, L., et al. 1999. Epoxomicin, a potent and selective proteasome inhibitor, exhibitsin vivo antiinflammatory activity. Proc. Natl. Acad. Sci. USA 96: 10403–10408.

109. MACK, P.C., et al. 2003. Integration of the proteasome inhibitor PS-341 (Velcade)into the therapeutic approach to lung cancer. Lung Cancer 41(Suppl. 1): S89–S96.

110. SUNWOO, J.B., et al. 2001. Novel proteasome inhibitor PS-341 inhibits activation ofnuclear factor-κ B, cell survival, tumor growth, and angiogenesis in squamous cellcarcinoma. Clin. Cancer Res. 7: 1419–1428.

111. BOLD, R.J., S. VIRUDACHALAM & D.J. MCCONKEY. 2001. Chemosensitization of pan-creatic cancer by inhibition of the 26S proteasome. J. Surg. Res. 100: 11–17.

112. CUSACK, J.C., Jr., et al. 2001. Enhanced chemosensitivity to CPT-11 with proteasomeinhibitor PS-341: implications for systemic nuclear factor-κB inhibition. CancerRes. 61: 3535–3540.

113. EGAN, L.J., et al. 1999. Inhibition of interleukin-1-stimulated NF-κB RelA/p65 phos-phorylation by mesalamine is accompanied by decreased transcriptional activity. J.Biol. Chem. 274: 26448–26453.

114. KHALED, A.R., et al. 1997. Inhibition of the p50 (NKκB1) subunit of NF-κB by phos-phorothioate-modified antisense oligodeoxynucleotides reduces NF-κB expressionand immunoglobulin synthesis in murine B cells. Clin. Immunol. Immunopathol.83: 254–263.

115. VOISARD, R., et al. 2001. Different effects of antisense RelA p65 and NF-κB1 p50oligonucleotides on the nuclear factor-κB mediated expression of ICAM-1 inhuman coronary endothelial and smooth muscle cells. BMC Mol. Biol. 2: 7.

116. WU, G.S., et al. 2001. Antisense NF-κB p65 prolongs experimental allo- and xeno-graft survival. Transplant. Proc. 33: 360.

117. HIGGINS, K.A., et al. 1993. Antisense inhibition of the p65 subunit of NF-κ B blockstumorigenicity and causes tumor regression. Proc. Natl. Acad. Sci. USA 90: 9901–9905.

118. TOMITA, N., et al. 2000. Transcription factor decoy for NFκB inhibits TNF-α-inducedcytokine and adhesion molecule expression in vivo. Gene Ther. 7: 1326–1332.

119. TOMITA, N., et al. 1998. Transcription factor decoy for nuclear factor-κB inhibitstumor necrosis factor-α-induced expression of interleukin-6 and intracellular adhe-sion molecule-1 in endothelial cells. J. Hypertens. 16: 993–1000.

120. TOMITA, T., et al. 2000. Transcription factor decoy for NFκB inhibits cytokine andadhesion molecule expressions in synovial cells derived from rheumatoid arthritis.Rheumatology (Oxford) 39: 749–757.

121. MORISHITA, R., et al. 1997. In vivo transfection of cis element “decoy” against nuclearfactor-κB binding site prevents myocardial infarction. Nat. Med. 3: 894–899.

122. TOMITA, T., et al. 1999. Suppressed severity of collagen-induced arthritis by in vivotransfection of nuclear factor κB decoy oligodeoxynucleotides as a gene therapy.Arthritis Rheum. 42: 2532–2542.

123. LIN, Y.Z., et al. 1995. Inhibition of nuclear translocation of transcription factor NF-κB by a synthetic peptide containing a cell membrane-permeable motif and nuclearlocalization sequence. J. Biol. Chem. 270: 14255–14258.

124. ABATE, A., S. OBERLE & H. SCHRODER. 1998. Lipopolysaccharide-induced expressionof cyclooxygenase-2 in mouse macrophages is inhibited by chloromethylketonesand a direct inhibitor of NF-κ B translocation. Prostaglandins Other Lipid Mediat.56: 277–290.

125. TORGERSON, T.R., et al. 1998. Regulation of NF-κ B, AP-1, NFAT, and STAT1 nuclearimport in T lymphocytes by noninvasive delivery of peptide carrying the nuclearlocalization sequence of NF-κ B p50. J. Immunol. 161: 6084–6092.

126. MAY, M.J., et al. 2000. Selective inhibition of NF-κB activation by a peptide that blocksthe interaction of NEMO with the IκB kinase complex. Science 289: 1550–1554.