throbber
http://www-ermm.cbcu.cam.ac.uk
`
`expert reviews
`
`on molecular medicine
`
`w
`.J
`Structure-function analysis and the molecular
`
`
`UJ
`C:
`
`
`
`
`origins of anti-DNA antibodies in systemic lupus
`erythematosus
`fl>
`(1)
`
`·-
`
`·-"'C
`0
`
`
`
`
`Jatinderpal Kalsi, Chelliah T. Ravirajan, Anisur Rahman and
`.c
`David A. Isenberg
`
`
`
`
`
`Patients with the autoimmune rheumatic disease systemic lupus erythematosus
`�
`
`
`
`(SLE or 'lupus') develop a wide variety of clinical and serological manifestations
`
`
`
`
`
`including the presence of antibodies to double-stranded DNA (dsDNA), which
`<C
`
`
`
`
`
`
`are often diagnostic and potentially pathogenic. In this review, we have examined Z
`
`
`
`
`the links between the structure and function of anti-dsDNA antibodies, 't
`
`
`
`
`
`their clinical associations. We have also reviewed studies involving ;:;
`emphasising
`
`
`
`
`
`animal models, the analysis of human antibody sequences and studies of, and C:
`
`
`
`using, computer modelling and crystal structure.
`<C
`
`....
`
`·-
`
`Systemic lupus erythematosus (SLE or 'lupus') is
`
`
`
`characterised by thickening of the skin), patients
`
`
`
`
`a major autoimmune rheumatic disease that is
`
`
`with SLE do not develop antibodies to the
`
`
`
`characterised clinically by photosensitive rashes
`DNA-binding enzyme Sci 70 (also known as
`
`and arthritis; in most cases the kidneys, lungs and
`
`
`topoisomerase 1). Furthermore, the relatively
`
`
`
`central nervous system are affected by the disease.
`
`
`
`
`restricted autoantibody profile that is typical of
`
`
`
`
`Its clinical diversity is, apparently, matched by its
`
`
`patients with SLE leads to the conclusion that
`
`
`serological diversity (see Table l; tabOOldil).
`
`random polyclonal activation of B cells cannot be
`
`
`
`Antibodies [immunoglobulins (Igs) produced by
`
`
`
`solely responsible for the production of anti­
`
`
`
`B lymphocytes (B cells)] that are found in patients
`
`
`DNA antibodies (Ref. 2). Precisely which of
`
`
`with SLE appear to target a range of 'self antigens'
`
`
`
`these SLE-associated autoantibodies are likely to
`
`
`
`(host-derived antigens). However, the diversity
`
`
`
`be pathogenic remains to be determined, and
`
`
`of the antibodies found in patients with SLE
`
`
`represents a considerable challenge.
`
`is actually rather narrow, given that a
`
`Anti-dsDNA antibodies
`
`
`
`typical mammalian cell contains physiologically
`
`
`significant quantities of as many as 2000 different
`
`
`Among the autoantibodies that are present in the
`
`
`proteins, which are all potential self antigens
`
`serum of patients with SLE, those that bind to
`
`
`(Ref. 1). For example, unlike patients with
`
`
`dsDNA remain of paramount interest. T hese
`
`
`scleroderma (an autoimmune disease that is
`
`
`antibodies were first identified in the serum of
`
`David A. Isenberg (Corresponding author)
`
`
`ARC Diamond Jubilee Professor of Rheumatology at University College London, Centre for
`
`
`
`
`
`
`
`
`
`
`Rheumatology /Bloomsbury Rheumatology Unit, Department of Medicine, University College
`
`
`
`
`
`London, Arthur Stanley House, 50 Tottenham Street, London WlP 9PG, UK, Tel: +44 (0)171 380
`
`
`9230; Fax: +44 (0)171 380 9278; E-mail: d.isenberg@ucl.ac.uk;
`
`
`
`
`
`Departmental website: http://www.ucl.ac.uk/ medicine /bloomsbury-rheumatology /
`
`Jatinderpal Kalsi, Chelliah T. Ravirajan and Anisur Rahman
`
`
`
`
`Centre for Rheumatology /Bloomsbury Rheumatology Unit, Department of Medicine, University
`
`
`
`
`
`
`
`
`
`
`College London, Arthur Stanley House, 50 Tottenham Street, London WlP 9PG, UK
`
`1
`{99)00042-3a.pdf (short code: txt001 dil); 16 February 1999
`
`Accession information:
`ISSN 1462-3994 ©1999 Cambridge University Press
`Pfizer v. Genentech
`IPR2017-01488
`
`Genentech Exhibit 2023
`
`

`

`http://www-ermm.cbcu.cam.ac.uk
`
`expert reviews
`in motecular medicine
`
`
`! Table 1. Summary of the frequency of serum antibodies (and the antigens they are
`detected with) that are commonly detected in patients with systemic lupus
`erythematosus (SLE) (tab001dil)
`
`
`
`
`
`Not studied
`55
`21
`Not studied
`
`5 2
`
`5 (for IgG); 35 (for IgM)
`
`
`
`inSLE
`lesIn
`
`
`
`Anti-DNAantibod
`
`
`
`
`
`Frequencyofpatients studied who have detectable antibodies (%)
`Published studies
`Series at the Bloomsbury
`Antigens the Igs bind to
`
`
`(Ab type)? (worldwide)
`Rheumatology Unit®
`
`
` Cardiolipin
` 34 (for IgG);12 (for IgM)
`20-50
`20-45
`Ctq
`
`
`40-90
`dsDNA
`
`
`~25
`Fc IgG (RF)
`30-80
`Histone
`5-40
`hsp 70
`
`
`5-50
`hsp 90
`10-15
`La
`10-20
`LAC
`
`
`>90
`‘Nucleus’ (ANA)
`20-35
`RNP
`
`
`30-40
`Ro
`
`
`5-30
`sm
`
`
`Not studied
`<70
`ssDNA
`
`
`
`20
`$35
`Thyroid
`
`
`
`
`@ For further details about the antigens used, see Table 13 in Ref. 106.
`
`
`> The Bloomsbury Rheumatology Unit is at University College, London, UK.
`
`Abbreviations used: Ab = antibody; ANA = anti-nuclear antibody; C1q = a complementprotein; dsDNA =
`
`double-stranded DNA; hsp 70 and hsp 90 = two types of human heat shock protein; histone = the protein
`associated with DNA; Ig = immunoglobulin (antibody); LAC = lupus anti-coagulant; RF = rheumatoid factors,
`antibodies (usually IgM) that bind to Ig and are produced by patients with rheumatoid arthritis but also with
`
`
`other diseases; ssDNA = single-stranded DNA; Sm, RNP, Ro and La = ribonuclear proteins; thyroid = an
`
`
`extract of human thyroid.
`
`Ref. 4) have concluded that levels of anti-
`patients with SLE over 40 years ago in four
`different laboratories (reviewed in Ref. 3). The
`dsDNAantibodies [usually measured using
`likely involvement of these antibodies in the
`enzyme-linked immunosorbent assays (ELISAs)
`pathogenesis of humanSLE, and in animal models
`or radioimmunoassays (RIAs)|} generally reflect
`of SLE,is indicated by (1) the close links between
`clinical diseaseactivity. This observation appears
`to be particularly true of renal (kidney) disease,
`disease ‘activity’ and levels of anti-dsDNA
`antibodies in the serum of many, though notall,
`and most of the evidence that anti-dsDNA
`patients; (2) the elution (removal and collection)
`antibodies are pathogenic has been collected from
`studies of the kidney. Thus, high levels of anti-
`of these antibodies from the kidneys of patients
`with SLE and lupus-prone mice; (3) direct
`dsDNAantibodies, as measured using the Farr
`evidenceofthese antibodies being associated with
`assay, which detects high-avidity antibodies
`(those that bind strongly to their target antigens),
`pathogenicity in isolated rat perfusion systems
`in which kidneys are dissected from the rat and
`and low values of the complement enzyme
`their function is maintained artificially for a
`marker CH50 were found predominantly in
`patients with lupus nephritis (i.e. who have
`few hours) and mice with severe combined
`immunodeficiency (SCID); and (4) the fact that
`kidney inflammation associated with SLE;Ref. 5).
`although antibodies to single-stranded DNA
`In contrast, antibodies to ssDNAare notspecific
`for patients with SLE, being present, for example,
`(ssDNA)are frequently found in therelatives of
`patients with SLE, those that bind to dsDNA are
`in many individuals with infectious diseases.
`Lloyd and Schur (Ref. 6) found that complement
`virtually never detected.
`depletion and raised titres of anti-dsDNA
`Clinical studies
`antibodies were associated more closely with
`Many papers published during the past 20
`renal than non-renal exacerbations in SLE. Ter
`Borg and colleagues (Ref. 7), in a prospective
`years (reviewed by Spronk and colleagues in
`aSSSee
`Accession information: (99)00042-3a.pdf (short code: txtO01dil); 16 February 1999
`ISSN 1462-3994 ©1999 Cambridge University Press
`
`

`

`http://www-ermm,.cbcu.cam.ac,uk
`
`expert reviews
`in molecular medicine
`
`Anti-DNAantibod
`
`at ‘the scene ofthe crime’. Koffler and colleagues
`study of 72 patients, showed that active lupus
`nephritis was usually associated with high
`(Ref. 12) eluted IgG, complement and IgM from
`the kidneys of patients who had died from lupus
`titres of anti-dsDNA antibodies. More
`recently, Okamura and colleagues (Ref. 8) have
`nephritis. These eluates possessed anti-nuclear-
`demonstratedaclose relationship between renal
`bindingactivity; although a specific test for anti-
`disease activity (assessed by biopsy) and the
`DNAbinding was not performed, the anti-
`isotype of Ig (e.g. IgG compared with IgM)
`nuclear-binding activity of the eluted antibodies
`could be partially inhibited by the addition of
`produced that bound DNA:activity correlated
`with IgG reactive against dsDNAbutnot with IgG
`dsDNA,suggestingthat at least some anti-dsDNA
`reactive against ssDNA or with IgM reactive
`antibodies were present.
`In a study (Ref. 13) of over 40 families affected
`against either dsDNA or ssDNA. Bootsma and
`colleagues (Ref. 9), using the conceptof a rise in
`by SLE (where 21% of 147relatives had antibodies
`levels of anti-dsDNA antibodies as a means of
`to ssDNA), only tworelatives had antibodies
`predictingaclinical relapse, showedthattreating
`to dsDNA,strongly implicating anti-dsDNA
`such patients with highlevels of prednisolone (30
`antibodies, but not the anti-ssDNA antibodies, in
`the disease process (Ref. 14).
`mg/day) reduced the relapse rate, compared with
`a control group, who were treated with either
`lower doses of prednisolone or no steroids.
`
`Spontaneous disease modeis of SLE;
`pathogenicity of anti-DNA antibodies
`Assessment of disease activity in SLE
`Somestrains of mice spontaneously develop
`A major problem with many of these earlier
`autoimmune disorders that have many of the
`features that are typical of human SLE (Ref. 15).
`studies was the unsatisfactory nature of the
`The most commonly studiedstrains of mice with
`indices of disease activity that were usedto assess
`murine lupusare: (1) New Zealand black (NZB),
`SLE. Validated and reliable global-score indices
`(i.e. score systems in which activity in each system
`(2) BWF1 [NZB x New Zealand white (NZW) F1],
`(3) MRL/++, (4) MRL-Ipr/Ipr, (5) MRL/Ipr, (6)
`is ‘lumped together’) have been developed
`only in the past decade. However, with a
`BXSB, and (7) SNF1 [(NZB) x (SWR) F1] (for
`disease as diverse as SLE, an activity index that
`a comparison of their features see Table 2;
`tab002dil). These mice produceelevatedlevels of
`demonstrates ‘at a glance’ the degree of disease
`activity in each of the major organs or systems
`total Igs and autoantibodies (such as anti-dsDNA)
`and are thought to develop nephritis and arteritis
`has obvious advantages. The British Isles Lupus
`Assessment Group (BILAG) has described and
`as a result of deposition of immune complexesin
`the kidneys or arteries. Like patients with SLE,
`validated such a system, based on the ‘physician’s
`these mice presenta diversity of disease patterns.
`intention to treat’ principle (Ref. 10). Thus, eight
`organsor systemsare distinguished, allowing the
`In NZB and BWF1 mice, disease occurs primarily
`in the females. The disease in NZB mice is
`easy correlation of activity in a particular organ
`or system with any serological marker. Using this
`characterised mostly by a type of haemolytic
`anaemia that is Coombspositive (with anti-red-
`system inaserial, longitudinal study of 14 Afro-
`Caribbean patients with SLE, antibodies to
`cell antibodies produced), and this can be
`associated with variable production of anti-
`dsDNA were shownto correlate with renal
`nuclear antibodies (ANAs). The disease in
`disease, cardiopulmonary disease and global
`BWF1 mice is characterised by the production
`scores, but not with musculoskeletal, central
`of ANAs and immune-complex-mediated
`nervous system or haematological involvement.
`glomerulonephritis (in their kidneys). In MRL/
`Blood samples taken over periods of follow-up
`in the range of 3-15 years were analysed in this
`Ipr mice, the lupus disease more equally affects
`study (Ref. 11).
`male and female mice, whereas in BXSB mice, the
`disease affects males, because of the influence of
`a Y-linked gene (Ref. 15).
`
`Historical evidence that anti-dsDNA
`antibodies can be pathogenic in SLE
`Therole of the [pr gene in mouse models
`Detection alone of anti-DNA antibodies in the
`of SLE
`kidneys of patients with SLE does not prove such
`MRL/++ and MRL/Ipr mice are used as models
`antibodies were responsible for the development
`of this complication; however, it does place them
`of SLE, and genetically differ at the Ipr locus,
`—————SSSSSa ee
`Accession information: (99)00042-3a.pdf (short code: txtOO1dil); 16 February 1999
`ISSN 1462-3994 ©1999 Cambridge University Press
`
`”i
`
`esin
`
`
`
`

`

`expert reviews
`in molecular medicine
`http://www-ermm.cbcu.cam.ac.uk
`eeeSSSe
`Table 2. Features of disease in lupus-prone mouse models (tab002dil)
`
`
`
`Anti-DNAantibodiesinSLE
`
`Major immunological
`features
`
`Production of anti-erythrocyte
`antibodies, hyperproduction of
`IgM, generalised lymphocyte
`dysfunction.
`
`Production of anti-nuclear and
`anti-DNAantibodies,
`generalised lymphocyte
`dysfunction.
`
`Production of anti-nuclear
`antibodies and rheumatoid
`factors, proliferation of Ly1*
`cells?, generalised lymphocyte
`dysfunction.
`
`Production of anti-DNA
`antibodies, anti-NTA and
`anti-erythrocyte antibodies,
`thymic atrophy occurs earlier
`than normal.
`
`Production of anti-DNA
`antibodies, anti-NTA and anti-
`erythrocyte antibodies,
`immunosuppression.
`
`Production of anti-DNA
`antibodies, hyperactivity of B
`lymphocytes.
`
`
`
`Mousestrain;
`MHC haplotype
`
`New Zealand black
`(NZB); H-29
`
`Majorclinical features
`
`Haemolytic anaemia,
`glomerulonephritis,
`lymphomas.
`
`Survival time for 50%
`of animals in typical
`group; sex of animal
`mostaffected
`
`18 months; both sexes
`affected equally.
`
`Severe immune-complex-
`mediated nephritis.
`
`7-8 months; females.
`
`Lymphoproliferation,
`immune-complex-mediated
`nephritis, rheumatoid
`arthritis, vasculitis.
`
`2—4 months; females.
`
`BWF1 [F1 of (NZB
`x New Zealand white
`(NZW)]; H-2%
`
`MRL-/pr/ipr, H-2*
`
`MRL**; H-2*
`
`BXSB; H-2°
`
`As for MRL-/pr/pr but less
`severe.
`
`18 months; females.
`
`As for MRL-/pr/pr but less
`severe.
`
`Haemolytic anaemia,
`lymphadenopathy,
`glomerulonephritis.
`
`2-4 months; males.
`
`Moth-eaten; H-2°
`
`Hair loss, glomerulonephritis,
`increased susceptibility
`to infections.
`
`4 month; both sexes
`affected equally.
`
`Palmerston—North;
`H-29
`
`Polyarteritis nodosa,
`immune-complex-mediated
`nephritis.
`
`Swan; H-2*
`
`Mild glomerulonephritis.
`
`11 months; females.
`
`18 months; both sexes
`affected equally.
`
`Production of anti-DNA
`antibodies, thymic atrophy
`occurs earlier than normal.
`
`SNF 1; H-2:
`
`Severe glomerulonephritis.
`
`4-8 months; females.
`
`Production of anti-DNA and
`anti-nucleosome antibodies.
`
`@Ly1* cells = mouse T lymphocytes(Tcells) that have Ly1 antigens ontheir cell surface, a T-helper subset.
`Abbreviations used: H-2 = mouse major histocompatibility complex (MHC) class I; IgM = isotype of
`immunoglobulin (Ig); NTA = natural thymocytotoxic antibody.
`
`whichis mutated in lpr mice and is an autosomal
`recessive gene(Ref. 16). In the homozygousstate,
`lpr causes an immunological
`illness that
`is characterised by lymphoproliferation
`and accelerated autoimmunity. MRL/++ mice,
`which havetwocopiesthe wild-type (unmutated)
`gene at the lpr locus, develop an autoimmune
`
`syndrome with a much longer survival time of
`the mice. Congeneic mice on various genetic
`backgroundsthat have twocopiesof the mutated
`Ipr gene (such as C57BL/6-Ipr/Ipr, B6-Ipr/Ipr,
`C3H/HeJ-lpr/lpr and AKR-Ipr/Ipr) all develop
`‘spontaneous’ lymphoproliferation and produce
`autoantibodies. Despite the fact that anti-DNA
`
`Accession information: (99)00042-3a.pdf (short code: txtOO1dil); 16 February 1999
`ISSN 1462-3994 ©1999 Cambridge University Press
`
`

`

`expert reviews
`in molecular medicine
`
`
`
`Anti-DNAantibodiesinSLE
`
`http://www-ermm.cbcu.cam.ac.uk
`___EeeeEee
`of CD4* T cells and CD8* T cells appeared to
`antibodies are produced by, and foundin,all of
`be normal (Ref. 22). Failure of apoptosis of
`the strains of mice, only MRL/Ipr mice (with one
`self-reactive CD8* T cells can then lead to
`wild-type and one mutant lpr gene) develop
`downregulation of CD8 and persistence as
`severe immune-complex renal disease, which is
`CD4- CD8- T cells, which contribute to the
`responsible for the death of these mice by 6-9
`lymphadenopathy. It should be noted that in the
`monthsof age. Therefore, the underlying or
`MRL/lIpr mice, the size of the thymus increased
`primary mechanism of autoimmunity in the MRL
`in parallel with the augmentedactivity of the Th
`modellies in the MRL/++ background, and the
`T cells (Ref. 23).
`Ipr gene appears to act by accelerating and
`increasing the severity of the disease.
`The Ipr gene is now knownto encode a
`defective configuration of the Fas receptor
`(Ref. 17), the normal form of which mediates a
`signalling pathway that initiates apoptosis
`(programmedcell death; Ref. 18). Fas is a cell-
`surface protein that plays a majorrole in the
`induction of apoptosis in lymphoid cells. Mice
`with mutations in Ipr and/or Ipr®, the gene(s)
`encoding Fasin mice (Ref. 19), are characterised
`by prolongedsurvivalof B cells and otherfeatures
`that are similar to those found in patients with
`SLE, suggestingthat in these animals interference
`with apoptosis might be important in the
`pathogenesis of their autoimmune disease (see
`below). Because of the involvement of Ipr in
`apoptosis, which is important to T-cell function,
`werecently investigated whether MRL/Ipr mice
`hadincreasedactivity in the T-helper (Th) subset
`of T cells (Ref. 20). T cells in the lymph nodes (of
`both mice and humans) produce soluble factors
`that enhance B-cell activation and, in turn,
`cause the accumulation of large numbers of
`lymphocytes in the spleen and lymph nodes.
`TheseT cells are either conventional CD4* T cells
`or CD4 CD8T cells (also known as ‘double
`negative’ T cells). In MRL/Ipr mice, a failure of
`apoptosis of CD4' T cells in the periphery of the
`thymusallowsself-reactive T cells to persist and
`ultimately to drive autoantibody production
`by B cells (Ref. 21). Our studies revealed no
`significant difference in the extent of apoptosis in
`the organs of MRL/Ipr mice compared with
`normal (non-autoimmune) BALB/c mice at one
`monthof age. At this age, the MRL/Ipr mice had
`low levels of serum anti-ssDNAantibodies and
`anti-dsDNAantibodies, and showed nodetectable
`impairment in renal functions. This observation
`confirms that in MRL/Ipr mice, at one month,
`when the distribution of apoptotic cells was
`normal, there was no detectable lupus disease.
`Initially, in the MRL/Ipr mice, there were no global
`defects in the negative selection of the T-cell
`repertoire, and the positive selection of subsets
`
`Anti-DNAantibodies in mice
`In mice, the genetic basis of the ability to produce
`anti-dsDNAantibodies has not been completely
`defined. In most strains, nephritis is associated
`with the co-inheritance of several genes including
`major histocompatibility complex (MHC)genes.
`In the SNF1 mouse, nephritis is linked to the I-A‘
`locus of the normal SWRparent, whereas in BWF1
`mice, it is linked to the I-E® chain from the NZW
`parent, a different MHC gene.
`Each of these autoimmune mouse strains
`has a polyclonal B-cell hyperactivity (many
`clones of B cells are affected) that causes
`hypergammaglobulinaemia (high levels of IgG
`antibodies) and an increased numberof antibody-
`forming cells, including those that produce
`antibodiesthat bindto nuclear antigens. Although
`the B cells that are capable of producing the high-
`affinity anti-dsDNA antibodies are present in
`normal mice, they do notproliferate and actually
`produce such antibodies. Experimentally,
`to investigate the relationship between
`autoimmunity and Fas-mediated apoptosis(see
`below), using appropriate fusion-partnercells
`(Ref. 24), autoantibody-producingcells can be
`‘immortalised’ to produce Igs in culture.
`
`Control of anti-DNA antibodies in mice
`by T cells and cytokines
`T cells probably play an importantrole in these
`mouse models of SLE because experimental
`elimination of Th T cells in SNF1 mice virtually
`prevented disease, and the removalof Th cells in
`BWF1 micealso improved disease outcome(Ref.
`25). CD4* T cells, which can induce B cells to
`secrete cationic IgG anti-dsDNAantibodies, have
`been cloned from SNF1 mice, which develop
`an SLE-like autoimmunity (Ref. 26). Studies on
`the role of
`those T cells
`that express
`CD40 ligand (CD40L) in the developmentof
`glomerulonephritis in SNF1 mice have shownthat
`blocking the interaction between CD40L on the
`pathogenic Th cells and CD40 on the lupusB cells,
`
`Accession information: (99)00042-3a.pdf (short code: txtOO1dil); 16 February 1999
`ISSN 1462-3994 ©1999 Cambridge University Press
`
`

`

`http://www-ermm.cbcu.cam.ac.uk
`
`expert reviews
`in molecular medicine
`
`cardiolipin and to nucleic acids (Fig. 1; fig001dil;
`at a particular window of time, delayed the
`Ref. 32). Together, these data suggest that DNA
`expansion of autoimmune memoryBcells, and
`”/)
`itself might not be the immunogenthat is
`resulted in long-term therapeutic benefits in the
`responsible for inducing what are commonly
`mice (Ref. 27). Thus,it is theoretically possible that
`knownas anti-DNA antibodies.
`patients with SLE might benefit from treatment
`with monoclonal antibodiesthatare reactive with
`CD4 or CD40.
`Studies of mouse models of SLE have suggested
`a role for two cytokines, interleukin 6 (IL-6) and
`interleukin 10 (IL-10), in the pathophysiology of
`SLE. Treatment of BWF1 mice with an anti-IL-10
`monoclonal antibody delayed the onset of
`autoimmune manifestations and the production
`of autoantibodies (Ref. 28). Similarly, treatment
`of BWF1 mice with an anti-IL-6-receptor antibody
`loweredthe production of IgG autoantibodies and
`suppressed the development of autoimmune
`disease (Ref. 29). Again, these data suggest that
`the use of appropriate monoclonalantibodies in
`patients with SLE mightbe beneficial (Ref. 30).
`
`
`
`cAnti-DNAantibodies
`
`Non-mammalian DNAasaneliciting
`agent
`As reviewed elsewhere (Ref. 33), mammalian
`DNAis poorly immunogenic in mammals. One
`of the key questions, therefore, concerns the
`identity of the agent(s) responsible for eliciting
`the anti-DNA-antibody responsein patients with
`SLE.Several candidates have been suggested, and
`have been reviewed below.
`Experiments have shownthat bacterial DNA
`is able to induce strong antibody responses in
`mice. Gilkeson and colleagues (Refs 34, 35) have
`proposed that bacterial DNA could be the
`immunogen that induces the production of
`antibodies that are reactive to DNAin patients
`with SLE. When bacterial DNA wasinjected into
`healthy mice, antibodies that lacked specificity for
`mammalian DNA were mostly found; these
`antibodies lacked the molecular characteristics
`(notably arginine residues in the V,, CDR3
`region) that are typical of the anti-dsDNA
`antibodies that are found in patients with SLE
`(Ref. 35). Autoantibodies in the serum of
`patients with SLE can bind to a widely shared
`bacterial epitope (on DNA), though these
`antibodies were mainly directed against ssDNA
`rather than dsDNA.
`a human dsDNA
`Studies using BK,
`polyomavirus, have produceddata that are more
`promising. Hyperimmunisation of BALB/c mice
`(Ref. 36) with this virus has been shownto induce
`the production of anti-DNA antibodies, and
`analysis of the variable-region genes of the
`antibodies revealed that the induced antibodies
`resembled the anti-DNA antibodies that are
`characteristic of lupus in mice. Rekvig and
`colleagues (Ref. 37) have shownthatthe injection
`of BK into young lupus-prone mice (NZB x NZW
`F1) before the natural onsetof the disease induced
`a strong and persistent anti-dsDNAresponse,
`which is more typical of the response found in
`patients with SLE. In a subsequent report
`(Ref. 38), the same group showed that in vivo
`expression of a single DNA-binding protein,
`the polyoma virus T antigen, was sufficient to
`initiate the production of anti-dsDNA andanti-
`histone antibodies (histones are the protein core
`
`In vivo stimulation of anti-DNA antibody
`production
`The anti-DNAantibody response in SLE has been
`shown to be heterogeneous (Ref. 31). Serum-
`derived antibodies and monoclonal anti-DNA
`antibodies recognised epitopes that occur in
`different nucleic acids (such as phosphodiester
`groups separatedby three adjacent carbon atoms,
`see Fig. 1; fig001dil) on apparently diverse
`molecules such as phospholipids, proteins,
`polysaccharides and cell-membranestructures
`(reviewedin Ref. 31). These observations were not
`due either to non-specific binding of low-affinity
`antibodies or merely to a charge interaction,
`because it has
`also been shown that
`anti-DNA’ antibodies do not necessarily bind
`polynucleotidesthat have similar patterns of ionic
`charge (Ref. 31). Furthermore, IgG antibodies that
`have high affinity for DNA(Ref. 31) can show the
`same degree of polyreactivity (i.e. ability to react
`with different epitopes) as some low-affinity IgM
`anti-DNAantibodies.
`This polyreactivity of DNA antibodies could
`be due to multiple bindingsites in variable regions
`in individual Igs or to the same(or similar)
`epitopes in different antigens. Furthermore,
`hybridomacells (a fusion of antibody-producing
`cells and a transformed cell line) that were
`prepared from mice that had been immunised
`with conjugates of cardiolipin (a phospholipid)
`and protein have been shown to produce
`monoclonal antibodies that bound both to
`
`Accession information: (99)00042-3a.pdf (short code: txtOO1dil); 16 February 1999
`ISSN 1462-3994 ©1999 Cambridge University Press
`
`

`

`
`
`Anti-DNAantibodiesinSLE
`
`http://www-ermm.cbcu.cam.ac.uk
`
`expert reviews
`in molecular medicine
`
`Phosphodiester
`group
`
`Three adjacent
`carbon atoms
`
`Putative
`shared epitope
`for anti-DNA-
`antibody binding
`
`Expert Reviews in Molecular Medicine ©1999 Cambridge University Press
`
`C,
`
`]Ho” O. BASE
`His Cc’
`H
`Oo He
`R
`
`Gh
`
`u
`
`H2C\ 9_¢G-R
`
`Single strand of DNA
`
`Cardiolipin (phospholipid)
`
`Potential shared epitope between DNAandcardiolipin
`
`Figure 1. Potential shared epitope between DNA and cardiolipin. Anti-DNA antibodies found in the serum
`of patients with systemic lupus erythematosus (SLE) have been shownto bind to many antigenic targets
`including single- and double-stranded DNA (ssDNA and dsDNA)and phospholipids such as cardiolipin. In
`addition, mouse monoclonal antibodies from mice immunised with cardiolipin can bind to nucleic acids and
`cardiolipin (Ref. 32). This cross-reactivity might be due to a shared epitopein their primary sequence, consisting
`of one or more phosphodiester groups (Ref. 107) separated by three adjacent carbon atoms (shownherefor
`ssDNAandcardiolipin). Abbreviation used: R = side chain. Modified from Ref. 107 (fig001dil).
`
`around which dsDNA ‘wraps’). They also
`provided further evidence that DNAaloneis
`not immunogenic in vivo, thus adding support
`to the increasingly popular theory that DNA
`that is complexed to histones in the form of
`nucleosomesis the key immunogenicparticle
`
`responsible for the production of anti-DNA
`antibodies.
`
`Nucleic-acid—protein conjugates as
`eliciting agents
`Nucleosomesare the fundamental repeating units
`
`Accession information: (99)00042-3a.pdf (short code: txtOO1dil); 16 February 1999
`ISSN 1462-3994 ©1999 Cambridge University Press
`
`

`

`
`
`Anti-DNAantibodiesinSLE
`
`expert reviews
`in molecular medicine
`
`http://www-ermm.cbcu.cam.ac.uk
`EESeeeSS
`several mammalian histone peptides and also
`of chromatin; they are generated during apoptosis
`other components that are common target
`by internucleosomal cleavage of the chromatin
`antigens in autoimmunediseases, such as
`(Ref. 39). Bell and colleagues (Ref. 40) have
`Sm-D antigens, ubiquitinated H2A antigen and
`demonstrated that B-cell hybridomacells from
`poly(ADP-ribose)antigens (Ref. 48). Recently, we
`patients with SLEin culture released nucleosomes
`have shownthat, in the mouse strain MRL/Ipr,
`that stimulated proliferation and Ig synthesis in
`whichis genetically susceptible to lupus, the
`normalB cells of mice.
`injection of aslittle as 1 1g of mouse histone-RNA
`In mouse models of SLE, the onset of the
`complex induced increased antibody responsesto
`autoimmune response has been shown to be
`murine histones, dsDNAand ubiquitinated H2A,
`characterised by the early emergence of antibodies
`and exaggeratedthe disease in vivo in young mice
`that recognise conformational epitopes of the
`(Ref. 49).
`nucleosomeparticles (Refs 41, 42). As the immune
`Antibodies to dsDNA were also induced in
`response progressed, autoantibodies that reacted
`non-autoimmune mice by immunisation with
`to dsDNA or histones became apparent. In
`complexes formed from mammalian DNA andthe
`addition, in mice with lupus, nucleosomes were
`Fus-1 protein from the parasite Trypanosoma cruzi
`a major immunogenfor inducing ThTcells that
`(Ref. 50). However, the significance of these
`could stimulate pathogenic autoantibodies
`observations to the aetiology of human SLE
`(Ref. 43).
`remains uncertain.
`Ourresearch group has produced from one
`patient with SLE a paneloffive IgG (andfive IgM)
`human hybridoma-derived monoclonal anti-
`DNAantibodies; two of these IgG antibodies
`bound to both dsDNAandhistones, whereas none
`of the IgM antibodiesdid (Ref. 44). Moreover, the
`binding of these human monoclonal antibodies
`to histones was enhancedbythe presence of DNA,
`which wasalso detectable in the supernatants of
`the hybridomas. In a subsequent study, we
`analysedthe pathogenic properties of monoclonal
`human hybridoma anti-DNAIgG antibodies by
`implanting and growing the hybridoma-secreting
`cells in the peritoneal cavity of SCID mice. Only
`the IgG antibodies that boundto both dsDNAand
`histones (or nucleosomes) deposited in the renal
`glomeruli of the mice, producing renal damage
`(Refs 45, 46). These data imply that some anti-
`dsDNAantibodies have the capacity to bind
`directly to the renal tissue and cause damage,
`perhaps withoutthe help of complement.
`Conjugatesof nucleic acid andproteins, where
`the nucleic acids can be DNAorother nucleic
`acids, and the proteins can be histone or other
`proteins, can also induce anti-DNAantibodies. For
`example, in normal, non-SLE-susceptible mice,
`bacterial DNA that is complexed to methylated
`bovine serum albumin (BSA) in a suitable
`adjuvant can inducethe production of hightitres
`of IgG anti-DNAantibodies (Ref. 47). However,
`anti-DNA antibodies induced in this way do not
`bind mammalian dsDNA,despite their reactivity
`with mammalian histone proteins that are
`complexed with mammalian RNA. These IgG
`antibodies react with mammalian histones,
`
`Molecular mimicry and cross-reactivity
`A microorganism that possesses both foreign
`epitopes and epitopes that mimic autoantigens
`has the potentialto elicit an autoimmune response
`in.a humanor animal that is exposedto it. Indeed,
`Diamondandcolleagues(Refs 51, 52) have shown
`that autoantibodies were generated as part of the
`immune response to a bacterial immunogen.
`This group described 99D.7E, a cross-reactive
`monoclonal mouse IgG antibody that bound to
`both dsDNA and phosphorylcholine, whichis a
`componentofthe bacterial cell wall. Although the
`antibody 99D.7E provided partial protection
`against the microorganism, it was pathogenicto
`the kidney. It is also possible that immune
`responses might arise against a particular
`componentbecauseofits close association with
`another component. Rabbits that were immunised
`with purified rabbit RNA polymerase 1 (Ref. 53)
`producedantibodies that werereactive against the
`RNApolymerase 1 enzymeand alsorabbit nucleic
`acids.It is possible that these antibodies against
`nucleic acids were induced not by cross-reactive
`epitopes on RNA polymerase 1, but because the
`physical association between RNA polymerase1
`and nucleic acids rendered the nucleic acids
`immunogenic. A recent experimental model of
`lupus (Ref. 54) that used peptide immunisation
`of normal rabbits extols the concept of epitope
`spreading as one of the mechanisms that might
`generate anti-DNA antibodies. Our own work,
`however, does not substantiate this model
`(Ref. 55).
`
`Accession information: (99)00042-3a.pdf (short code: txtOO1dil); 16 February 1999
`ISSN 1462-3994 ©1999 Cambridge University Press
`
`

`

`Lu
`and
`
`ins
`lies
`
`
`
`Anti-DNAantibod
`
`codingfor Fasor the Fas ligands. However, levels
`of soluble Fas are elevated ina minority of patients
`whohave SLEonly (Ref. 57).
`Bcl-2 is one of a family of proto-oncogene
`products; the principal Bcl-2 protein is unusual
`among such products in that it enhances the
`survival of lymphoid cells by interfering
`with apoptosis, rather than by promoting cell
`proliferation. Much fundamental research work
`is in progress to explore how apoptosis is
`Nuclear DNA damage induced by phagocytes
`regulated; some members of the Bcl-2 family
`Activated
`phagocytic
`cells
`(such
`as
`enhance apoptosis whereasothers blockit (Ref.
`polymorphonuclear leucocytes and macrophages)
`57). Althoughnotall studies agree, it is probable
`are capable of releasing highly reactive oxygen
`that in patients with SLE, T cells, but notBcells,
`species (ROS), which might be able to penetrate
`overexpress Bcl-2, and that this overexpression
`cell membranes, and thereby interact with, and
`correlates with SLE disease activity (Ref. 57).
`damage, nuclear DNA. Subsequentrelease of this
`‘altered’ DNA might, in turn, stimula

This document is available on Docket Alarm but you must sign up to view it.


Or .

Accessing this document will incur an additional charge of $.

After purchase, you can access this document again without charge.

Accept $ Charge
throbber

Still Working On It

This document is taking longer than usual to download. This can happen if we need to contact the court directly to obtain the document and their servers are running slowly.

Give it another minute or two to complete, and then try the refresh button.

throbber

A few More Minutes ... Still Working

It can take up to 5 minutes for us to download a document if the court servers are running slowly.

Thank you for your continued patience.

This document could not be displayed.

We could not find this document within its docket. Please go back to the docket page and check the link. If that does not work, go back to the docket and refresh it to pull the newest information.

Your account does not support viewing this document.

You need a Paid Account to view this document. Click here to change your account type.

Your account does not support viewing this document.

Set your membership status to view this document.

With a Docket Alarm membership, you'll get a whole lot more, including:

  • Up-to-date information for this case.
  • Email alerts whenever there is an update.
  • Full text search for other cases.
  • Get email alerts whenever a new case matches your search.

Become a Member

One Moment Please

The filing “” is large (MB) and is being downloaded.

Please refresh this page in a few minutes to see if the filing has been downloaded. The filing will also be emailed to you when the download completes.

Your document is on its way!

If you do not receive the document in five minutes, contact support at support@docketalarm.com.

Sealed Document

We are unable to display this document, it may be under a court ordered seal.

If you have proper credentials to access the file, you may proceed directly to the court's system using your government issued username and password.


Access Government Site

We are redirecting you
to a mobile optimized page.





Document Unreadable or Corrupt

Refresh this Document
Go to the Docket

We are unable to display this document.

Refresh this Document
Go to the Docket