throbber
AantibodiesinSLE
`
`
`
`Anti-DN
`
`expert reviews
`in molecular medicme
`http://www—ermm.cbcu.cam.ac.uk
`”-
`
`Structure—function analysis and the molecular
`origins of anti-DNA antibodies in systemic lupus
`erythematosus
`
`— J
`
`atinderpal Kalsi, Chelliah T. Ravirajan, Anisur Rahman and
`David A. lsenberg
`
`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
`are often diagnostic and potentially pathogenic. In this review, we have examined
`the links between the structure and function of anti-dsDNA antibodies,
`emphasising their clinical associations. We have also reviewed studies involving
`animal models, the analysis of human antibody sequences and studies of, and
`using, computer modelling and crystal structure.
`
`Systemic lupus erythematosus (SLE or ’lupus’) is
`a major autoimmune rheumatic disease that is
`characterised clinically by photosensitive rashes
`and arthritis; in most cases the kidneys, lungs and
`central nervous system are affected by the disease.
`Its clinical diversity is, apparently, matched by its
`serological diversity (see Table 1; tabOOldil).
`Antibodies [immunoglobulins (Igs) produced by
`B lymphocytes (B cells)] that are found in patients
`with SLE appear to target a range of ’self antigens’
`(host—derived antigens). However, the diversity
`of the antibodies found in patients with SLE
`is actually rather narrow, given that
`a
`typical mammalian cell contains physiologically
`significant quantities of as many as 2000 different
`proteins, which are all potential self antigens
`(Ref. 1). For example, unlike patients with
`scleroderma (an autoimmune disease that is
`
`characterised by thickening of the skin), patients
`with SLE do not develop antibodies to the
`DNA-binding enzyme SCI 70 (also known as
`topoisomerase 1). Furthermore, the relatively
`restricted autoantibody profile that is typical of
`patients with SLE leads to the conclusion that
`random polyclonal activation of B cells cannot be
`solely responsible for the production of anti—
`DNA antibodies (Ref. 2). Precisely which of
`these SLE-associated autoantibodies are likely to
`be pathogenic remains to be determined, and
`represents a considerable challenge.
`
`Anti-dsDNA antibodies
`Among the autoantibodies that are present in the
`serum of patients with SLE, those that bind to
`dsDNA remain of paramount interest. These
`antibodies were first identified in the serum of
`
`
`David A. lsenberg (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 W1P 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 WIP 9PG, UK
`# 1
`Accession information: (99)OOO42-3a.pdf (short code: txt001 dil); 16 February 1999
`ISSN 1462-3994 ©1999 Cambridge University Press
`Celltrion v. Genentech
`IPR2017-01374
`Genentech Exhibit 2023
`
`

`

`expert reviews
`in molecular medicine
`
`http://www-ermmtcbcucam.ac.uk
`
`#T
`
`I.”
`able 1. Summary of the frequency of serum antibodies (and the antigens they are _|
`(I)
`detected in patients with systemic lupus
`y
`detected with) that are commonl
`thematosus (SLE) (tab001dil)

`ntib
`Fre u no of atie
`t
`WWW
`Series at the Bloomsbury
`Published studies
`Rheumatology Unitb
`
`Antigens the lgs bind to
`(Ab type)“
`
`(worldwide)
`
`
`
`Anti-DNAantibodies
`
`34 (for lgG);12 (for IgM)
`Not studied
`55
`21
`Not studied
`
`5 2
`
`5 (for lgG); 35 (for IgM)
`13
`14
`97
`20
`32
`10
`Not studied
`20
`
`Cardiolipin
`C1 q
`dsDNA
`Fc lgG (RF)
`Histone
`hsp 70
`hsp 90
`La
`LAC
`‘Nucleus' (ANA)
`RNP
`Ro
`Sm
`ssDNA
`Thyroid
`
`20—50
`20—45
`40—90
`~25
`30—80
`5—40
`5-50
`10—1 5
`10—20
`>90
`20—35
`30—40
`5—30
`s70
`$35
`
`a For further details about the antigens used. see Table 13 in Ref. 106.
`b The Bloomsbury Rheumatology Unit is at University College, London, UK.
`Abbreviations used: Ab = antibody; ANA = anti-nuclear antibody; C1q = a complement protein; dsDNA =
`double-stranded DNA; hsp 70 and hsp 90 = two types of human heat shock protein; histone = the protein
`associated with DNA; lg = immunoglobulin (antibody); LAC = lupus anti-coagulant; RF = rheumatoid factors,
`antibodies (usually IgM) that bind to lg and are produced by patients with rheumatoid arthritis but also with
`
`other diseases; ssDNA = single-stranded DNA; Sm, RNP. R0 and La = ribonuclear proteins; thyroid = an
`
`extract of human thyroid.
`
`patients with SLE over 40 years ago in four
`different laboratories (reviewed in Ref. 3). The
`likely involvement of these antibodies in the
`pathogenesis of human SLE, and in animal models
`of SLE, is indicated by (1) the close links between
`disease ’activity’ and levels of anti-dsDNA
`antibodies in the serum of many, though not all,
`patients; (2) the elution (removal and collection)
`of these antibodies from the kidneys of patients
`with SLE and lupus—prone mice; (3) direct
`evidence of these antibodies being associated with
`pathogenicity in isolated rat perfusion systems
`in which kidneys are dissected from the rat and
`their function is maintained artificially for a
`few hours) and mice with severe combined
`immunodeficiency (SCID); and (4) the fact that
`although antibodies to single—stranded DNA
`(ssDNA) are frequently found in the relatives of
`patients with SLE, those that bind to dsDNA are
`virtually never detected.
`
`Clinical studies
`Many papers published during the past 20
`years (reviewed by Spronk and colleagues in
`
`Ref. 4) have concluded that levels of anti-
`dsDNA antibodies [usually measured using
`enzyme-linked immunosorbent assays (ELISAS)
`or radioimmunoassays (RIAs)] generally reflect
`clinical disease activity. This observation appears
`to be particularly true of renal (kidney) disease,
`and most of the evidence that anti-dsDNA
`antibodies are pathogenic has been collected from
`studies of the kidney. Thus, high levels of anti-
`dsDNA antibodies, as measured using the Farr
`assay, which detects high—avidity antibodies
`(those that bind strongly to their target antigens),
`and low values of the complement enzyme
`marker CHSO were found predominantly in
`patients with lupus nephritis (i.e. who have
`kidney inflammation associated with SLE; Ref. 5).
`In contrast, antibodies to ssDNA are not specific
`for patients with SLE, being present, for example,
`in many individuals with infectious diseases.
`Lloyd and Schur (Ref. 6) found that complement
`depletion and raised titres of anti-dsDNA
`antibodies were associated more closely with
`renal than non-renal exacerbations in SLE. Ter
`Borg and colleagues (Ref. 7), in a prospective
`
`fA
`
`ccession information: (99)OOO42—3a.pdf (short code: txt001dil); 16 February 1999
`ISSN 1462-3994 @1999 Cambridge University Press
`
`

`

`http://www-ermm.cbcu.cam.ac.uk
`
`study of 72 patients, showed that active lupus
`nephritis was usually associated with high
`titres of anti-dsDNA antibodies. More
`recently, Okamura and colleagues (Ref. 8) have
`demonstrated a close relationship between renal
`disease activity (assessed by biopsy) and the
`isotype of Ig (e.g. IgG compared with IgM)
`produced that bound DNA: activity correlated
`with IgG reactive against dsDNAbut not with IgG
`reactive against ssDNA or with IgM reactive
`against either dsDNA or ssDNA. Bootsma and
`colleagues (Ref. 9), using the concept of a rise in
`levels of anti-dsDNA antibodies as a means of
`predicting a clinical relapse, showed that treating
`such patients with high levels of prednisolone (30
`mg/ day) reduced the relapse rate, compared with
`a control group, who were treated with either
`lower doses of prednisolone or no steroids.
`
`Assessment of disease activity in SLE
`A major problem with many of these earlier
`studies was the unsatisfactory nature of the
`indices of disease activity that were used to assess
`SLE. Validated and reliable global-score indices
`(i.e. score systems in which activity in each system
`is ’lumped together’) have been developed
`only in the past decade. However, with a
`disease as diverse as SLE, an activity index that
`demonstrates ’at a glance’ the degree of disease
`activity in each of the major organs or systems
`has obvious advantages. The British Isles Lupus
`Assessment Group (BILAG) has described and
`validated such a system, based on the ’physician’s
`intention to treat’ principle (Ref. 10). Thus, eight
`organs or systems are distinguished, allowing the
`easy correlation of activity in a particular organ
`or system with any serological marker. Using this
`system in a serial, longitudinal study of 14 Afro-
`Caribbean patients with SLE, antibodies to
`dsDNA were shown to correlate with renal
`disease, cardiopulmonary disease and global
`scores, but not with musculoskeletal, central
`nervous system or haematological involvement.
`Blood samples taken over periods of follow-up
`in the range of 3—15 years were analysed in this
`study (Ref. 11).
`
`Historical evidence that anti-dsDNA
`antibodies can be pathogenic in SLE
`Detection alone of anti-DNA antibodies in the
`kidneys of patients with SLE does not prove such
`antibodies were responsible for the development
`of this complication; however, it does place them
`
`
`
`Anti-DNAantibodiesinSLE
`
`expert reviews
`In molecular medicine
`
`at ’the scene of the crime’. Koffler and colleagues
`(Ref. 12) eluted IgG, complement and IgM from
`the kidneys of patients who had died from lupus
`nephritis. These eluates possessed anti-nuclear-
`binding activity; although a specific test for anti-
`DNA binding was not performed, the anti-
`nuclear-binding activity of the eluted antibodies
`could be partially inhibited by the addition of
`dsDNA, suggesting that at least some anti-dsDNA
`antibodies were present.
`In a study (Ref. 13) of over 40 families affected
`by SLE (where 21% of 147 relatives had antibodies
`to ssDNA), only two relatives had antibodies
`to dsDNA, strongly implicating anti-dsDNA
`antibodies, but not the anti-ssDNA antibodies, in
`the disease process (Ref. 14).
`
`Spontaneous disease models of SLE;
`pathogenicity of anti-DNA antibodles
`Some strains of mice spontaneously develop
`autoimmune disorders that have many of the
`features that are typical of human SLE (Ref. 15).
`The most commonly studied strains of mice with
`murine lupus are: (1) New Zealand black (N23),
`(2) BWF1 [NZB x New Zealand white (NZW) F1],
`(3) MRL/++, (4) MRL-lpr/lpr, (5) MRI. /1pr, (6)
`BXSB, and (7) SNFl [(NZB) x (SWR) F1] (for
`a comparison of their features see Table 2;
`tabOOZdil). These mice produce elevated levels of
`total Igs and autoantibodies (such as anti-dsDNA)
`and are thought to develop nephritis and arteritis
`as a result of deposition of immune complexes in
`the kidneys or arteries. Like patients with SLE,
`these mice present a diversity of disease patterns.
`In N213 and BWF1 mice, disease occurs primarily
`in the females. The disease in N23 mice is
`characterised mostly by a type of haemolytic
`anaemia that is Coombs positive (with anti~red-
`cell antibodies produced), and this can be
`associated with variable production of anti-
`nuclear antibodies (ANAs). The disease in
`BWF1 mice is characterised by the production
`of ANAs and immune-complewmediated
`glomerulonephritis (in their kidneys). In MRL/
`lpr mice, the lupus disease more equally affects
`male and female mice, whereas in BXSB mice, the
`disease affects males, because of the influence of
`a Y-linked gene (Ref. 15).
`
`The role of the lpr gene in mouse models
`of SLE
`MRL/ ++ and MRL/ lpr mice are used as models
`of SLE, and genetically differ at the lpr locus,
`
`Accession information: (99)OOO42-3a.pdf (short code: txt001dil); 16 February 1999
`ISSN 1462-3994 @1999 Cambridge University Press
`
`

`

`http://www-ermmcbcucam.ac.uk
`
`expert reviews
`in molecular medicine
`
`# T
`
`
`
`
`
`
`
`Anti-DNAantibodiesinSLE
`
`able 2. Features of disease in lupus-prone mouse models itab002dill
`
`
`Survival time for 50%
`of animals in typical
`
`
`Mouse strain;
`group; sex of animal Major immunological
`MHC haplotype
`Major clinical features
`most affected
`features
`
`
`New Zealand black
`Haemolytic anaemia,
`18 months; both sexes Production of anti-erythrocyte
`
`
`(NZB); H-2d
`glomerulonephritis,
`affected equally.
`antibodies, hyperproduction of
`lymphomas.
`IgM, generalised lymphocyte
`dysfunction.
`
`
`BWF1 [F1 of (N28
`Severe immune-complex— 7-8 months; females.
`Production of anti-nuclear and
`x New Zealand white mediated nephritis.
`anti-DNA antibodies,
`
`
`(NZW)]; H-2"’z
`generalised lymphocyte
`
`
`dysfunction.
`
`
`
`MRL-Ipr/lpr, H~2k
`Lymphoprollferatlon,
`2—4 months; females.
`Production of anti-nuclear
`
`
`immune-complex-mediated
`antibodies and rheumatoid
`nephritis. rheumatoid
`factors, proliferation of Ly1‘
`
`
`arthritis, vasculitls.
`cells“, generalised lymphocyte
`dysfunction.
`
`
`
`
`MRL'“; H-2k
`As for MRL-Ipr/Ipr but less
`18 months; females.
`As for MRL-lpr/Ipr but less
`severe.
`severe.
`
`
`
`
`
`BXSB; H-2h
`Haemolytic anaemia,
`2—4 months; males.
`Production of anti-DNA
`antibodies, anti-NTA and
`lymphadenopathy,
`
`
`
`
`glomerulonephrltis.
`anti-erythrocyte antibodies,
`thymic atrophy occurs earlier
`than normal.
`
`Moth-eaten; H-2b
`Hair loss, glomerulonephritis, 1 month; both sexes
`Production ofanti~DNA
`
`
`increased susceptibility
`affected equally.
`antibodies. anti-NTA and anti-
`to infections.
`erythrocyte antibodies,
`immunosuppression.
`
`
`
`Palmerston—North;
`Polyarteritis nodosa,
`11 months; females.
`Production of anti-DNA
`H-Z“
`immune-complex-mediated
`antibodies, hyperactivity of B
`
`
`nephritis.
`lymphocytes.
`
`
`Swan; H-2k
`Mild glomerulonephritis.
`18 months; both sexes Production of anti-DNA
`affected equally.
`antibodies. thymic atrophy
`occurs earlier than normal.
`
`
`SNF1; H-2il’d
`Severe glomerulonephritis.
`4—8 months; females.
`Production of anti-DNA and
`anti-nucleosome antibodies.
`
`“ Ly1‘ cells = mouse T lymphocytes (T cells) that have Ly1 antigens on their cell surface, a T—helper subset.
`
`Abbreviations used: H-2 = mouse major histocompatibility complex (MHC) class I; lgM = isotype of
`immunoglobulin (lg); NTA = natural thymocytotoxic antibody.
`
`
`
`syndrome with a much longer survival time of
`which is mutated in lpr mice and is an autosomal
`the mice. Congeneic mice on various genetic
`recessive gene (Ref. 16). In the homozygous state,
`backgrounds thathave two copies of the mutated
`lpr causes an immunological
`illness that
`lpr gene (such as C57BL/6-lpr/lpr, B6-lpr/lpr,
`is characterised by lymphoproliferation
`and accelerated autoimmunity. MRL/++ mice, C3H/He]~lpr/lpr and AKR-lpr/Zpr) all develop
`which have two copies the wild-type (unmutated)
`’spontaneous’ lymphoproliferation and produce
`gene at the lpr locus, develop an autoimmune
`autoantibodies. Despite the fact that anti—DNA
`f 4
`Accession information: (99)OOO42-3a.pdf (short code: txt001 dil); 16 February 1999
`ISSN 1462-3994 ©1999 Cambridge University Press
`
`

`

`expert reviews
`In molecular medicme
`
`downregulation of CD8 and persistence as
`CD4" CD8' T cells, which contribute to the
`lymphadenopathy. It should be noted that in the
`MRL/ [pr mice, the size of the thymus increased
`in parallel with the augmented activity of the Th
`T cells (Ref. 23).
`
`Anti-DNA antibodies in mice
`In mice, the genetic basis of the ability to produce
`anti-dsDNA antibodies 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 SNFl mouse, nephritis is linked to the I-Aq
`locus of the normal SWR parent, whereas in BWFl
`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 number of antibody-
`forming cells, including those that produce
`antibodies that bind to nuclear antigens. Although
`the B cells that are capable of producing the high-
`affinity anti-dsDNA antibodies are present in
`normal mice, they do not proliferate and actually
`produce such antibodies. Experimentally,
`to investigate the relationship between
`autoimmunity and Fas-mediated apoptosis (see
`below), using appropriate fusion-partner cells
`(Ref. 24), autoantibody-producing cells can be
`’immortalised’ to produce Igs in culture.
`
`http://www-ermm.cbcu.cam.ac.uk
`#m
`of CD4+ T cells and CD8" T cells appeared to
`antibodies are produced by, and found in, all of
`be normal (Ref. 22). Failure of apoptosis of
`the strains of mice, only MRL / lpr mice (with one
`self-reactive CD8+ T cells can then lead to
`wild-type and one mutant Zpr gene) develop
`severe immune-complex renal disease, which is
`responsible for the death of these mice by 6—9
`months of age. Therefore, the underlying or
`primary mechanism of autoimmunity in the MRL
`model lies in the MRL/ ++ background, and the
`lpr gene appears to act by accelerating and
`increasing the severity of the disease.
`The lpr gene is now known to encode a
`defective configuration of the Fas receptor
`(Ref. 17), the normal form of which mediates a
`signalling pathway that initiates apoptosis
`(programmed cell death; Ref. 18). Fas is a cell—
`surface protein that plays a major role in the
`induction of apoptosis in lymphoid cells. Mice
`with mutations in lpr and/ or lpr‘S, the gene(s)
`encoding Fas in mice (Ref. 19), are characterised
`by prolonged survival of B cells and other features
`that are similar to those found in patients with
`SLE, suggesting that in these animals interference
`with apoptosis might be important in the
`pathogenesis of their autoimmune disease (see
`below). Because of the involvement of lpr in
`apoptosis, which is important to T-cell function,
`we recently investigated whether MRL/lpr mice
`had increased activity 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.
`These T cells are either conventional CD4+ T cells
`or CD4' CD8‘ T cells (also known as ’double
`negative’ T cells). In MRL/ lpr mice, a failure of
`apoptosis of CD4+ T cells in the periphery of the
`thymus allows self-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/lpr mice compared with
`normal (non-autoimmune) BALB/ c mice at one
`month of age. At this age, the MRL/ lpr mice had
`low levels of serum anti-ssDNA antibodies and
`anti-dsDNA antibodies, and showed no detectable
`impairment in renal functions. This observation
`confirms that in MRL/ lpr mice, at one month,
`when the distribution of apoptotic cells was
`normal, there was no detectable lupus disease.
`Initially, in the MRL / lpr mice, there were no global
`defects in the negative selection of the T-cell
`repertoire, and the positive selection of subsets
`
`Control of anti-DNA antibodies in mice
`by T cells and cytokines
`T cells probably play an important role in these
`mouse models of SLE because experimental
`elimination of Th T cells in SNFl mice virtually
`prevented disease, and the removal of Th cells in
`BWFl mice also improved disease outcome (Ref.
`25). CD4+ T cells, which can induce B cells to
`secrete cationic IgG anti-dsDNA antibodies, have
`been cloned from SNFl mice, which develop
`an SLE-like autoimmunity (Ref. 26). Studies on
`the role of
`those T cells that express
`CD40 ligand (CD40L) in the development of
`glomerulonephritis in SNFI mice have shown that
`blocking the interaction between CD4OL on the
`pathogenic Th cells and CD40 on the lupus B cells,
`
`CD
`
`C m 0
`
`
`
`Anti-DNAantibod
`
`Accession information: (99)OOO42-3a.pdf (short code: txt001dll); 16 February 1999
`ISSN 1462—3994 @1999 Cambridge University Press
`
`

`

`expert reviews
`In mo|ecu|ar medicine
`
`http://www-ermm.cbcu.cam.ac.uk
`’ I.“
`cardiolipin and to nucleic acids (Fig. 1; figOOldil; _l
`at a particular window of time, delayed the
`Ref. 32). Together, these data suggest that DNA
`expansion of autoimmune memory B cells, and
`itself might not be the immunogen that is ‘0
`resulted in long-term therapeutic benefits in the
`responsible for inducing what are commonly :
`mice (Ref. 27). Thus, it is theoretically possible that
`known as anti-DNA antibodies.
`--
`patients with SLE might benefit from treatment
`with monoclonal antibodies that are 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 BWFl mice with an anti-IL-IO
`monoclonal antibody delayed the onset of
`autoimmune manifestations and the production
`of autoantibodies (Ref. 28). Similarly, treatment
`of BWFl mice with an anti-IL-6-receptor antibody
`lowered the production of IgG autoantibodies and
`suppressed the development of autoimmune
`disease (Ref. 29). Again, these data suggest that
`the use of appropriate monoclonal antibodies in
`patients with SLE might be beneficial (Ref. 30).
`
`8
`Non-mammalian DNA as an eliciting
`agent
`As reviewed elsewhere (Ref. 33), mammalian '5
`DNA is poorly immunogenic in mammals. One
`of the key questions, therefore, concerns the .0
`identity of the agent(s) responsible for eliciting j;
`the anti-DNA-antibody response in patients with c
`SLE. Several candidates have been suggested, and 5
`have been reviewed below.
`Experiments have shown that bacterial DNA <
`is able to induce strong antibody responses in
`mice. Gilkeson and colleagues (Refs 34, 35) have D
`proposed that bacterial DNA could be the
`I
`immunogen that induces the production of 1:
`antibodies that are reactive to DNA in patients :
`with SLE. When bacterial DNA was injected into
`healthy mice, antibodies thatlacked specificity for <
`mammalian DNA were mostly found; these
`antibodies lacked the molecular characteristics
`(notably arginine residues in the VH 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,
`polyoma virus, have produced data that are more
`promising. Hyperimmunisation of BALB/c mice
`(Ref. 36) with this virus has been shown to 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 shown that the injection
`of BK into young lupus-prone mice (N23 x NZW
`F1)before the natural onset of the disease induced
`a strong and persistent anti-dsDNA response,
`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 and anti-
`histone antibodies (histones are the protein core
`
`In vivo stimulation of anti-DNA antibody
`production
`The anti—DNA antibody 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 separated by three adjacent carbon atoms;
`see Fig. 1; figOOldil) on apparently diverse
`molecules such as phospholipids, proteins,
`polysaccharides and cell-membrane structures
`(reviewed in 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
`polynucleotides that 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-DNA antibodies.
`This polyreactivity of DNA antibodies could
`be due to multiple binding sites in variable regions
`in individual Igs or to the same (or similar)
`epitopes in different antigens. Furthermore,
`hybridoma cells (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: txt001dll); 16 February 1999
`ISSN 1462-3994 @1999 Cambrldge University Press
`
`

`

`http://www-ermm.cbcu.cam.ac.uk
`
`expert reviews
`in molecular medicine
`
`f
`
`
`
`Anti-DNAantibodiesinSLE
`
`
`
`Phosphodiester
`group
`
`
`Putative
`shared epitope
`for anti-DNA-
`antibody binding
`
`Three adjacent
`carbon atoms
`
`
`
`
`
`
`
`
`9H2
`9H2
`9
`
`
`
`
`
`
`
`|
`C\
`H?\C/ H
`9 H?
`R
`
`Single strand of DNA
`
`|
`O
`H20\ O‘ICI‘R
`
`Cardiolipin (phospholipid)
`
`
`
`
`Potential shared epitope between DNA and cardiolipin
`Expert Reviews in Molecular Medicine @1999 Cambridge University Press
`Figure 1. Potential shared epitope between DNA and cardiollpin.AntivDNA antibodies found in the serum
`of patients with systemic lupus erythematosus (SLE) have been shown to bind to many antigenic targets
`including single- and double-stranded DNA (ssDNA and dsDNA) and phosphclipids 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 epitope in their primary sequence, consisting
`of one or more phosphodiesler groups (Ref. 107) separated by three adjacent carbon atoms (shown here for
`ssDNA and cardiolipin). Abbreviation used: R = side chain. Modified from Ref. 107 (fi9001dil).
`
`around which dsDNA ’wraps’). They also
`provided further evidence that DNA alone is
`not immunogenic in Vivo, thus adding support
`to the increasingly popular theory that DNA Nucleic—acid—protein conjugates as
`that is complexed to histones in the form of
`eliciting agents
`nucleosomes is the key immunogenic particle Nucleosomes are the fundamental repeating units
`f 7
`Accession information: (99)OOO42-3a.pdf (short code: txtOO‘ldil); 16 February 1999
`ISSN 1462-3994 01999 Cambridge University Press
`
`responsible for the production of anti—DNA
`antibodies.
`
`

`

`expert reviews
`in molecular medicine
`
`
`
`-DNAantibodiesinSL
`
`http://www-ermm.cbcu.cam.ac.uk
`#m
`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 autoimmune diseases, such as
`(Ref. 39). Bell and colleagues (Ref. 40) have
`Sm-D antigens, ubiquitinated H2A antigen and
`demonstrated that B-cell hybridoma cells from
`poly(ADP-ribose) antigens (Ref. 48). Recently, we
`patients with SLE in culture released nucleosomes
`have shown that, in the mouse strain MRL/ lpr,
`that stimulated proliferation and lg synthesis in
`which is genetically susceptible to lupus, the
`normal B cells of mice.
`injection of as little as 1 rig of mouse histone-RNA
`In mouse models of SLE, the onset of the
`complex induced increased antibody responses to
`autoimmune response has been shown to be
`murine histones, dsDNA and ubiquifinated H2A,
`characterised by the early emergence of antibodies
`and exaggerated the disease in vivo in young mice
`that recognise conformational epitopes of the
`(Ref. 49).
`nucleosome particles (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 and the
`addition, in mice with lupus, nucleosomes were
`Fus-l protein from the parasite Trypanosoma cruzi
`a major immunogen for inducing Th T cells that
`(Ref. 50). However, the significance of these
`could stimulate pathogenic autoantibodies
`observations to the aetiology of human SLE
`(Ref. 43).
`remains uncertain.
`Our research group has produced from one
`patient with SLE a panel of five IgG (and five IgM)
`human hybridoma-derived monoclonal anti-
`DNA antibodies; two of these IgG antibodies
`bound to both dsDNA and histones, whereas none
`of the IgM antibodies did (Ref. 44). Moreover, the
`binding of these human monoclonal antibodies
`to histones was enhanced by the presence of DNA,
`which was also detectable in the supernatants of
`the hybridomas. In a subsequent study, we
`analysed the pathogenic properties of monoclonal
`human hybridoma anti-DNA lgG antibodies by
`implanting and growing the hybridoma-secreting
`cells in the peritoneal cavity of SCID mice. Only
`the IgG antibodies thatbound to both dsDNA and
`histones (or nucleosomes) deposited in the renal
`glomeruli of the mice, producing renal damage
`(Refs 45, 46). These data imply that some anti-
`dsDNA antibodies have the capacity to bind
`directly to the renal tissue and cause damage,
`perhaps without the help of complement.
`Conjugates of nucleic acid and proteins, where
`the nucleic acids can be DNA or other nucleic
`acids, and the proteins can be histone or other
`proteins, can also induce anti-DNA antibodies. For
`example, in normal, non-SLE-susceptible mice,
`bacterial DNA that is complexed to methylated
`bovine serum albumin (BSA) in a suitable
`adjuvant can induce the production of high titres
`of IgG anti-DNA antibodies (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 potential to elicit an autoimmune response
`in a human or animal that is exposed to it. Indeed,
`Diamond and colleagues (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, which is a
`component of the bacterial cell wall. Although the
`antibody 99D.7E provided partial protection
`against the microorganism, it was pathogenic to
`the kidney. It is also possible that immune
`responses might arise against a particular
`component because of its close association with
`another component. Rabbits that were immunised
`with purified rabbit RNA polymerase 1 (Ref. 53)
`produced antibodies that were reactive against the
`RNA polymerase 1 enzyme and also rabbit 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 polymerase 1
`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).
`
`1:
`
`c <
`
`Accession information: (99)OOO42-3a.pdf (short code: txtOO‘i dll); 16 February 1999
`ISSN 1462-3994 @1999 Cambridge University Press
`
`

`

`http://www—ermm.cbcu.cam.ac.uk
`
`expert reviews
`in molecular medicine
`
`# O
`
`ther stimuli of anti-dsDNA-antibody
`production
`Abnormalities of the immune system in human
`SLE no doubt serve to enhance or perpetuate
`the anti-DNA-antibody response. It has been
`suggested that several other factors might also
`’assist’ in the generation of the autoantibody
`response.
`
`Nuclear DNA damage induced by phagocytes
`Activated
`phagocytic
`cells
`(such
`as
`polymorphonuclear leucocytes and macrophages)
`are capable of releasing highly reactive oxygen
`species (ROS), which might be able to penetrate
`cell membranes, and thereby interact with, and
`damage, nuclear DNA. Subsequent release of this
`’altered’ DNA might, in turn, stimulate the
`production of anti-DNA antibodies by B cells.
`This possibility was explored by Gordon
`and colleagues (Ref. 56), who compared 41
`patients with SLE with 37 people with other
`autoimmune diseases and 20 healthy controls.
`Serum antibodies to ’damaged' DNA induced by
`ROS from patients with SLE had a statistically
`greater capacity for binding than anti-DNA
`antibodies from

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