10
RAG2–/–, IkB-a–/– Chimeras Display a Psoriasiform Skin Disease Chih-Li Chen,* Fiona E. Yull,² Nancy Cardwell,²‡ Nagendra Singh,* William David Strayhorn,² Lillian B. Nanney,²‡ and Lawrence D. Kerr*²§¶ Departments of *Microbiology and Immunology, ²Cell Biology, and Plastic Surgery, §Vanderbilt University School of Medicine, Vanderbilt-Ingram Cancer Center, Nashville, Tennessee, U.S.A.; The National Institutes of Health, Transplantation and Immunogenetics Section, Division of Allergy, Immunology, and Infectious Disease, Bethesda, Maryland, U.S.A. Nuclear factor-kB, a ubiquitous transcription factor involved in inflammatory and immune responses, is inappropriately activated in several immuno-related diseases, such as allograft rejection, or bronchial asthma. As nuclear factor-kB activity is regulated by inhibitor of kB (IkB), the gene encoding IkB-a was disrupted in mice to observe the in vivo effects of hyperactivation of nuclear factor-kB. IkB-a–/– mice have constitutive nuclear factor-kB activity, severe skin disease, and neonatal lethality. To determine the role of IkB-a deficient immunocytes in the patho- genesis of the skin disease in adult mice, we utilized the RAG2-deficient blastocyst complementation sys- tem to generate RAG2–/–, IkB-a–/– chimeras. These animals display a psoriasiform dermatitis char- acterized by hyperplastic epidermal keratinocytes and dermal infiltration of immunocytes, including lymphocytes. Skin grafts transferred from diseased chimeras to recipient nude mice produce hyperproli- ferative psoriasiform epidermal keratinocytes in response to stimulation. Furthermore, adoptive transfer of lymph node cells from diseased chimeras to RAG2–/– recipient mice recapitulates the disease. Taken together, these characterizations provide evi- dence to suggest that constitutive activation of nuclear factor-kB, due to deficiency in IkB-a, can invoke severe psoriasiform dermatitis in adult mice. Key words: IkB-a/keratinocytes/lymphocytes/psoriasis. J Invest Dermatol 115:1124–1133, 2000 T he ubiquitous transcription factor family, nuclear factor-kB (NF-kB) is composed of five vertebrate DNA-binding proteins, including NF-kB1 (p50), NF-kB2 (p52), cRel, RelA, and RelB. These proteins bind DNA as dimers in a sequence-specific manner to alter gene expression. NF-kB/Rel activity has been demonstrated to be essential in regulating the expression of a number of genes involved in inflammatory responses, immune responses, acute phase reactions, and viral infection, as well as a number of basic cellular functions, including proliferation, differentiation and programmed cell death. In most cells, homo- dimeric or heterodimeric NF-kB is retained in the cytoplasm by association with an inhibitor of kB (IkB). The inhibitors of NF-kB form an evolutionarily conserved multigene family with seven vertebrate members, including IkB-a,IkB-b, Bcl3, IkB-g, p105, p100, and IkB-e. Of these, IkB-a is recognized as a major regulator of NF-kB activity. NF-kB is activated by a variety of signals, including cytokines, bacterial and viral products, T and B cell receptor cross-linking, T cell CD28, and B cell CD40. These signal pathways converge on the phosphorylation and degradation of IkB, releasing NF-kB complexes to translocate to the nucleus where NF-kB can transcriptionally regulate target genes (reviewed in Baldwin, 1996; Ghosh et al, 1998; Sha, 1998). Several observations suggest that the aberrant activation of the NF-kB/IkB family is involved in the development or progression of diseases, such as autoimmune diseases, atherosclerosis, cancer, and acute phase responses such as septic shock (Chen et al, 1999). Several groups have observed a central role for NF-kB in these disease processes. NF-kB is activated in the arthritic synovium (Handel et al, 1995), and NF-kB activity may provide the link between inflammation and hyperplasia that occurs in the arthritic joint (Miagov et al, 1998). Inhibition of NF-kB in a T cell specific fashion in mice results in a reduced incidence of collagen-induced arthritis (Seetharaman et al, 1999). Like arthritis, the initiation and progression of atherosclerosis involves inflammatory responses, as well as the proliferation of vascular smooth muscle cells. Both processes have been shown to involve NF-kB activation (Nakajima et al, 1994; Berliner et al, 1995). Interestingly, NF-kB activity is greatly elevated in mice susceptible to atherosclerosis, particularly when fed with an atherogenic diet (Thurberg and Collins, 1998). These diseases are all involved, to some extent, with the deregulation of the immune response. Psoriasis is a complex disorder of the skin and the immune system and also involves genetic influences (Christophers, 1996). The two major abnormal characteristics of psoriasis are the excessive growth of epidermal keratinocytes in the epidermis and leukocyte infiltration. The proliferating keratinocytes are juxta- posed with immunocytes, including T lymphocytes, monocyte/ macrophages, and dermal dendritic cells (Barker, 1997). Mice that are deficient in IkB-a, the major inhibitor of NF-kB, display a 0022-202X/00/$15.00 · Copyright # 2000 by The Society for Investigative Dermatology, Inc. 1124 Manuscript received April 10, 2000; revised August 24, 2000; accepted for publication August 29, 2000. Reprint requests to: Dr. Fiona E. Yull, Vanderbilt University School of Medicine, A-4320 MCN, 1161 21st Avenue South, Nashville, TN 37232- 2363. Email: fi[email protected] Abbreviations: RAG, recombination activating gene; PCNA, proliferat- ing cellular nuclear antigen.

RAG2–/–, IκB-α–/– Chimeras Display a Psoriasiform Skin Disease

Embed Size (px)

Citation preview

Page 1: RAG2–/–, IκB-α–/– Chimeras Display a Psoriasiform Skin Disease

RAG2±/±, IkB-a±/± Chimeras Display a Psoriasiform SkinDisease

Chih-Li Chen,* Fiona E. Yull,² Nancy Cardwell,²³ Nagendra Singh,* William David Strayhorn,²Lillian B. Nanney,²³ and Lawrence D. Kerr*²§¶Departments of *Microbiology and Immunology, ²Cell Biology, and ³Plastic Surgery, §Vanderbilt University School of Medicine, Vanderbilt-Ingram

Cancer Center, Nashville, Tennessee, U.S.A.; ¶The National Institutes of Health, Transplantation and Immunogenetics Section, Division of Allergy,

Immunology, and Infectious Disease, Bethesda, Maryland, U.S.A.

Nuclear factor-kB, a ubiquitous transcription factorinvolved in in¯ammatory and immune responses, isinappropriately activated in several immuno-relateddiseases, such as allograft rejection, or bronchialasthma. As nuclear factor-kB activity is regulated byinhibitor of kB (IkB), the gene encoding IkB-a wasdisrupted in mice to observe the in vivo effects ofhyperactivation of nuclear factor-kB. IkB-a±/± micehave constitutive nuclear factor-kB activity, severeskin disease, and neonatal lethality. To determine therole of IkB-a de®cient immunocytes in the patho-genesis of the skin disease in adult mice, we utilizedthe RAG2-de®cient blastocyst complementation sys-tem to generate RAG2±/±, IkB-a±/± chimeras.These animals display a psoriasiform dermatitis char-

acterized by hyperplastic epidermal keratinocytesand dermal in®ltration of immunocytes, includinglymphocytes. Skin grafts transferred from diseasedchimeras to recipient nude mice produce hyperproli-ferative psoriasiform epidermal keratinocytes inresponse to stimulation. Furthermore, adoptivetransfer of lymph node cells from diseased chimerasto RAG2±/± recipient mice recapitulates the disease.Taken together, these characterizations provide evi-dence to suggest that constitutive activation ofnuclear factor-kB, due to de®ciency in IkB-a, caninvoke severe psoriasiform dermatitis in adult mice.Key words: IkB-a/keratinocytes/lymphocytes/psoriasis. JInvest Dermatol 115:1124±1133, 2000

The ubiquitous transcription factor family, nuclearfactor-kB (NF-kB) is composed of ®ve vertebrateDNA-binding proteins, including NF-kB1 (p50),NF-kB2 (p52), cRel, RelA, and RelB. Theseproteins bind DNA as dimers in a sequence-speci®c

manner to alter gene expression. NF-kB/Rel activity has beendemonstrated to be essential in regulating the expression of anumber of genes involved in in¯ammatory responses, immuneresponses, acute phase reactions, and viral infection, as well as anumber of basic cellular functions, including proliferation,differentiation and programmed cell death. In most cells, homo-dimeric or heterodimeric NF-kB is retained in the cytoplasm byassociation with an inhibitor of kB (IkB). The inhibitors of NF-kBform an evolutionarily conserved multigene family with sevenvertebrate members, including IkB-a, IkB-b, Bcl3, IkB-g, p105,p100, and IkB-e. Of these, IkB-a is recognized as a major regulatorof NF-kB activity. NF-kB is activated by a variety of signals,including cytokines, bacterial and viral products, T and B cellreceptor cross-linking, T cell CD28, and B cell CD40. These signalpathways converge on the phosphorylation and degradation of IkB,releasing NF-kB complexes to translocate to the nucleus where

NF-kB can transcriptionally regulate target genes (reviewed inBaldwin, 1996; Ghosh et al, 1998; Sha, 1998).

Several observations suggest that the aberrant activation of theNF-kB/IkB family is involved in the development or progressionof diseases, such as autoimmune diseases, atherosclerosis, cancer,and acute phase responses such as septic shock (Chen et al, 1999).Several groups have observed a central role for NF-kB in thesedisease processes. NF-kB is activated in the arthritic synovium(Handel et al, 1995), and NF-kB activity may provide the linkbetween in¯ammation and hyperplasia that occurs in the arthriticjoint (Miagov et al, 1998). Inhibition of NF-kB in a T cell speci®cfashion in mice results in a reduced incidence of collagen-inducedarthritis (Seetharaman et al, 1999). Like arthritis, the initiation andprogression of atherosclerosis involves in¯ammatory responses, aswell as the proliferation of vascular smooth muscle cells. Bothprocesses have been shown to involve NF-kB activation (Nakajimaet al, 1994; Berliner et al, 1995). Interestingly, NF-kB activity isgreatly elevated in mice susceptible to atherosclerosis, particularlywhen fed with an atherogenic diet (Thurberg and Collins, 1998).These diseases are all involved, to some extent, with thederegulation of the immune response.

Psoriasis is a complex disorder of the skin and the immunesystem and also involves genetic in¯uences (Christophers, 1996).The two major abnormal characteristics of psoriasis are theexcessive growth of epidermal keratinocytes in the epidermis andleukocyte in®ltration. The proliferating keratinocytes are juxta-posed with immunocytes, including T lymphocytes, monocyte/macrophages, and dermal dendritic cells (Barker, 1997). Mice thatare de®cient in IkB-a, the major inhibitor of NF-kB, display a

0022-202X/00/$15.00 ´ Copyright # 2000 by The Society for Investigative Dermatology, Inc.

1124

Manuscript received April 10, 2000; revised August 24, 2000; acceptedfor publication August 29, 2000.

Reprint requests to: Dr. Fiona E. Yull, Vanderbilt University School ofMedicine, A-4320 MCN, 1161 21st Avenue South, Nashville, TN 37232-2363. Email: ®[email protected]

Abbreviations: RAG, recombination activating gene; PCNA, proliferat-ing cellular nuclear antigen.

Page 2: RAG2–/–, IκB-α–/– Chimeras Display a Psoriasiform Skin Disease

severe skin phenotype that is reminiscent of human psoriasis, buttypically die within 9 d after birth. The elevated level of NF-kBactivity in hematopoietic cells of these mice may affect chronicin¯ammatory responses resulting in the disease phenotype. Micelacking IkB-a also display excessive growth of keratinocytes andthe in®ltration of immunocytes (Beg et al, 1995; Klement et al,1996). Several cytokines such as tumor necrosis factor (TNF) -a,interleukin (IL) ±1, and IL-8, known to be involved in theprogression of psoriasis are also downstream targets of NF-kB.Based on these observations and the role of NF-kB in multipleother immuno-related diseases, it is reasonable to hypothesize thatthe activation of NF-kB is involved in the pathology of psoriasis.

The neonatal lethality of IkB-a de®cient mice precludes study ofthe function of IkB-a in adult lymphocytes or in the pathogenesisof skin disease; however, use of the RAG2 (recombination activatinggene 2) blastocyst complementation system to generate chimericmice provides an alternative approach to study these questions(Chen et al, 1993). This technique generates chimeras by injectionof normal or mutant embryonic stem cells (ES) cells into RAG2-de®cient blastocysts. RAG2-de®cient mice do not have mature Band T cells due to a de®ciency of the RAG2. Therefore, all maturelymphocytes in the chimeras must be derived exclusively frominjected ES cells. The contribution of the otherwise normalRAG2-de®cient cells to other cells of the resulting chimeras is ableto rescue the neonatal lethality phenotype enabling study of anadult animal. This strategy has been used extensively to study therole of several genes in B and T cell function, in circumstances thatwould otherwise not be possible due to embryonic lethality (Chen,1996). Using this system, we have created IkB-a±/±, RAG2±/±chimeras. These chimeras display a psoriasiform dermatitis that canbe recapitulated by transferring the chimeric lymph node fromaffected mice to RAG2±/± recipients. In response to stimulation,skin grafts from affected chimeras to nude mice also generatehyperplastic epidermal keratinocytes. Our characterizations provideevidence to suggest that such chimeras will serve as a good model tostudy psoriasis.

MATERIALS AND METHODS

Targeting vectors A mouse IkB-a gene, mad3, fragment was clonedfrom a mouse 129 genomic library into gt11 phage. A KpnI (6.5 kb)fragment was subcloned into pBluescript (pBS, Stratagene, La Jolla, CA).The KpnI fragment contains both 5¢ and 3¢ ¯anking regions of IkB-a asdetermined by diagnostic southern analysis. The fragment was used tocreate the targeting construct. NarI was used to remove the 300 bpnucleotides containing the 5¢-untranslated region and part of exon I region,which was replaced with a PGK-Neo cassette in order to construct thetargeting vector, PGK-Neo IkB-a. This single selection construct, PGK-Neo IkB-a was veri®ed by southern blot using external and internalprobes. To create the double selection construct, an XbaI fragmentcontaining pGK-Neo was subcloned into a PGK-tk plasmid to create aPGK-tk Neo IkB-a construct.

ES cell culture and generation of mice Linearized targeting vector(50 mg; was introduced into TL-1 ES cells (a gift from Trish Labosky) andG418-resistant colonies were selected and cultured. DNA isolated from 610colonies was digested with HindIII and SmaI and screened by southernblotting using the KpnI/HindIII fragment as the external probe (probe a)and Neo cassette as the internal probe (probe b). The probe a fragment wasisolated from a SmaI fragment subcloned into pBluescript from the samegt11 phage clone. Four clones from a single selection and ®ve clones from adouble selection with a single targeted copy of IkB-a were identi®ed. Twoclones (nos 332 and 393) with a single targeted copy were used to generatedouble targeted copies as described (Mortensen et al, 1991). Brie¯y, theIkB-a+/± heterozygous ES cells were subjected to selection using 1 mgper ml G418. The surviving clones were selected after 14 d. The doubletargeted ES clones were veri®ed by southern blot using an internal probe(probe c) and an external probe (probe a, data not shown). Eighty percentof the surviving clones were double IkB-a±/± ES clones. Two clones wereselected and microinjected into RAG2-de®cient blastocysts as describedpreviously (Hogan et al, 1986). RAG2-de®cient mice (129/SvEvTacfBr-[KO] rag-2) were obtained from Taconic Farms (Germantown, NY).Chimeric offspring with reconstituted lymphocytes were veri®ed by¯uorescence-activated cell sorter (FACS) analysis using peripheral blood

from 3 wk old mice. All ES cell lines and RAG2 null mice used were fromthe same 129 SV strain background. All the mice used in this study weremaintained in a germ-free environment consisting of autoclaved water,cage, and food. Animals were examined by veterinary staff and no signs ofinfection were detected in affected animals or littermates.

Preparation of nuclear and cytoplasmic extracts Nuclear andcytoplasmic extracts were prepared from spleen, thymus, and peripheralblood cells as previously described (Alkalay et al, 1995). Brie¯y,5 3 106 cells were resuspended in buffer A [10 mM 4-(2-hydroxyethyl)-1-piperazine-ethane-sulfonic acid (pH 7.9); 10 mM KCl, 0.1 mMethylenediamine tetraacetic acid, 0.1 mM ethyleneglycol-bis(b-aminoethyl ether)-N,N,N¢,N¢,-tetraacetic acid, 1 mM dithiothreitol;1 mM phenylmethylsulfonyl¯uoride], and incubated ice for 15 min.Nonidet P-40 was added to 1% vol/vol ®nal concentration, sampleswere vortexed for 10 s and centrifuged at 12,000 3 g for 5 min. Theresulting supernatant containing the cytoplasmic fraction was frozen at±70°C. Buffer C [420 mM NaCl, 20 mM 4-(2-hydroxyethyl)-1-piperazine-ethane-sulfonic acid (pH 7.9; 1.5 mM MgCl2, 0.2 mMethylenediamine tetraacetic acid, 25% glycerol, 1 mM dithiothreitol,1 mM phenylmethylsulfonyl¯uoride, 4 mg per ml leupeptin] was added tothe pellet, and the sample was agitated at 4°C for 30 min. The extract wascentrifuged at 12,000 3 g for 10 min and the supernatant containing thenuclear fraction was frozen at ±70°C. Protein concentrations weredetermined with Lowrie Assay (Bio-Rad, Herculus, CA).

Electrophoretic mobility shift assay An oligonucleotideencompassing a kB DNA binding site (Stratagene, La Jolla, CA) wasend-labeled using g-P32-adenosine triphosphate. A reaction mixturecontaining nuclear extract (4 mg) labeled probe (12,000 cpm); poly(dI±dC) and bovine serum albumin (10 mg) was incubated in the presence ofbinding buffer (20 mM HEPES pH 7.9, 5% vol/vol glycerol, 1 mMethylenediamine tetraacetic acid, 1% nonidet P-40, and 5 mMdithiothreitol). DNA±protein complexes were resolved on a non-denaturing 5% polyacrylamide gel and visualized by autoradiography(Singh et al, 1986).

Ribonuclease protection assay Cytokine mRNA levels in the skinwere analyzed using the RiboQuant Multi-Probe RNase Protection AssaySystem according to the manufacturer's instructions (PharMingen, SanDiego, CA). 32P-labeled anti-sense RNA templates speci®c for IL-4, IL-5,TNF-a, granulocyte-macrophage colony-stimulating factor, macrophagein¯ammatory protein-1a, IL-2, IL-6, g interferon-g), and controls L32 andglyceraldehyde-3-phosphate dehydrogenase were transcribed using anmCK-1 Custom Template Set (PharMingen). Total RNA was isolatedfrom the skin of the indicated mice using Tri-Reagent (MolecularResearch Center, Cincinnati, OH) according to the manufacturer'sinstructions. Twenty micrograms of total RNA was analyzed per reaction.

Immuno¯uorescence staining and ¯ow cytometry Isolation ofsingle cell populations from thymus, spleen, peripheral blood, and bonemarrow was performed as described elsewhere (Boothby et al, 1997).Dispersed cells were surface stained as described previously (Sha et al, 1995),with directly ¯uoresceinated monoclonal antibodies (PharMingen). Stainedcells were subjected to analysis by FACScan ¯ow cytometer (BectonDickinson, San Jose, CA) The monoclonal antibodies used were: anti-B220(PharMingen) (01125A), anti-CD4 (01065A), anti-CD8 (01044A), anti-IgD (02214D), anti-IgM (1021-09; Southern Biotech, Birmingham, AL).The FACS analysis was performed using Cell Quest program.

Preparation of puri®ed splenic B and T cells Spleen cells fromchimeras were ®rst depleted of erythrocytes by hydrolysis, followed byincubation at 37°C in complete medium for 1 h to allow macrophageattachment to the bottom of the plate. After macrophage depletion, thesuspended cells were gently transferred to 15 ml tubes and counted.3 3 107 cells were incubated with 1.5 ml of anti-Thy 1.2, anti-CD4(hybridoma G.K. 1.5), and anti-CD8 (hybridoma T105) hybridoma mediaon ice for 40 min followed then washed. Cells were then transferred toplates coated with anti-rat IgG (212±005±103; Jackson ImmunoresearchLab., West Grove, PA) to deplete the T cell population and plated at 4°Cfor 1 h. Spleen cells (3 3 107) were incubated in media containing anti-IgM(Biosource Internal Inc, Camarilla, CA) bound plate at 4°C for 1 h todeplete B cells. After panning out of B or T cells, the purity of T and B cellpopulations was veri®ed by FACS analysis using anti-CD3 (01048A), anti-B220, and anti-Mac-1 (01715B) antibodies (PharMingen). Normally, 80±85% purity of B or T cells was obtained.

VOL. 115, NO. 6 DECEMBER 2000 RAG2±/±, IkB-a±/± CHIMERAS 1125

Page 3: RAG2–/–, IκB-α–/– Chimeras Display a Psoriasiform Skin Disease

Proliferation assays of B and T cells in culture All primary cells werecultured in complete medium containing RPMI-1640 (GIBCO/BRL,Rockville, MD) supplemented with 10% heat-inactivated fetal bovineserum, 50 mM 2-mercaptoethanol, and 200 mM L-glutamine. Spleen cellswere plated at 1 3 106 leukocytes per ml. Puri®ed B and T cells werecultured at concentrations of 1 3 106 cells per ml. For proliferation assays,the puri®ed B or T lymphocytes were dispensed into 96-well microtiterplates at 200 ml per well. B lymphocytes were stimulated with anti-IgM20 mg per ml (Jackson Immunoresearch Laboratory, West Grove, PA), anti-CD40 2 mg per ml (Pharmingen), and lipopolysaccharide 10 mg per ml(Sigma, St Louis, MO). T lymphocytes were stimulated with concanavalinA (2.5 mg per ml, Pharmacia), anti-CD3 (1/30 dilution, culture supernatantof anti-CD3 hybridoma 140C11) in the presence of irradiated splenic cells(5 3 105) from C57BL/6 mice for 48 h. Fresh medium containing 0.75 mCi[3H]thymidine (Amersham, Piscataway, NJ) per well was added andincubated for 16 h. Cells were harvested with a cell harvester (Tomtec,Orange, CT), and uptake of radioactivity was measured with a betaplatereader (Wallac, Gaithersburgh, MD).

Histologic analysis and immunohistochemistry Skin and spleensfrom the 13 wk old chimeras (bone marrow recipients 5 wk after transfer)were ®xed in 4% paraformaldehyde, embedded in paraf®n, and sectioned at

5 mm. Sections were stained with hematoxylin and eosin or used forimmunohistochemistry. Proliferating cellular nuclear antigen (PCNA)immunoperoxidase staining was performed using a Vector ABC EliteMouse IgG Kit (PK-6102) and using anti-PCNA antibody (1486-772;Roche, Indianapolis, IN). Epidermal thickness was measured as described(Fraki et al, 1983). To rule out the possibility that the psoriasiformdermatitis was induced by either bacterial or fungal agents, additional gramstaining and periodic acid-Schiff staining was performed on skin sections.No evidence of bacterial or fungal infection was detected when sectionswere examined from regions of skin most heavily affected by thepsoriasiform phenotype. Thus it seems highly unlikely that thepsorasiform dermatitis was induced by Corynebacterium bovis orunclassi®ed fungal agents.

Bone marrow and lymph node transfer Bone marrow or lymph nodecells were taken from the chimeras at 13 wk of age as described elsewhere(Boothby et al, 1997). To deplete mature B and T cells, bone marrow cellswere incubated with anti-thy 1.2, anti-CD4 (hybridoma G.K 1.5) and anti-CD8 (hybridoma T105) for 45 min at 4°C. After washing with fresh coldmedium twice, bone marrow cells were incubated in plates precoated withanti-rat IgG and anti-IgM m chain (Jackson Immunoresearch Laboratory)antibodies for 45 min at 4°C. Then, 5 3 105 bone marrow cells wereinjected into irradiated RAG2-de®cient mice. Lymph node cellswere injected into unirradiated-RAG2-de®cient mice at 2 3 106,5 3 106, and 10 3 106 for RAG2±/±, IkB-a±/±, 4 (or 5) 3 106 forRAG2±/±, IkB-a+/+.

Skin grafts Skin grafts were completed as described (Nanney et al, 1992).Brie¯y, athymic nude mice (Taconic Farms) were anesthetized with anintramuscular injection (0.1 ml) of Ketaset plus acepromazine and thepotential graft site, a full-thickness recipient bed measuring 2.5 3 2.5 cm,was created on the dorsal surface of the mouse. The skins from chimeraswere divided into four individual grafts and one graft per mouse was ®xedinto position using Dermalon suture (Davis-Geck, Davis, CA). Anonadherent bolster dressing (Release, Johnson and Johnson, Wayne,PA) was sutured over the wound. Grafted mice were housed individually in®lter-topped cages. The bolster dressings and sutures were removed after7 d, and wound margins were allowed to heal for 8 wk. The grafted micewere stripped with wax after 8 wk. Three days later, the stripped mice werekilled. The skins were prepared for histologic analysis.

RESULTS

Generation of RAG2±/±, IkB-a±/± chimeras The neonatallethality of IkB-a null mice precludes studying the effect ofconstitutive NF-kB activity on adult lymphocytes and other tissues.The RAG2-de®cient blastocyst complementation system providesan alternative method to carry out these studies. RAG2-de®cientmice have no mature B or T cells due to an inability to initiateV(D)J recombination (Shinkai et al, 1992); however, injection ofnormal or mutant ES cells into RAG2±/± blastocysts leads to thegeneration of somatic chimeras. In such chimeras, all mature B andT cells are derived from the injected ES cells. To investigate theeffect of constitutive NF-kB activation on adult lymphocytes, IkB-a±/± ES cells were generated and injected into RAG2±/±blastocysts to create chimeric adult mice. IkB-a±/± ES cells weregenerated by subjecting two different IkB-a+/± ES cell lines toelevated G418 selection. IkB-a±/± clones were veri®ed bySouthern analysis (Fig 1). Two representative IkB-a±/± clones(nos 72 and 89) derived from two different ES IkB-a+/± lines (nos332 and 393, respectively) were injected into RAG2±/± blastocysts.Six injections into RAG2-de®cient blastocysts, two for each cloneof IkB-a±/± and control IkB-a+/+ ES cells, were completed.Twenty-one pups were generated from IkB-a±/± ES cellinjections whereas 24 pups were generated from IkB-a+/+ EScell injections. As the ES cell clones and the RAG2±/± recipientblastocysts are from the same 129 SV background, selection ofchimeras using coat color was not feasible. Thus selection ofchimeras for analysis was completed on the basis of reconstitution oflymphocyte populations as determined by FACS analysis. ThreeRAG2±/±, IkB-a±/± chimeras (72, 89, 72.2) and 12 controlRAG2±/±, IkB-a+/+ with reconstituted lymphocyte populationswere generated.

Figure 1. Targeted disruption of IkB-a gene in TL-1 ES cells. (A)Schematics of IkB-a targeting constructs. Two targeting vectors, PGK-tk-Neo IkB-a (double selection) and PGK-Neo IkB-a (single selection),were used to disrupt the IkB-a gene via homologous recombination. Aschematic showing restriction enzyme sites within the IkB-a gene andrecombination locus is depicted. (B) Southern blot analysis identi®es IkB-a±/± ES clones. Shown are examples of wild-type (+/+), heterozygous(+/±), and homozygous (±/±) ES clones. DNA (10 mg) isolated from ESclones was digested with HindIII/SmaI or BamHI and used for southernblot analysis. The sizes of diagnostic fragments detected by southern blotanalysis are indicated. Probe a, an external probe, is a 0.7 kb HindIII/KpnIfragment from the 3¢ end of IkB-a gene. Probe c, an internal probe, is a0.4 kb fragment from the promoter region of IkB-a. The HindIII/SmaIdigest generates a 5 kb fragment diagnostic for the targeted allele. TheBamHI digest generates diagnostic fragments in which the 3 kb fragmentrepresents the targeted allele.

1126 CHEN ET AL THE JOURNAL OF INVESTIGATIVE DERMATOLOGY

Page 4: RAG2–/–, IκB-α–/– Chimeras Display a Psoriasiform Skin Disease

RAG2±/±, IkB-a±/± chimeras develop severe skindisease RAG2±/±, IkB-a±/± chimeras were normal at birth.The most dramatic phenotype began to appear in two (nos 72 and89) of the three chimeras at approximately 5 wk after birth whenthe mice developed alopecia, characterized by hair loss around theirsnout, ears, head, and neck regions. The underlying skin of thesechimeras was red, dry, scaly, ¯aky, and itchy, all signs indicative ofan in¯ammatory response of the skin. Skin punches taken from earlobes of RAG2±/±, IkB-a±/± mice contained evidence ofhyperkeratosis, epidermal hyperplasia, neutrophilic in®ltration tothe epidermis with microabscesses, and pustular psoriasis in theaffected animals. These effects were not observed in the RAG2±/±,IkB-a+/+ animals or nonchimeric littermates. The severe skinpathology ultimately extended over the entire surface of the mouse.Upon sacri®ce, histologic specimens were prepared from snout,ears, neck, dorsal surface, tail, and footpads. Histologic analyzesreveal clear evidence of epidermal thickening and microabscessformation (Fig 2A, C). In order to compare the epidermalthickness of IkB-a±/± or wild-type reconstituted animals, astandard formula was used to measure the skin layers (Fig 2B).

Dramatic differences were noted between RAG2±/±, IkB-a+/+and RAG2±/±, IkB-a±/± mice in which 4.5-, 3.9-, 2.7, and 2.9-fold increases in epidermal thickness were documented in snout,ears, dorsal, and footpad regions, respectively (Fig 2B).

In order to determine if epidermal thickness in RAG2±/±, IkB-a±/± mice might be due to hyperproliferation of skin keratino-cytes, immunohistochemistry was performed using an antibodyagainst PCNA. A dramatic increase in PCNA-positive cells wasobserved in RAG2±/±, IkB-a±/± skin within the epidermis(Fig 2D). These proliferative keratinocytes were also presentsuperior to the basal layer, although increased PCNA+ cells werenoted in dermis as well. These results demonstrate one of thehallmark characteristics of psoriasiform skin, the hyperproliferationof epidermal keratinocytes in the presence of in®ltrating lympho-cytes.

Lymphocyte development in RAG2±/±, IkB-a±/± mice Allmature B and T cells of chimeras should be derived from IkB-a-de®cient ES cells, as RAG2 de®ciency precludes the generation ofmature B and T cells. To con®rm this and to determine the IkB-a±/± contribution to the skin of chimeric animals, Southern

Figure 2. Histologic analysis of skin from diseased RAG2±/±, IkB-a±/± chimeras. (A) Hematoxylin and eosin-stained skin micrographs contrastingthe skin disease in the right panel showing a RAG2±/±, IkB-a±/± chimera with the left panel showing a RAG2±/±, IkB-a+/+ control chimera. (B) Tableshowing the average epidermal thickness. Shown are representative measurements from two RAG2 ±/±, IkB-a±/± chimeras and two IkB-a+/+ controlchimeras. The data suggest that de®ciency in IkB-a alters the epidermal thickness in multiple body regions. (C) Hematoxylin and eosin staining of ear skinremoved at 5 wk of age from a severely affected RAG2±/±, IkB-a±/± chimera and an unaffected RAG2±/±, IkB-a+/+ control chimera. Mice de®cient forIkB-a display a greatly thickened acanthotic epidermis with microabscesses and a dermal in®ltrate. (D) Immunoperoxidase staining comparing thedistribution and abundance of cells positive for PCNA in ear skin removed at 5 wk of age from an affected RAG2±/±, IkB-a±/± chimera and an unaffectedRAG2±/±, IkB-a+/+ chimera. Mice lacking in IkB-a show a dense lymphocytic in®ltrate in the thickened dermal region, and a greatly expandedproliferative population within the basal epidermal compartment. Additional psoriasiform features include prominent acanthosis of the epidermis as well as afocal parakeratosis (arrows). (A) 20 3; (C, D) 40 3. +/+: RAG2±/±, IkB-2+/+; ±/±: RAG2±/±, IkB-a±/±.

VOL. 115, NO. 6 DECEMBER 2000 RAG2±/±, IkB-a±/± CHIMERAS 1127

Page 5: RAG2–/–, IκB-α–/– Chimeras Display a Psoriasiform Skin Disease

analysis was performed to detect diagnostic fragments in genomicDNA of chimeric tissue (Fig 3A). The results show variation in theef®ciency of reconstitution of the three chimeras (nos 72, 89, 72.2).The vast majority of lymphocytes from thymus and spleen chimerasare IkB-a±/± cells. This would be expected as reconstitution of thelymphocyte population was criterion used to select chimeras foranalysis. Interestingly, the two chimeras (nos 72, 89) that displaydisease have more than 50% of skin derived from IkB-a±/± ES cells

whereas the chimera that did not develop disease had less than 10%of skin derived from IkB-a±/± ES cells.

To investigate the effects of IkB-a de®ciency on the activationof NF-kB, nuclear extracts from peripheral blood, thymus, orspleen cells were analyzed for NF-kB DNA-binding activity viaelectrophoretic mobility shift assay. As expected, signi®cantlyelevated levels of NF-kB binding activity were evident in all IkB-a±/± chimeras, consistent with IkB-a±/± de®ciency (Fig 3B).This indicates that the absence of IkB-a leads to constitutiveactivation of NF-kB in adult immunocytes.

To elucidate the effects of IkB-a de®ciency on the expression ofgenes downstream of NF-kB that are potentially involved in skindisease and in¯ammation, RNase protection assays were performedusing RNA from the skin of sick or control chimeras. TNF-a, acytokine known to be involved in acute in¯ammatory or acutephase responses (Arai et al, 1990; Baumann and Gauldie, 1994) andmacrophage in¯ammatory protein-1a, involved in macrophageactivation (Schall et al, 1993), were elevated in the skin ofRAG2±/±, IkB-a±/± chimeras relative to control littermates(Fig 3C). Other cytokines such as granulocyte-macrophagecolony-stimulating factor, IL-2, and interferon-g were not elevated(data not shown). The results suggest that proin¯ammatorycytokines regulated by NF-kB are upregulated in the absence ofIkB-a and may contribute to the dermatologic phenotype.

Given the critical role of NF-kB family members in lymphocytedevelopment (Sha, 1998), it might be hypothesized that constitu-tive NF-kB activation advances immunocyte differentiation. Totest this idea, thymocytes, splenocytes, and peripheral lymphocytescollected from RAG2±/±, IkB-a±/± chimeras and controls werestained with antibodies that detect stage-speci®c markers of B andT cells. FACS analysis of these cells with the T cell differentiationmarkers, CD4 and CD8, revealed that the proportions of CD4+ orCD8+ single positive cells from RAG2±/±, IkB-a±/± chimeras arecomparable with those from RAG2±/±, IkB-a+/+ chimeras(Fig 4A). Interestingly, analysis of cells from RAG2±/±, IkB-a±/±chimeras stained with B cell differentiation markers (B220, IgM,and IgD) revealed that the proportion of cells expressing high levelsof surface IgM in the RAG2±/±, IkB-a±/± spleen cells wereelevated (approximately 2-fold) relative to controls (Fig 4B). Thelevels of IgM on the IgD-positive population are elevated on thesurface of splenic cells from the RAG2±/±, IkB-a±/± chimeras(Fig 4C±E). These results suggest that constitutive NF-kBactivation skews the development of B cells towards mature stages.

B and T cells from RAG2±/±, IkB-a±/± chimeras displayproliferation defects Animals de®cient in individual NF-kBmembers have demonstrated that kB transcriptional activity iscritical for lymphocyte proliferation in response to B and T cellspeci®c stimuli (for review, Sha, 1998). In addition, use of thetrans-dominant inhibitor IkB-aDN has revealed that functionalinhibition of NF-kB transcriptional activation alters T and B cell exvivo proliferation (Boothby et al, 1997; Bendall et al, 1999). Thus,we were interested in investigating the effects of constitutive NF-kB activation on the proliferation of mature B and T cells. Spleencells isolated from RAG2±/±, IkB-a±/± chimeras and controlmice, and equal numbers of the enriched B and T cells were used tomeasure ex vivo [3H]thymidine incorporation to investigateinduction of proliferation ability in response to indicated stimuli(Fig 5). Analysis of T cells stimulated with anti-CD3 orconcanavalin A in the presence of antigen-presenting cellsshowed decreased [3H]thymidine uptake in T cells fromRAG2±/±, IkB-a±/± chimeras as compared with those fromRAG2+/+, IkB-a+/+ animals. In contrast to results from T cells,analysis of B cells treated with speci®c stimulators of B cells(lipopolysaccharide, anti-IgM, anti-IgM plus anti-CD40, anti-CD40) demonstrated an increased uptake of [3H]thymidine in Bcells from RAG2±/±, IkB-a±/± chimeras relative to wild-typecontrols. These results indicate that constitutive NF-kB activationalters the ex vivo proliferative ability of mature B and T cells in a

Figure 3. IkB-a±/± ES cells reconstitute lymphocyte populationsand disruption of IkB-a expression results in constitutive NF-kBDNA binding activity and increased target gene expression. (A)Southern blot analysis to con®rm that ES clones (72, 89) have reconstitutedthymus and spleen cells of RAG2±/±, IkB-a±/± chimeras (72, 72.2, 89).Genomic DNA isolated from immunocytes was digested with BamHI andthe blot was probed with probe C from Fig 1. (B) Nuclear extracts (4 mg)from secondary lymph organs of RAG2±/±, IkB-a±/± chimeras andcontrol littermates were subjected to electrophoretic mobility shift assayusing a human immunode®ciency virus-long terminal repeat kB bindingelement as probe. Three independent experiments were completed fromthree different mice (72, 72.2, 89). Shown are representative resultsgenerated from the clone 89 chimera. Competitor oligo: 100 3. (C) RNaseprotection assay indicates that the transcriptional levels of several cytokines,including TNF-a and macrophage in¯ammatory protein-1a are elevatedin the skin from affected chimeras as compared with those from controlanimals. L32 and glyceraldehyde-3-phosphate dehydrogenase are loadingcontrols. +/+: RAG2±/±, IkB-a+/+; ±/±: RAG2±/±, IkB-a±/±; 0:RAG2 null control; IkB-a±/±: IkB-a null control.

1128 CHEN ET AL THE JOURNAL OF INVESTIGATIVE DERMATOLOGY

Page 6: RAG2–/–, IκB-α–/– Chimeras Display a Psoriasiform Skin Disease

very different manner. The results shown are representative of allthree chimeras.

Lymph node adoptive transfer reproduces the psoriatic skinphenotype in RAG2-de®cient mice It has been demonstratedthat adoptive transfer of a speci®c subset of lymphocytes canrecapitulate several immune diseases, including chronicin¯ammation (Davidson et al, 1996). To explore the etiology ofthe skin disorder of RAG2±/±, IkB-a±/± chimeras, cells from thelymph node and bone marrow of affected chimeras or RAG2±/±,IkB-a+/+ controls were adoptively transferred to untreated orirradiated RAG2±/± de®cient mice. Nine untreated RAG2-de®cient mice received lymphocytes from the lymph nodes ofRAG2±/±, IkB-a±/± chimeras. Six of the nine RAG2±/±recipients received lymphocytes from chimeras 72 and 89, i.e.,obviously sick mice. These recipients developed skin disease within5 wk post-transfer whereas the recipients of cells from the 72.2chimera, which itself did not develop skin disease, remainedhealthy. Analysis of peripheral blood con®rmed that thereconstituted lymphocyte population from affected chimeras iscomparable with those from controls (Fig 6A). Histologic analysisof ear punches from sick recipients revealed a near identicalpsoriasiform dermatitis to that observed in the RAG2±/±, IkB-a±/± chimeras (Fig 6B). Interestingly, when the mice weresacri®ced at 6 mo after transfer, the skin disease appeared lesssevere, suggesting that as IkB-a±/± immunocytes aged and wereeliminated from the system, disease progression was attenuated.These observations suggest that IkB-a±/± lymphocytessigni®cantly contribute to the skin disease of affected chimeras

and that NF-kB activity may contribute to the progression of thisdisease.

Six irradiated RAG2±/± mice received the mature B and T celldepleted bone marrow cells from RAG2±/±, IkB-a chimeras.Mice receiving bone marrow cells from affected chimeras appearednormal for 4 wk, and then became ruf¯ed and hunched. Tworecipients died at approximately 6 wk post transfer. The remainingfour animals were sacri®ced for analysis. Recipients of wildtypebone marrow remained healthy until killed at 6 wk post-transfer.FACS analysis of bone marrow cells from recipients of RAG2±/±,IkB-a±/± chimeras, sacri®ed prior to pathogenic death, revealed areduction in B cell precursors (Fig 6C). In agreement with this®nding, fewer T and B cells were evident in the secondary lymphorgans analyzed including lymph nodes, spleen and thymus viaFACS relative to the cells from wild-type bone marrow recipients(data not shown). Spleen sections from animals that received bonemarrow of affected mice contained reduced numbers of lympho-cytes and lacked primary follicle structures (Fig 6D). Secondarylymph organs of wild-type bone marrow recipients showed normalreconstitution of lymphocyte populations (data not shown). Thecause of death of RAG2±/±, IkB-a±/± bone marrow recipients isnot known. Potentially, other hematopoietic stem cell populationsare also abnormal in bone marrow derived RAG2±/±, IkB-a±/±animals and are therefore unable to reconstitute other critical celltypes.

Skin from affected chimeras retains the psoriatic phenotypewhen grafted on nude mice Several lines of evidence suggestthat human skin diseases such as psoriasis can be transferred by skingraft on to an immune-de®cient host (Sundberg et al, 1994;Nickoloff et al, 1995). We wished to investigate whether the skindisease in RAG2±/±, IkB-a±/± chimeras would be maintained ingrafts transferred to nude mice. Eight or 12 wk following grafting,mice were stripped with wax to initiate an in¯ammatory responseand skin grafts were harvested after 3 d. Interestingly, the graftsfrom affected chimeras still show the skin disease syndrome

Figure 5. Constitutive NF-kB activation alters T and B cellproliferation in response to stimulation. Thymocytes and enriched Band T cells (2 3 105) were plated in 96 wells in the absence or presence ofindicated stimulators. [3H]thymidine (0.75 mCi) was added to the wells after48 h stimulation. Cells were collected 16 h after addition of [3H]thymidineand then counted. Concanavalin A (5 mg per ml) and anti-CD3 (1:30dilution from supernatant from hybridoma culture) in the presence ofirradiated antigen presenting cells, and phorbol myristate acetate (33.3 ngper ml) and ionomycin (5 mM; were used to stimulate puri®ed T cells. IgM(25 mg per ml) with or without anti-CD40 (2 mg per ml), anti-CD40 (2 mgper ml) alone, or lipopolysaccharide (10 mg per ml) were used as B cellspeci®c stimulators. Shown are the results of RAG2±/±, IkB-a+/+ inblack bars and RAG2±/±, IkB-a±/± in white bars. Results arerepresentative of those from all three chimeras. Cells from each chimerawere analyzed in triplicate. Error bar = SD.

Figure 4. The effect of constitutive NF-kB activity on the devel-opment of lymphocytes. (A) FACS analysis demonstrates the normaldevelopment of IkB-a±/± T cells in RAG2±/±, IkB-a±/± chimeras.Lymphocytes isolated from thymus, spleen, or peripheral blood werestained with ¯uorescence-labeled CD4 and CD8 antibodies to determinethe differentiation status of T cells. (B) FACS analysis indicates theproportion of IgMhiIgDhi cells is elevated in spleens of RAG2±/±, IkB-a±/± chimeras. (C) The gate used for the histogram analysis of (D) and (E).Histogram of FACS analysis indicates that the IgDlo (D) and IgDhi (E) IkB-a±/± B cells have higher levels of IgM as compared with IkB-a+/+ Bcells. Shown are representatives of three mice.

VOL. 115, NO. 6 DECEMBER 2000 RAG2±/±, IkB-a±/± CHIMERAS 1129

Page 7: RAG2–/–, IκB-α–/– Chimeras Display a Psoriasiform Skin Disease

(Fig 7A). No involvement of recipient's skin was observed. Toinvestigate the proliferative status of keratinocytes, skin sectionsfrom 8 wk and 12 wk grafts were examined for expression ofPCNA, a marker of cellular proliferation. Proliferatingkeratinocytes in the skin grafts derived from affected chimeraswere greater in number than keratinocytes in grafts from wild-typechimeras (Fig 7B). The thickness of grafts from affected chimeras isthree to four times greater than the grafts from control animals.Interestingly, the thickness of the grafts was reduced at 12 wk ascompared with 8 wk (Fig 7C). These results indicate thatpsoriasiform dermatitis can be maintained by grafts transferredfrom affected chimeras, consistent with studies of human psoriasis(Krueger et al, 1975; Nickoloff et al, 1995).

DISCUSSION

Mice lacking IkB-a have been independently created by threegroups, including the mice described here (Beg et al, 1995; Klementet al, 1996). A phenotype common to all studies is a severepsoriasiform dermatitis. The neonatal lethality of this knockout,however, precludes the possibility of studying the interplaybetween the keratinocytes and immunocytes in adult animals, bothof which are known to play crucial parts in the development of skindiseases of humans (Nickoloff, 1998). Using a RAG2-de®cientblastocyst complementation assay, we have created mosaic chimerasof RAG2±/±, IkB-a±/± mice. Unlike the neonatal lethalitydisplayed by IkB-a-de®cient mice, these chimeric mice surviveinto adulthood. Chimeras, however, display a skin disease similar tothat of IkB-a knockout mice, and also defects in the differentiationand function of lymphocytes. This suggests that neonatal lethality ofIkB-a±/± is not exclusively due to the defects in the skin of theseanimals. Furthermore, these chimeras provide a model to study notonly lymphocyte development but also pathogenesis of psoriasiformdermatitis.

Effects of constitutive NF-kB activity on lymphocytedevelopment and proliferative response De®ciency inindividual NF-kB/IkB family members has minor effects on thedevelopment of lymphocytes, but major effects on theirproliferative responses and immune function (reviewed in Sha,1998). In addition, NF-kB activity is known to be involved inregulating apoptosis. Our study shows that constitutive NF-kBactivity skews B cell development towards an advanceddifferentiation stage, but has no apparent effect on T cell

Figure 6. Lymph node adoptive transfers reproduce the phenotypeof RAG2±/±, IkB-a±/± in RAG2-de®cient mice. Bone marrowtransfer recipients die. (A) FACS analysis indicates that reconstitutedlymphocytes derived from the lymph nodes of RAG2±/±, IkB-a±/±chimeras are comparable with those from RAG2±/±, IkB-a+/+ chimeras.(B) Lymphocytes collected from lymph nodes of RAG2±/±, IkB-a+/+ orRAG2±/±, IkB-a±/± chimeras were injected into RAG2±/± mice. Earpunch of recipients was sectioned and stained with hematoxylin and eosin.Shown are results from skin sections of the recipients receiving 5 3 106 cellsfrom RAG2±/±, IkB-a+/+ or RAG2±/±, IkB-a±/± chimeras. Results arerepresentative of six recipients. The affected epidermis is greatly thickened,increased dermal lymphocytic in®ltration is apparent, and a microabscess ispresent at the epidermal surface. Table of the thickness of epidermis of skinfrom the ear of lymph node recipients of RAG2±/±, IkB-a±/± andRAG2±/±, IkB-a+/+. Results are representative of six recipients. (C)FACS analysis of bone marrow cells taken from recipients 6 wk post-transfer indicates that the B cell precursors are reduced in the recipients ofRAG-2±/±, IkB-a±/± bone marrow; however, the elevated level of CD43indicates that the lymphoid or myeloid precursors are increased in therecipients of RAG2±/±, IkB-a±/± bone marrow cells. Shown arerepresentatives of four recipients. (D) Histologic analysis of spleen indicatesthat lymphocytes are reduced and spleen structure is altered in therecipients of RAG2±/±, IkB-a±/± cells. Results are representative of fourrecipients. (B) 40 3; (D) 20 3.

Figure 7. RAG2±/±, IkB-a±/± skin grafts demonstrate sustainedepidermal thickness in the absence of endogenous lymphocytes,retain extensive proliferation and respond to trauma with an evengreater increase in epidermal thickness. Skin grafts from diseased andcontrol mice were transplanted on to nude recipients. Eight week post-transfer, grafts were stimulated by the mild trauma of wax-stripping andmice were killed 3 d later. (A) Histologic analysis indicating stripped skingrafts from RAG2±/±, IkB-a±/± affected chimeras and controls. (B)PCNA immunoperoxidase staining indicating the proliferating keratino-cytes in the epidermal layer of skin grafts from RAG2±/±, IkB-a±/±chimera. (C) Measurement of epidermal thickness of skin grafts fromaffected chimeras to athymic nude mice. Results are representative of fourmice. (A) 20 3; (B) 40 3.

1130 CHEN ET AL THE JOURNAL OF INVESTIGATIVE DERMATOLOGY

Page 8: RAG2–/–, IκB-α–/– Chimeras Display a Psoriasiform Skin Disease

development. In addition, our data show that IkB-a±/± B cellshave hyperproliferative responses to speci®c stimuli, whereas T cellsexhibit impaired proliferative responses. Our observations areconsistent with the ®ndings from studies of lymphocytes de®cientin p105, another IkB family member (Ishikawa et al, 1998).Whether these ®ndings are due to the effects of constitutive NF-kBactivity on apoptosis, differentiation, or cell cycle of lymphocytesremains unclear.

Psoriasis, a chronic hyperproliferative skin disease affectingapproximately 1.5±2.0% of the Caucasian population, is character-ized histologically by prominent keratinocyte hyperplasia and anearly in¯ammatory cell in®ltrate, including T lymphocytes andmacrophages. Studies on the pathophysiology of psoriasis have alsoindicated that cytokines, including TNF-a, IL-1, IL-8, andadhesion molecules, including integrins, endothelial cell adhesionmolecules, and vascular cell adhesion molecules are correlated withthis disease (Nickoloff, 1991). Several lines of evidence suggest thatthe activity of NF-kB may be involved in the development ofpsoriasis. Transgenic mice, which express human TNF-a under theregulation of a keratin promoter, and mice expressing IL-1a fromthe keratin 14 promoter (Cheng et al, 1992; Groves et al, 1995),exhibit immune cell in®ltration of the dermis. These two cytokinesare well known as NF-kB activators (Beg et al, 1993). Severalcytokines and adhesion molecules involved in development ofpsoriasis have been postulated to be downstream target genes ofNF-kB, including IL-1, IL-6, TNF-a, interferon-g, IL-8, vascularcell adhesion molecule-1, endothelial cell adhesion molecule-1,and intercellular adhesion molecule-1 (Nickoloff and Mitra, 1992;Ghosh et al, 1998). This study detected changes in the levels ofTNF-a, and macrophage in¯ammatory protein-1a in the skin ofRAG2±/±, IkB-a±/± chimeras relative to control littermates.Consistent with the ®ndings of other groups (Klement et al, 1996),however, granulocyte-macrophage colony-stimulating factor, acytokine involved in the differentiation of granulocytes, and thesteady-state levels of IL-2 and interferon-g are not signi®cantlychanged (data not shown). High NF-kB activity has been observedin the skin of transgenic mice expressing a reporter gene under thecontrol of NF-kB DNA binding elements (Frucht et al, 1991; Buzyet al, 1995; F.E. Yull unpublished data). Alteration of NF-kBactivity affects the proliferation or apoptosis of keratinocytes (Seitzet al, 1998; Wrone-Smith et al, 1997; Chaturvedi et al, 1999; Li et al,1999). Psoriasis is known to occur in association with humanimmunode®ciency virus infection, another well described inducerof NF-kB (Johnson et al, 1985; Duvic, 1990). Based on theseobservations and the fact that lack of IkB-a results in a severepsoriasiform dermatitis characterized by hyperplastic keratinocytesand immunocyte in®ltration, it is proposed that NF-kB activityplays a pivotal stimulatory role in the pathogenesis of psoriasis.

In this study, we showed that RAG2±/± IkB-a±/± chimerasdevelop a disease with features of psoriasiform dermatitis (Liu andParsons, 1983). Although the mice appear normal at 3 wk of age, by5±6 wk old, the animals develop alopecia and mild scaling of theskin that subsequently becomes more severe. After 7 wk the animalssuffer from severe pruritus demonstrated by frequent scratching.Histologic analysis reveals a hyperproliferative epidermis, paraker-atosis, microabcesses, and immunocyte in®ltration. This histo-pathology is consistent with human psoriasis. Adoptive transfer oflymph node cells from sick mice to unirradiated RAG2-de®cientmice recapitulates the chronic psoriasiform dermatitis indicatingthat hematopoietic cells, including lymphocytes are involved in thedevelopment of this skin phenotype. Skin grafts from the affectedchimeras to athymic nude mice can sustain the hyperplastickeratinocytes for periods of at least 12 wk. In summary, theseobservations suggest that the RAG2±/±, IkB-a±/± chimerasdevelop psoriasiform dermatitis.

The use of a severe combined immunode®cient mouse±humanskin model has revealed that psoriasis is an immunologicallymediated skin disease linked to several different class I majorhistocompatibility complex alleles. Using this model, studiesreported from several groups indicated that T lymphocytes and

natural killer cells are involved in the pathogenesis of psoriasis(Nickoloff and Wrone-Smith, 1997; Nickoloff, 1999; Nickoloffet al, 1999). In this study, as only two of three RAG2±/±, IkB-a±/± chimeras in which IkB-a±/± ES cells successfully recon-stituted the lymphocyte populations display the cutaneousphenotype, it appears that IkB-a±/± lymphocytes alone may notbe able to initiate the skin disease. Therefore, an interplay betweenkeratinocytes and immunocytes may be important for the process ofdisease initiation and progression. This theory is strengthened bythe observation that there appears to be a requirement for an IkB-a±/± component to the skin in addition to IkB-a null lymphocytesin order to have a strong display of psoriasiform dermatitis. Thechimera, 72.2, which did not develop skin disease, had a very lowproportion of skin derived from IkB-a±/± ES cells. If aninteraction between IkB-a±/± skin and immunocytes was critical,we would expect that lymph node or bone marrow adoptivetransfer from the sick mice would not be suf®cient to recapitulateskin disease; however, some degrees of skin disease were producedby the lymph node transfers. Perhaps an interplay betweenkeratinocytes and immunocytes is important for the initiation ofskin disease followed by a systemic effect, possibly upon cytokinesecretion, or adhesion molecule expression. This systemic effectmay result in fundamental changes to the lymphocyte population.This lymphocyte population might then be suf®cient to initiate thedisease process after lymph node transfer. We have observedincreased expression of IL-2 receptors on the cell surface of T cellsfrom sick mice in the peripheral blood (data not shown), and theelevated basal proliferation of T and B cells would suggest that theselymphocytes are functionally changed. In addition, the lymph nodetransfers from the nonskin disease RAG2±/±, IkB-a±/± chimera(72.2) to the RAG2-de®cient mice did not produce psoriasiformdermatitis, an observation consistent with the above hypothesis.

Skin grafts from the affected chimeras to athymic nude miceretain the psoriasiform dermatitis for an extended time and provideevidence to suggest that keratinocytes are important in diseaseprogression (Krueger et al, 1975). The observation that the severityof skin disease is reduced after a lengthy period of time suggests thatthe absence of abnormal hematopoietic cells allows the skin patchto heal. Thus factors from abnormal hematopoietic IkB-a±/± cellsmay be important in maintaining the skin disease.

Recently, numerous rodent models have emerged that exhibitvarious psoriasiform abnormalities that appear similar to aspects ofhuman disease (SchoÈn, 1999). Studies with spontaneous mousemutations known as chronic proliferative dermatitis (cpd)(HogenEsch et al, 1993) and ¯aky skin (fsn) suggest that epidermalhyperproliferation and mixed in¯ammatory in®ltrate with neu-trophilic accumulations in microabscesses (Sundberg et al, 1994) isnot dependent on T or B cell lymphocytes. Transgenic mice withtargeted epidermal overexpression of amphiregulin (Cook et al,1997), bone morphogenesis protein-6 (Blessing et al, 1996) orsuprabasal integrin expression (Carroll et al, 1995) likewise developa psoriasiform phenotype. On the other hand, the T cell-mediatedimmunopathogenesis of psoriasis has been well-established innumerous human and mouse studies (Krueger et al, 1995;Gottlieb et al, 1995; Wrone-Smith and Nickoloff, 1996; SchoÈn,1999). The aberrant expression of multiple cytokines suggests thatno single signal transduction cascade is responsible for thepsoriasiform dermatitis (SchoÈn, 1999). This study providescorroborating evidence of the pivotal nature of the NF-kBpathway in the cascade of events that lead to the development ofpsoriasis.

As lack of IkB-a results in constitutive NF-kB activity, increasedexpression of targets such as cytokines and adhesion molecules maycontribute to the skin disease. The interaction of cytokines oradhesion molecules with IkB-a±/± hematopoietic cells, includinglymphocytes may result in activation and recruitment of thelymphocytes into the epidermal and dermal regions. The interplaybetween the keratinocytes and activated lymphocytes may theninitiate keratinocyte hyperplasia as observed in the RAG2±/±, IkB-

VOL. 115, NO. 6 DECEMBER 2000 RAG2±/±, IkB-a±/± CHIMERAS 1131

Page 9: RAG2–/–, IκB-α–/– Chimeras Display a Psoriasiform Skin Disease

a±/± animals. We are currently designing experiments to test thismodel.

To further develop this model, highly ef®cient reconstitution ofchimeras by IkB-a±/± ES cells is required. The low ef®ciency ofreconstitution by the IkB-a±/± ES cells used in this study wasnoted. To circumvent this issue, we are generating IkB-a±/± EScells from 3.5 d IkB-a±/± blastocysts. Large numbers of chimeraswill be generated from these ES cells to study factors or adhesionmolecules, which are required for the interplay between keratino-cytes and immunocytes. These chimeras will be used to study bothtopical and systemically delivered therapeutic agents, which mayattenuate or reverse the psoriatic phenotype.

In summary, IkB-a±/± de®ciency results in a mouse model ofpsoriasiform dermatitis, which can be studied in vivo in adultanimals by variety of methods, including blastocyst complementa-tion, adoptive transfer, and skin grafting. The interplay between theimmune system and the keratinocytes is based on the resultsdescribed herein. Further studies will utilize this powerful system tostudy the physiologic role of cytokines and adhesion molecules inthe pathogenesis of psoriasis, and to develop the therapeutic agentsto treat this disease.

We thank Annapurna Venkatarishnam from the laboratory of Dr. Timothy

Blackwell at Vanderbilt University for providing labeled NF-kB probes for the

electrophoretic mobility shift assay. We would also like to thank Trish Labosky and

Bridget Hogan for providing ES cells and David Martin for technical advice. We are

grateful to Dr. M.H. Kosco-Vibois for generously providing anti-FDC-M1

antibody and to Drs. Singh and Dekoter for the training in the technique of fetal

liver cell transfer. Special thanks to members of the Kerr lab and to Drs. Earl Ruley,

Luc Van Kaer, Terry Dermody, Barney Graham, Roland Stein, Gene Oltz and

Mark Boothby at Vanderbilt University for helpful comments on the manuscript.

This work was supported by NIH grant R01 GM51249, a Center grant from the

National Cancer Institute (CA 68485), and P30AR41943.

REFERENCES

Alkalay I, Yaron A, Hatzubai A, et al: In vivo stimulation of I kappa Bphosphorylation is not suf®cient to activate NF-kappa B. Mol Cell Biol15:1294±1301, 1995

Arai K, Nishida J, Hayashida K, et al: Coordinate regulation of immune andin¯ammatory responses by cytokines. Rinsho Byori 38:347±353, 1990

Baldwin AS Jr: The NF-kappa B and I kappa B proteins: new discoveries andinsights. Annu Rev Immunol 14:649±683, 1996

Barker J: Psoriasis. J R Coll Phys London 31:238±240, 1997Baumann H, Gauldie J: The acute phase response. Immunol Today 15:74±80, 1994Beg AA, Finco TS, Nantermet PV, Baldwin AS Jr: Tumor necrosis factor and

interleukin-I lead to phosphorylation and loss of I kappa B alpha: a mechanismfor NF-kappa B activation. Mol Cell Biol 13:3301±3310, 1993

Beg AA, Sha WC, Bronson RT, Baltimore D: Constitutive NF-kappa B activation,enhanced granulopoiesis, and neonatal lethality in I kappa B alpha-de®cientmice. Genes Dev 9:2736±2746, 1995

Bendall HH, Sikes ML, Ballard DW, Oltz EM: an intact NF-kappa B signalingpathway is required for maintenance of mature B cell subsets. Mol Immunol36:187±195, 1999

Berliner JA, Navab M, Fogelman AM, et al: Atherosclerosis: basic mechanisms.Oxidation, in¯ammation, and genetics. Circulation 91:2488±2496, 1995

Blessing M, Schirmacher P, Kaiser S: Overexpression of bone morphogeneticprotein-6 (BMP-6) in the epidermis of transgenic mice: inhibition orstimulation of proliferation depending on the pattern of transgene expressionand formation of psoriatic lesions. J Cell Biol 135:227±239, 1996

Boothby MR, Mora AL, Scherer DC, Brockman JA, Ballard DW: Perturbation ofthe T lymphocyte lineage in transgenic mice expressing a constitutive repressorof nuclear factor (NF) -kappaB. J Exp Med 185:1897±1907, 1997

Buzy JM, Lindstrom LM, Zink MC, Clements JE: HIV-1 in the developing CNS.developmental differences in gene expression. Virology 210:361±371, 1995

Carroll JM, Romero MR, Watt FM: Suprabasal integrin expression in the epidermisof transgenic mice results in developmental defects and a phenotype resemblingpsoriasis. Cell 83:957±968, 1995

Chaturvedi V, Qin JZ, Denning MF, Choubey D, Diaz MO, Nickoloff BJ: apoptosisin proliferating, senescent, and immortalized keratinocytes. J Biol Chem274:23358±23367, 1999

Chen J: Analysis of gene function in lymphocytes by RAG-2-de®cient blastocystcomplementation. Adv Immunol 62:31±59, 1996

Chen F, Castranova V, Shi X, Demers LM: New insights into the role of nuclearfactor-kappaB, a ubiquitous transcription factor in the initiation of diseases.Clin Chem 45:7±17, 1999

Chen J, Lansford R, Stewart V, Young F, Alt FW: RAG-2-de®cient blastocystcomplementation: an assay of gene function in lymphocyte development. ProcNatl Acad Sci USA 90:4528±4532, 1993

Cheng J, Turksen K, Yu QC, Schreiber H, Teng M, Fuchs E: Cachexia and graft-vs.-host-disease-type skin changes in keratin promoter-driven TNF alphatransgenic mice. Genes Dev 6:1444±1456, 1992

Christophers E: The immunopathology of psoriasis. Int Arch Allergy Immunol110:199±206, 1996

Cook PW, Piepkorn M, Clegg CH, Plowman GD, DeMay JM, Brown JR,Pittelkow MR: Transgenic expression of the human amphiregulin geneinduces a psoriasis-like phenotype. J Clin Invest 100:2286±2294, 1997

Davidson NJ, Leach MW, Fort MM, et al: T helper cell I-type CD4+ T cells, but notB cells, mediate colitis in interleukin 10-de®cient mice. J Exp Med 184:241±251, 1996

Duvic M: Immunology of AIDS related to psoriasis. J Invest Dermatol 95:38S±40S,1990

Fraki JE, Briggaman RA, Lazarus GS: Transplantation of psoriatic skin onto nudemice. J Invest Dermatol 80(Suppl.):31±35s, 1983

Frucht DM, Lamperth L, Vicenzi E, Belcher JH, Martin MA: Ultraviolet radiationincreases HIV-long terminal repeat-directed expression in transgenic mice.AIDS Res Hum Retroviruses 7:729±733, 1991

Ghosh S, May MJ, Kopp EB: NF-kappa B and Rel proteins: evolutionarily conservedmediators of immune responses. Annu Rev Immunol 16:225±260, 1998

Gottlieb SL, Gilleaudeau P, Johnson R, Estes L, Woodworth TG, Gottlieb AB:Krueger JG: Response of psoriasis to a lymphocyte-selective toxin (DABS891L-2) suggests a primary immune, but not keratinocyte, pathogenic basis.Nat Med 1:442±447, 1995

Groves RW, Mizutani H, Kieffer JD, Kupper TS: In¯ammatory skin disease intransgenic mice that express high levels of interleukin 1 alpha in basalepidermis. Proc Natl Acad Sci USA 92:11874±11878, 1995

Handel ML, McMorrow LB, Gravallese EM: Nuclear factor-kappa B in rheumatoidsynovium. Localization of p50 and p65. Arthritis Rheum 38:1762±1770, 1995

Hogan B, Beddington R, Costantini F, Lacy E: Manipulating the Mouse Embryo: aLaboratory Manual. Cold Spring Harbor, NY: Cold Spring Harbor LaboratoryPress, 1986

HogenEsch H, Gijbels MJ, van Offerman E, Hooft J, Bekkum DW, Zurcher C: Aspontaneous mutation characterized by chronic proliferative dermatitis inC57BL mice. Am J Pathol 143:972±982, 1993

Ishikawa H, Claudio E, Dambach D, Raventos-Suarez C, Ryan C, Bravo R:Chronic in¯ammation and susceptibility to bacterial infections in mice lackingthe polypeptide (p)105 precursor (NF-kappaB1) but expressing p50. J Exp Med187:985±996, 1998

Johnson TM, Duvic M, Rapini RP, Rios A: AIDS exacerbates psoriasis [letter]. NEngl J Med 313:1415, 1985

Klement JF, Rice NR, Car BD, et al: IkappaBalpha de®ciency results in a sustainedNF-kappaB response and severe widespread dermatitis in mice. Mol Cell Biol16:2341±2349, 1996

Krueger GG, Manning DD, Malouf J, Ogden B: Long-term maintenance of psoriatichuman skin on congenitally athymic (nude) mice. J Invest Dermatol 64:307±312,1975

Krueger JG, Wolfe JT, Nabeya RT, et al: Successful ultraviolet B treatment ofpsoriasis is accompanied by a reversal of keratinocyte pathology and by selectivedepletion of intraepidermal T cells. J Exp Med 182:2057±2068, 1995

Li Q, Lu Q, Hwang JY, Buscher D, Lee KF, lzpisua-Belmonte JC, Verma IM:IKK1-de®cient mice exhibit abnormal development of skin and skeleton.Genes Dev 13:1322±1328, 1999

Liu SC, Parsons CS: Serial cultivation of epidermal keratinocytes from psoriaticplaques. J Invest Dermatol 81:54±61, 1983

Miagkov AV, Kovalenko DV, Brown CE, et al: NF-kappaB activation provides thepotential link between in¯ammation and hyperplasia in the arthritic joint. ProcNatl Acad Sci USA 95:13859±13864, 1998

Mortensen RM, Zubiaur M, Neer EJ, Seidman JG: Embryonic stem cells lacking afunctional inhibitory G-protein subunit (alpha i2) produced by gene targetingof both alleles. Proc Natl Acad Sci USA 88:7036±7040, 1991

Nakajima T, Kitajima I, Shin H, et al: Involvement of NF-kappa B activation inthrombin-induced human vascular smooth muscle cell proliferation. BiochemBiophys Res Commun 204:950±958, 1994

Nanney LB, Ellis DL, Levine J, King LE: Epidermal growth factor receptors inidiopathic and virally induced skin diseases. Am J Pathol 140:915±925, 1992

Nickoloff BJ, Mitra RS: Intraepidermal psoriatic cytokine network involves gammainterferon, transforming growth factor-alpha, and their cell surface receptors:dysregulation rather than de®ciency. J Invest Dermatol 99:882±883, 1992

Nickoloff BJ: The cytokine network in psoriasis. Arch Dermatol 127:871±884, 1991Nickoloff BJ: Pathogenesis and immunointervention strategies for psoriasis [In

Process Citation]. Mol Med Today 4:512±513, 1998Nickoloff BJ: The immunologic and genetic basis of psoriasis. Arch Dermatol

135:1104±1110, 1999Nickoloff BJ, Wrone-Smith T: animal models of psoriasis [letter]. Nat Med 3:475±

476, 1997Nickoloff BJ, Kunkel SL, Burdick M, Strieter RM: Severe combined

immunode®ciency mouse and human psoriatic skin chimeras. Validation of anew animal model. Am J Pathol 146:580±588, 1995

Nickoloff BJ, Wrone-Smith T, Bonish B, Porcelli SA: Response of murine andnormal human skin to injection of allogeneic blood-derived psoriaticimmunocytes: detection of T cells expressing receptors typically present onnatural killer cells, including CD94, CD158, and CD161. Arch Dermatol135:546±552, 1999

1132 CHEN ET AL THE JOURNAL OF INVESTIGATIVE DERMATOLOGY

Page 10: RAG2–/–, IκB-α–/– Chimeras Display a Psoriasiform Skin Disease

Schall TJ, Bacon K, Camp RD, Kaspari JW, Goeddel DV: Human macrophagein¯ammatory protein alpha (MIP-I alpha) and MIP-I beta chemokines attractdistinct populations of lymphocytes. J Exp Med 177:1821±1826, 1993

SchoÈn MP: Animal models of psoriasisÐwhat can we learn from them? J InvestDermatol 112:405±410, 1999

Seetharaman R, Mora AL, Nabozny G, Boothby M, Chen J: Essential role of T cellNF-kappa B activation in collagen-induced arthritis. J Immunol 163:1577±1583,1999

Seitz CS, Lin Q, Deng H, Khavari PA: Alterations in NF-kappaB function intransgenic epithelial tissue demonstrate a growth inhibitory role for NF-kappaB. Proc Natl Acad Sci USA 95:2307±2312, 1998

Sha WC: Regulation of immune responses by NF-kappa B/Rel transcription factor. JExp Med 187:143±146,1998, 1998

Sha WC, Liou HC, Tuomanen EI, Baltimore D: Targeted disruption of the p50subunit of NF-kappa B leads to multifocal defects in immune responses. Cell80:321±330, 1995

Shinkai Y, Rathbun G, Lam KP, et al: RAG-2-de®cient mice lack maturelymphocytes owing to inability to initiate V(D)J rearrangement. Cell 68:855±867, 1992

Singh H, Sen R, Baltimore D, Sharp PA: A nuclear factor that binds to a conservedsequence motif in transcriptional control elements of immunoglobulin genes.Nature 319:154±158, 1986

Sundberg JP, Dunstan RW, Roop DR, Beamer WG: Full-thickness skin grafts from¯aky skin mice to nude mice: maintenance of the psoriasiform phenotype. JInvest Dermatol 102:781±788, 1994

Thurberg BL, Collins T: The nuclear factor-kappa B/inhibitor of kappa Bautoregulatory system and atherosclerosis. Curr Opin Lipidol 9:387±396, 1998

Wrone-Smith T, Nickoloff BJ: Dermal injection of immunocytes induces psoriasis. JClin Invest 98:1878±1887, 1996

Wrone-Smith T, Mitra RS, Thompson CB, Jasty R, Castle VP, Nickoloff BJ:Keratinocytes derived from psoriatic plaques are resistant to apoptosis comparedwith normal skin. Am J Pathol 151:1321±1329, 1997

VOL. 115, NO. 6 DECEMBER 2000 RAG2±/±, IkB-a±/± CHIMERAS 1133