Lupus nephritis: Lessons from experimental animal models

Lupus nephritis: Lessons from experimental animal models

REVIEW ARTICLE Lupus nephritis: Lessons from experimental animal models C. J. PEUTZ-KOOTSTRA, E. DE HEER, Ph. J. HOEDEMAEKER, C. K. ABRASS, and J. A. ...

179KB Sizes 4 Downloads 178 Views

REVIEW ARTICLE Lupus nephritis: Lessons from experimental animal models C. J. PEUTZ-KOOTSTRA, E. DE HEER, Ph. J. HOEDEMAEKER, C. K. ABRASS, and J. A. BRUIJN UTRECHT and LEIDEN, THE NETHERLANDS, and SEATTLE, WASHINGTON

Lupus nephritis is a frequent and severe complication of SLE. In the last decades, animal models for SLE have been studied widely to investigate the immunopathology of this autoimmune disease because abnormalities can be studied and manipulated before clinical signs of the disease become apparent. In this review an overview is given of our current knowledge on the development of lupus nephritis, as derived from animal models, and a hypothetical pathway for the development of lupus nephritis is postulated. The relevance of the studies in experimental models in relationship with our knowledge of human SLE is discussed. (J Lab Clin Med 2001;137:244-60)

Abbreviations: GvHD = graft-versus-host disease; ICAM-1 = intercellular adhesion molecule 1; IgG = immunoglobulin G; IgM = immunoglobulin M; IL = interleukin; MHC = major histocompatibility complex; mRNA = messenger RNA; PDGF = platelet-derived growth factor; RANTES = regulated upon activation normal T cells expressed and secreted; SLE = systemic lupus erythematosus; TGF-β = transforming growth factor β; TNF-α = tumor necrosis factor α; TXA = thromboxane

S

ystemic lupus erythematosus is a chronic autoimmune disease affecting multiple organs that is characterized by periods of remittance and relapse. Genetic, immunologic, hormonal, infectious, and environmental factors are all involved in the development of the disease. Autoantibodies are almost invariably present in the circulation.1-3 In Northern Europe and North America approximately 1 of 750 people will have SLE, with 80% of the cases occurring

From the Departments of Pathology, Utrecht University Medical Center, Utrecht, and Leiden University Medical Center, Leiden; and the Department of Nephrology, Veteran Affairs Medical Center, Seattle. Submitted for publication May 25, 2000; revision submitted December 11, 2000; accepted December 15, 2000. Reprint requests: C. J. Peutz-Kootstra, MD, PhD, Department of Pathology, Utrecht University Medical Center, PO Box 85500, 3508 GA Utrecht, The Netherlands. Copyright © 2001 by Mosby, Inc. 0022-2143/2001 $35.00 + 0 5/1/113755 doi:10.1067/mlc.2001.113755 244

in women during their childbearing years.1 Patients with SLE show diverse manifestations of the disease. Dermatitis, polyarthritis, glomerulonephritis, and vasculitis, as well as pulmonary, cardiovascular, hematologic, and central nervous system involvement, may occur, either together or separately.4 Glomerulonephritis in the form of lupus nephritis becomes clinically apparent in 50% to 80% of patients with SLE, resulting in end-stage renal failure in 10% to 20% of patients.2,5,6 Inflammation of the glomerulus eventually results in protein leakage from the circulation into the urinary space (ie, the development of proteinuria). Lupus nephritis is characterized by the presence of immunoglobulins of almost all isotypes in the glomerulus in combination with the presence and activation of complement components. Electron-dense deposits (immune aggregates) are observed in the mesangium, as well as subendothelially and subepithelially along the glomerular capillary walls. Histologically, a spectrum of glomerular lesions occurs, ranging from no abnormalities or mild mesangial proliferation to full-blown proliferative and

J Lab Clin Med Volume 137, Number 4

Peutz-Kootstra et al

245

Table I. World Health Organization morphologic classification of lupus nephritis (modified) I.

Normal glomeruli A. No abnormalities (by all techniques) B. Normal, as determined by means of light microscopy, but deposits seen with electron microscopy or immunofluorescence microscopy

II.

Pure mesangial alterations (mesangiopathy) A. Mesangial widening, mild hypercellularity (+), or both B. Moderate hypercellularity

III.

Focal segmental glomerulononephritis (associated with mild or moderate mesangial alterations) A. Active necrotizing lesions B. Active and sclerosing lesions C. Sclerosing lesions

IV.

Diffuse glomerulonephritis (severe mesangial, endocapillary or mesangiocapillary proliferation, and/or extensive subendothelial deposits) A. Without segmental lesions B. With active necrotizing lesions C. With active and sclerosing lesions D. With sclerosing lesions

V.

Diffuse membranous glomerulonephritis A. Membranous glomerulopathy B. Associated with lesions of Category II (a or b)

VI.

Advancing sclerosing glomerulonephritis

crescentic glomerulonephritis. In its classification for renal involvement in SLE, the World Health Organization combines observations made with light microscopy, immunofluorescence, and electron microscopy (Table I).7 This classification has proven to be very useful in clinical practice because in renal biopsy specimens taken from then untreated patients, the World Health Organization classification correlates well with prognosis and response to further therapy.2,5,8,9 To investigate the mechanisms underlying the development of glomerulonephritis in patients with SLE, animal models are widely used. 10 In human SLE it is obvious that disease variables can be studied with noninvasive methods only in a retrospective way. In animal models it is possible to study and manipulate immunoregulatory abnormalities and glomerular alterations occurring before the clinical signs of glomerulonephritis become apparent. In this review an overview is given of the way in which renal disease develops in 3 different models of SLE, followed by a summary of the autoantibody reactivity toward various antigens in murine SLE and the mechanisms that may account for the observed B-cell hyperreactivity. How autoantibodies in the circulation contribute to immune complex formation in the glomerulus is subsequently discussed. After glomerular injury is inflicted by immunoglobulins, local alterations in the glomerulus result in further deterioration of renal function. A summary of such local glomerular alterations is given, emphasizing the role of the glomerular matrix in the disturbance of glomerular structure and function. The clinical impli-

cations of these new insights are discussed in the final section of the article. MODELS OF SLE

Table II11-22 lists the majority of murine models of SLE that are being studied at present, excluding those of drug-induced SLE and genetically engineered animals (transgenes and knockouts). In this review studies in NZB/W F1 mice and MRL/lpr mice and in mice with chronic GvHD are discussed because these 3 models have been most intensively studied and severe glomerulonephritis develops in these mice. Other animal models of SLE have been reviewed in depth elsewhere.3,10 Development of an SLE-like disease in MRL/lpr mice can be largely attributed to their lpr trait, resulting in marked lymphoproliferation with lymphadenopathy, splenomegaly, and hypergammaglobulinemia.10,11 Autoantibodies reactive with a number of different antigens have been detected (Table III).23-45 At the approximate age of 6 months, MRL/lpr mice have severe immune complex glomerulonephritis with mesangial and endothelial cell proliferation, occasional crescent formation, thickening of the glomerular capillary wall, and tubulointerstitial nephritis with vasculitis.10,23 In the glomeruli immunoglobulins (predominantly IgG2a and IgG324) and C3 are found. The mice die of endstage renal failure with azotemia and high levels of proteinuria approximately 3 months later.10 NZB/W F1 mice are obtained by crossing NZW mice with NZB mice. The F1 hybrid mice also have auto-

J Lab Clin Med April 2001

246 Peutz-Kootstra et al

Table II. Most commonly studied murine models of SLE

Spontaneous models Mice with lpr mutation MRL/MP-lpr/lpr mice (MRL/lpr) C3H-lpr/lpr mice Mice with New Zealand background NZBxNZW mice (NZB/W F1) NZBxSWR mice (SNF1) NZB mice (NZB) BXSB mice Palmerston North mice SWR x SJL F1 mice Induced models Chronic GvHD DBA/2 in C57BLI10*DBA/2 FI BALB/c in C57BL10*BALB/c F1 BALB.D2 in C57BL10*BALB.D2 F1 BALB/c in BALB/c*A/J F1 Host-versus-graft-disease BALB/c*A/J F1 in BALB/c 16/6 idiotype-bearing human anti-DNA mAb

Glomerulonephritis

Reference No.

Severe No

11 11

Severe Severe Mild Severe in males Moderate Mild

Severe Moderate Moderate Mild Moderate Moderate

12 13, 14 15 16 * 17

18 19 19 20 21 22

*Waller SE, Gray RH, Fulton M, Wigley RD, Schnitzer B. Palmerston North mice: a new animal model of systemic lupus erythematosus. J Lab Clin Med 1978;92:932-45.

antibodies reactive with a variety of antigens (Table III). Histologically, various alterations in the kidneys occur, including mesangial proliferation, thickening of the glomerular capillary wall, glomerulosclerosis, tubular atrophy, and interstitial inflammation.10,12,23 Immunoglobulins (predominantly IgG2a and IgG2b) and C3 are found in the glomeruli from 6 months of age onward.10,46 Mice die of end-stage renal failure with proteinuria and azotemia at the age of 6 to 12 months.10 Induction of chronic GvHD by means of injection of DBA/2 lymphocytes in (C57BL/10*DBA/2) F1 hybrids results in the development of a lupus-like disease characterized by the presence of autoantibodies in the circulation with a reactivity pattern reminiscent of SLE.18,26 Six to 8 weeks after disease induction, immunoglobulins of all IgG isotypes are detected in the glomeruli, with IgG1 as the most prominent.47 At the same time, mesangial proliferation, mesangial matrix expansion, and thickening of the glomerular basement membranes with spike formations occur. Glomerulosclerosis develops, and the mice die of end-stage renal failure with high proteinuria levels, ascites, and hyperlipidemia 3 months after disease induction.47,48 B-CELL ACTIVATION AND ANTIBODY PRODUCTION IN MURINE SLE

The presence in the circulation of autoantibodies with various specificities (Table III) suggests a major role

for polyclonal B-cell activation in the pathogenesis of SLE. Indeed, in various experimental models of immune complex glomerulonephritis, including models of lupus nephritis, polyclonal B-cell activation underlies the development of autoantibodies with subsequent glomerular disease.49 An antigen-independent polyclonal B-cell stimulation on the basis of repetetive structures in proteins, such as those present on the surface of pneumococcus or in lipopolysaccharides, leads to the presence of predominantly IgM-type autoantibodies in the circulation, which subside after several weeks.49,50 In contrast, in murine SLE autoantibodies associated with disease development, the so-called pathogenic autoantibodies are of another immunoglobulin isotype (IgG instead of IgM),51-53 have a higher affinity for certain antigens,54,55 and have somatic mutations in their immunoglobulin Vh gene regions.56-61 These findings indicate that factors associated with an antigen-dependent propagation of B-cell clones are required for the presence of pathogenic autoantibodies in the circulation. The concept that next to polyclonal B-cell stimulation additional triggers are necessary for the secretion of pathogenic autoantibodies by B cells in lupus nephritis has been studied and reviewed by others.62-64 Evidence for polyclonal B-cell activation was found in MRL/lpr mice,65 NZB/W F1 mice,65,66 and mice with chronic GvHD.51 Because of the lpr mutation, MRL/lpr mice lack expression of the Fas antigen, lead-

J Lab Clin Med Volume 137, Number 4

Peutz-Kootstra et al

247

Table III. Selection of observed reactivities of autoantibodies in murine SLE Mouse strain

MRL/lpr

NZB/W F1

GvHD

Nuclear antigens Double-stranded DNA Single-stranded DNA Histones Nucleosomes Sm P Transfer RNA

+23 +25 +27 +30 +32,33 –33 ND

+25 +23 +27 +31 –32,33 –33 +25

+26 +26 +28,29 +31 –32,33 –33 +34

Cytoskeletal proteins Myosin Actin Vimentin

+35 +35 +36

+35 +35 ND

ND ND ND

Cell surface proteins on Thymocytes Erythrocytes Renal cells

± 23,37 ± 23 +38

+23 +23 ND

+26 +26 +39

Circulating and cell-surface proteins Complement factors (Clq) Immunoglobulins

+40 +23

+40 +23

+ ND

Basement membrane proteins Proteoglycans Collagens Laminins Fibronectin

+41 +42 +43-45 ND

+41 ND ND ND

+41 +39 +39,45 +39

ND, Not determined.

ing to impaired apoptosis.67,68 This defective apoptosis subsequently leads to an increased number of activated B cells, which is not associated with an altered education of T cells in the thymus but probably results from a failure in peripheral tolerance.35,69-71 In NZB/W F1 mice a T cell–independent intrinsic B-cell defect most likely underlies the polyclonal B-cell activation.71,72 In addition, a lack of suppressor CD8+ T-cell activity may play a role in NZB/W F1 mice.73 In mice with chronic GvHD, a decreased number of functionally active CD8+ T cells in the donor inoculate, normally suppressing the alloreactive donor CD4+ T cells, is responsible for the polyclonal activation of host B cells.74,75 The additional triggers that direct B cells to secrete pathogenic autoantibodies may very well be provided by T cells. CD4+ T cells have the tools to induce B-cell proliferation and differentiation by secreting cytokines, such as IL-4, IL-6, and IL-10,76-79 and T cells play an important role in the development of murine SLE.80-84 The investigation of aberrant CD4+ T-cell activity has resulted in intriguing new insights into SLE.85-88 However, caution is required in regarding the CD4+ T cells as the sole initiators of B-cell differentiation in lupus because, for example, natural killer cells are involved

in the development of lupus as well,89-91 and these cells can stimulate autoantibody production by activated B cells.92 Evidence from several experimental models indicates that T-cell help with the expression of adhesion molecules involved in cognate T-B–cell interaction93,94 is essential for the generation of pathogenic autoantibodies. How B cells secreting these pathogenic autoantibodies are generated is a topic of current research. The development of these autoantibodies may, for example, be associated with diminished elimination or aberrant selection of self-reactive B cells in germinal centers,95,96 but it may also be associated with expression of novel disease-relevant autoantigens in the periphery. This so-called epitope spreading of the immune response has been described only for antinuclear autoantibodies in lupus nephritis.97,98 In a later section of this article, we will argue that alterations in the glomerular matrix might also drive B cells to secrete pathogenic autoantibodies. Cytokines may very well play a crucial role in initiating and maintaining lupus disease, and in both processes they can have protective, as well as diseaseaggravating, effects.99-101 To provide a simplified model for cytokine secretion by T cells, secretion pat-

248 Peutz-Kootstra et al

J Lab Clin Med April 2001

Fig 1. Schematic representation of a hypothetical pathway for the development of experimental lupus nephritis. GBM, Glomerular basement membrane.

terns can be divided into a TH1 type (IL-2 and interferon γ), which is primarily involved in delayed-type hypersensitivity responses, and a TH2 type (IL-4, IL-5, IL-6, and IL-10), which is primarily involved in antibody responses.102 Imposing this TH1/TH2 paradigm on lupus shows that disease development is not strictly driven by either TH1- or TH2-type cytokines in MRL/lpr103-106 and NZB/W F1 mice.104,107-111 Chronic GvHD, however, has unequivocally been shown to be a disease mediated by TH2-type cytokines.112,113 GLOMERULAR IMMUNE COMPLEX FORMATION IN LUPUS NEPHRITIS

Glomerular immune complex formation in lupus nephritis can be caused by immune complex trapping from the circulation, by binding of autoantibodies with antigens deposited in the glomerulus, by direct binding of autoantibodies to intrinsic glomerular antigens, or by a combination of these mechanisms.114-116 Antigen-independent trapping of immune complexes from the circulation in the glomerulus may depend, for instance, on the (positive) charge of the antibodies,117 on antigen-independent sticking to matrix components (eg, fibronectin), on reaction with immune-complex

binding proteins (eg, mesangial matrix protein 50/100),118 or on binding of immune complexes to Fc receptors present on glomerular mesangial cells.119 In addition, the site of the glomerulus where circulating immune complexes were trapped was found to be dependent on the ratio between antigens and antibodies, which is influenced by the affinity of the antibodies for the antigen.120 In murine serum sickness large complexes formed with antibodies having a high affinity for antigen are found predominantly subendothelially and in the mesangium.121 A more membranous type of nephropathy with immune complexes predominantly present subepithelially along the glomerular capillary wall develops when antibodies have a low affinity for the antigen.121 Although attempts were made to extrapolate these findings to the involvement of antinuclear autoantibodies in lupus nephritis, hypothesizing that complexes of DNA and anti-DNA were trapped in glomeruli from the circulation, evidence for the presence of DNA/anti-DNA complexes in the circulation of patients with SLE has never been found.122 However, complexes of DNA and histones (ie, nucleosomes) have been found in the circulation of patients with SLE,123 pointing toward a possible

J Lab Clin Med Volume 137, Number 4

role for circulating complexes composed of these nucleosomes and their antinucleosomal antibodies in glomerular immune complex aggregation. A second mechanism for immune complex formation in lupus nephritis is binding of autoantibodies to antigens planted in the glomerulus. Evidence for this hypothesis emerged when the reactivity of antinuclear autoantibodies with cell-surface antigens was found to be mediated by nuclear components.41,124,125 Berden et al showed that antinuclear autoantibodies bind in the glomerulus by means of nucleosomes (ie, complexes of DNA and histones).126,127 The observation that nucleosomes bind to the glomerular capillary wall is presumably related to their strong (charge-based) affinity for components of the glomerular basement membrane,128 such as collagen type IV,129 and the negatively charged heparan sulfate proteoglycans.41 Nucleosomal particles have been detected in the circulation of patients with SLE123; in MRL/lpr mice it was found that these particles were released in the circulation as a consequence of aberrant apoptosis.127 A third mechanism accounting for immune complex formation is a direct, antigen-specific interaction of autoantibodies with glomerular antigens. Components of the glomerular basement membrane, in particular laminin, were found to be targets of autoantibodies in MRL/lpr mice43-45,130 and in mice with chronic GvHD (see below).45,131,132 Furthermore, monoclonal autoantibodies derived from MRL/lpr mice are known to interact with antigens isolated from glomerular mesangial and endothelial cells and to bind in the glomerulus, where they cause distinct histologic abnormalities.38,133 The observations that autoantibodies in patients with SLE react with nuclear and cytoplasmic antigens that are crucial for protein synthesis and cell replication134 led to the intriguing hypothesis that these autoantibodies may interfere directly with vital cell functions.135-137 MEDIATORS OF GLOMERULAR INFLAMMATION

The formation or deposition of immune complexes in the glomerulus can by itself lead to glomerular dysfunction.138,139 However, glomerular damage on immune complex formation is thought to be a consequence of activation of cellular and humoral mediator systems.115 The fact that immunoglobulins may be present along the glomerular capillary wall in the absence of functional glomerular damage illustrates the complexity of mechanisms leading to glomerular malfunction.140-142 In MRL/lpr mice it has been shown that the presence of B cells is essential for the development of lupus nephritis.143 It is believed that the site of immune complex formation determines the type of the glomerular lesion: a membranous glomerulopathy develops when subepithelial immune complexes are present with

Peutz-Kootstra et al

249

decreased activation and influx of leukocytes, whereas a more proliferative type of glomerulopathy is observed with mesangial and subendothelial immune complexes.144 In experimental lupus nephritis autoantibodies with different reactivity patterns can induce different histologic lesions after passive transfer into naive mice.38,133 Knowledge of factors involved in glomerular inflammation in lupus nephritis is largely deduced from studies in animal models of glomerular diseases other than lupus nephritis and from cell culture studies. Complement activation in glomeruli occurs through the classical pathway after immune complex binding and plays an important role in immune complex– mediated glomerular disease in several experimental models. 145 Complement components are synthesized by many different cell types, including renal cells,145 and their expression is enhanced by immune complex binding146 and by various cytokines.145,147 In addition to the role of complement in the clearance of immune complexes, complement factors are instrumental in the initiation of glomerular inflammation. For example, in vitro studies have shown that C5b-9 (or the membrane attack complex) stimulates glomerular mesangial and epithelial cells to synthesize inflammatory mediators, such as IL-1, TNF-α, prostaglandin E2, and TXA2.148,149 Furthermore, complement factors can directly induce cytoskeletal alterations 150 and secretion of extracellular matrix components 151,152 in glomerular epithelial cells. In experimental lupus nephritis increased synthesis of complement components has been found in the renal cortex in association with deteriorated renal function.153 Damage caused by factors present in the glomerulus, such as immune complexes and complement components, may result in the production of leukocyte-attracting factors. Platelet-activating factor,154 leukotrienes,155 PDGF,156 TXA2, IL-8,157 prostanoids,154 glomerular macrophage colony-stimulating factor, 158 colonystimulating factor, 159 monocyte chemoattractant protein 1,158,160,161 and RANTES162 are chemotactic factors that can be produced by glomerular cells, and some of these may even alter glomerular cell phenotype. In addition to the expression of chemoattractants, resident glomerular cells can express adhesion molecules, such as selectins and ICAM-1, thereby contributing to the influx of leukocytes in the glomerulus.163-167 Increased expression of glomerular macrophage colonystimulating factor, colony-stimulating factor, vascular cell adhesion molecule 1, ICAM-1, RANTES, and osteopontin has been found in glomeruli of MRL/lpr mice,159,168-171 and increased ICAM-1 expression was found in glomeruli of mice with chronic GvHD.172 Studies that prevented the influx of leukocytes or eliminated their presence altogether showed that these

250 Peutz-Kootstra et al

cells are involved in the development of glomerulonephritis.173-177 In MRL/lpr and NZB/W F1 mice glomerular F4/80-positive macrophages are detected as glomerular disease develops153,178; however, in mice with chronic GvHD, they remain absent.179 By using different strain combinations of mice for the induction of chronic GvHD, we found that the presence of CD11a-, CD45-, and MHC class II–positive cells in the glomeruli correlates with the development of glomerulosclerosis in lupus nephritis.179 The precise identity of these intriguing cells and their functional role in the development of glomerulosclerosis in chronic GvHD still remain to be established. The most important effector mechanism of leukocytes in tissues is the secretion of mediators that alter the phenotype of local cells, resulting in changes of tissue structure and function. The involvement of these mediators in modulating glomerular function has been reviewed extensively by others. In brief, leukocytes secrete oxidants, eicosanoids, catalytic enzymes, and many different cytokines that affect the glomerulus.180 Secreted cytokines (eg, IL-1 and TNF-α) induce and enhance the expression of chemotactic factors and adhesion molecules by glomerular cells,162,181,182 finally leading to local alterations, such as glomerular matrix accumulation or mesangial proliferation. For example, TGF-β,183-185 basic fibroblast-like growth factor,186 PDGF,187,188 and TXA2189,190 are involved in glomerular matrix accumulation. Mediators affecting the proliferation of glomerular cells are, among others, PDGF, 187,188 basic fibroblast-like growth factor, 191 IL-1, 181 and IL-6.192 Most of these mediators can be secreted both by glomerular cells and by infiltrating leukocytes, thus exemplifying the delicate balance between normal glomerular function and the development of glomerular disease. In MRL/lpr and NZB/W mice, increased mRNA expression, protein expression, or both of IL-1, TNFα, TXA2, TGF-β, and PDGF has been found in the renal cortex.193-197 In mice with chronic GvHD, no glomerular expression of TGF-β1 mRNA could be detected.198 Systemic modulation of the effect of certain mediators has shown that IL-1, TXA2,199 IL6,106,110 IL-4,113 IL-10,108 endothelin,200 and nitric oxide201,202 can enhance the development of renal disease in murine SLE, whereas development of the disease is slowed down by the administration of prostaglandin E analogues.203 It should be kept in mind, however, that these factors may have systemic, as well as local, glomerular effects, as shown for IL-1.204-206 Therefore the results obtained with systemically administered substances with respect to their local glomerular effects in lupus nephritis should be interpreted with caution.

J Lab Clin Med April 2001

Several other contributors to the genesis of lupus nephritis need to be mentioned as well, although their roles will not be discussed in detail here. For instance, the interstitium is not an innocent bystander in the development of end-stage renal disease, as has been reviewed extensively by others.207-210 On the contrary, interstitial inflammation and tubular atrophy correlate well with deterioration of renal function in glomerulonephritis.211 In MRL/lpr mice and in NZB/W F1 mice interstitial nephritis and glomerular disease coincide10; however, in mice with chronic GvHD, interstitial leukocyte infiltrates are only found in mice with end-stage renal failure.142 Furthermore, the development of vasculitis, a severe complication of SLE, can contribute to renal failure in experimental lupus nephritis. MRL/lpr mice have systemic vasculitis with prominent involvement of the arteries in the kidneys.212,213 Interestingly, the development of glomerulonephritis and arteritis in these mice can be segregated genetically.213 Vasculitis can also occur in NZB/W F1 mice but is less severe and less common than in MRL/lpr mice.23,214,215 In mice with chronic GvHD, vasculitis is not a prominent histologic feature. In summary, the development of glomerular lesions in lupus nephritis is a multifactorial process in which glomerular immunoglobulins, complement, mediators secreted by leukocytes and by resident glomerular cells, and systemic factors are involved. ALTERATIONS IN THE GLOMERULAR MATRIX

The disturbed glomerular structure and function in lupus nephritis are associated with quantitative and qualitative alterations in the glomerular matrix, ultimately leading to the development of glomerulosclerosis and chronic renal failure. Extracellular matrix components can bind to each other, and they can affect various biologic functions, such as cell attachment, spreading, proliferation, and differentiation.216,217 The important role of extracellular matrix components in modulating tissue function is exemplified by their altered expression during glomerular development.218221 Physiologic turnover of the extracellular matrix is maintained by a balanced interplay between matrix synthesis and matrix degradation.222 The altered expression of matrix components during the development of glomerular disease and the underlying mechanisms have been studied and reviewed by us and others.223,224 Here alterations in the glomerular matrix, in relation to the development of lupus nephritis, will be considered. In experimental lupus nephritis expansion of the mesangial matrix and thickening of the glomerular capillary wall occur. In MRL/lpr mice the expression of heparan sulfate, the side chain of heparan sulfate proteoglycan, seems to be decreased in the glomerular cap-

J Lab Clin Med Volume 137, Number 4

illary walls as the disease progresses.225 In NZB/W mice remodeling of the glomerular matrix is associated with increased expression of the α1 chains of collagen types I, III, and IV and of laminin and heparan sulfate proteoglycan.200 During development of glomerular disease in these mice, an additional increase of tissue inhibitor of matrix metalloproteases and of matrix metalloproteases occurs, thus regulating matrix degradation.200 In mice with chronic GvHD, expansion of the mesangial matrix and thickening of the glomerular basement membranes are associated with increased glomerular expression of collagen type IV, laminin, and heparan sulfate proteoglycan at the mRNA and protein levels.48,226 Furthermore, qualitative alterations occur in these matrix components in domains involved in cell-matrix and matrix-matrix interactions223,227,228 and thus may play a prominent role in the development of renal failure.217,224 The alterations in kidney laminin expression will be discussed in more detail below because laminin is an important component of the glomerular basement membrane, and autoantibodies against laminin play a nephritogenic role in MRL/lpr mice and in mice with chronic GvHD.45,227,229 The laminins are a family of at least 11 trimeric glycoproteins composed of an α, a β, and a γ chain and are involved in modulating cell behavior in developmental, normal, and disease states. Laminin fragments can influence cell adhesion, differentiation, migration, and proliferation. Furthermore, laminins play an important role in maintaining tissue integrity by binding to other basement membrane components, such as nidogen and collagen type IV.230,231 During the development of chronic GvHD, laminin 1 protein expression is increased in the mesangium and in the glomerular basement membrane, which is preceded by increased steady-state mRNA levels for the laminin β1 and γ1 chains.48,226 By using monoclonal antibodies generated against proteolytically digested fragments of laminin 1 involved in matrix-matrix and cell-matrix interactions, it was found that, in this model, certain laminin epitopes that are present in the normal glomerular basement membrane during embryogenesis only219 reappear in the glomerular basement membrane as the disease progresses.227 This is at least in part due to altered expression of laminin chains because the glomerular expression of the laminin α1 chain increases, whereas expression of the β1 chain decreases and β2 and γ2 chain expression remains unaltered.229 Mechanisms that may be involved in this altered composition of the glomerular basement membrane are altered local matrix protein synthesis or degradation, redistribution of matrix components within the glomerulus, and also conformational folding of matrix components and

Peutz-Kootstra et al

251

their binding proteins in the glomerular capillary wall. As exemplified below, binding of autoantibodies in the glomerular capillary wall may affect its composition as well and thus the development of immune complex glomerulonephritis. During development of glomerular disease in mice with chronic GvHD (and in MRL/lpr mice), autoantibodies reactive with renal tubular epithelial antigens and in particular with dipeptidyl peptidase IV (CD26), which is involved in cell-matrix interaction, appear in the circulation. Glomerular binding of anti-CD26 autoantibodies results in a patchy distribution of CD26 on the surface of glomerular visceral epithelial cells in mice with chronic GvHD. This altered CD26 distribution may be involved in the detachment of the epithelial cells from the glomerular basement membrane and the increased proteinuria in these mice.232 Likewise, antilaminin autoantibodies are detected in sera and glomerular eluates of MRL/lpr mice and mice with chronic GvHD,44,45,131 and antiglomerular basement membrane autoantibodies are a conditio sine qua non for the full-blown development of lupus nephritis in mice with chronic GvHD.19,131,233 Recently, we found that autoantibodies react with only laminin chains in glomerular matrix extracts of diseased mice and that, as the disease progresses, the laminin chain reactivity of autoantibodies alters in conjunction with altered glomerular laminin chain expression in mice with chronic GvHD. In particular, autoantibodies reactive with the laminin α1 chain and with the lamininbinding molecule filamin appear in the circulation, whereas this epitope is neoexpressed in glomeruli of diseased mice.229 It is tempting to speculate that the appearance of the laminin α1 chain and filamin in the glomerulus has elicited epitope spreading and a maturation of antilaminin α1/antifilamin autoantibody– producing B cells in this model. Indeed, in HgCl2induced glomerulonephritis in rats, a model also associated with a polyclonal activation of B cells, antigen-driven formation of autoantibodies reactive with the P1 fragment of laminin occurs.234 How these novel glomerular epitopes can be presented to T cells, B cells, or both remains speculative at present; however, glomerular cells express proteins that are capable of modulating the immune response in lupus nephritis. In MRL/lpr mice glomerular expression of vascular cell adhesion molecule 1,168 ICAM-1,169 and MHC class II molecules235 is increased as lupus nephritis develops. Several investigators have shown that tubular epithelial and mesangial cells are capable of expressing MHC class II antigens and of interacting with T cells.236-240 Altogether, these findings raise the intriguing possibility that exposure of novel glomerular antigens may lead to specific T-cell activation and the secretion of patho-

252 Peutz-Kootstra et al

genic, antigen-specific autoantibodies by B cells. The identification of epitopes inducing pathogenic autoantibodies may ultimately lead to more specific immunotherapy, as has recently been shown for nucleosomal epitopes in NZB mice.241 CLINICAL IMPLICATIONS AND FUTURE PERSPECTIVES

The mechanisms for the development of experimental lupus nephritis discussed above and summarized in Fig 1 can be largely extrapolated to human SLE. Briefly, in human subjects SLE is also characterized by increased activation of B cells. Lymphocyte abnormalities and alterations in the cytokine secretion profile are associated with this increased B-cell activity and the generation of pathogenic autoantibodies.63,100,242,243 Autoantibodies reactive with the antigens listed in Table III can be detected in the circulation of patients with SLE.244,245 In particular, autoantibodies reactive with the extracellular matrix components fibronectin,246 heparan sulfate proteoglycans,247 and laminin248 are detectable in sera of patients with SLE. Immunoglobulins bind in the glomerulus, presumably according to the same mechanisms unraveled in experimental models for SLE. Leukocytes infiltrate glomeruli249 concomitantly with the expression of glomerular adhesion molecules250 and MHC class II molecules251 on glomerular cells. An increased expression of all three TGF-β isoforms and their receptors has been found on glomerular cells of patients with lupus nephritis, which correlated with increased fibronectin ED-A expression and with disease activity.252,253 Furthermore, increased levels of connective tissue growth factor,254 IL-4,255 IL4 receptor,255 and nitric oxide synthases256 have been reported in biopsy specimens of patients with lupus nephritis. With the expansion of the glomerular matrix, an increased expression of laminin, fibronectin, and collagen type IV is observed in glomerular disease,257,258 whereas expression of heparan sulfate in the glomerular capillary walls was decreased in lupus nephritis.259 However, at present, there is no gold standard to predict a relapse of glomerular disease in patients with SLE, and therapeutic interventions resulting in the prevention of glomerular disease are lacking. Studies described in this review may help to develop strategies aimed at the prevention of renal disease development in SLE. First, it is conceivable that predictive parameters for the redevelopment of renal disease may be found either systemically or in the glomerulus itself. Regarding serologic parameters, until now, attention was focused largely on the role of antinuclear autoantibodies. The detection of autoantibodies reactive with renal antigens, and especially the shift of autoantibody reactivity toward a more antigen-driven response, may turn out

J Lab Clin Med April 2001

to be a valuable predictive parameter as well. By means of immunohistochemistry and molecular biologic techniques, it becomes more and more feasible to gather other than histologic data from renal biopsy specimens when, by means of conventional histologic analysis, no abnormalities are detectable.260 In chronic GvHD we found that steady-state mRNA levels for matrix components are increased as early as 4 weeks after disease induction,236 and altered mRNA splicing was detectable 2 weeks after disease induction.228 Histologically, however, glomerular matrix expansion and hypercellularity are first detectable 6 to 8 weeks after disease induction.48 Interestingly, therapeutic intervention with antiadhesion molecule autoantibodies 2 weeks after disease induction122 or with cyclosporin up to 6 weeks after disease induction142 attenuated renal disease development, thus showing that the early assessment of glomerular abnormalities may result in the commencement of early therapies preventing the development of renal disease. Furthermore, the glomerular expression of adhesion molecules, leukocyte subsets, and cytokines may be predictive for the development of more severe or end-stage renal disease as well.179 These findings indicate that measuring glomerular steady-state mRNA levels, preferably by using quantitative techniques, and performing additional immunohistochemical analyses will provide novel predictive parameters for the development of renal disease to the pathologist and the clinician. The analysis of early glomerular biopsy specimens (ie, specimens from patients with SLE without clinical signs of renal involvement) may ultimately contribute to the preservation of renal function in these patients. At present, lupus nephritis is treated with aspecific, aggressive immunosuppressive agents only.261 Investigations with experimental models aimed at gaining insight into disease mechanisms may ultimately lead to the development of specific therapeutic strategies. Systemically, the development of pathogenic autoantibodies may be prevented by blocking T-B–cell interactions (eg, with antiadhesion molecule monoclonal antibodies).93,94,172 Also, cytokine secretion patterns can be successfully manipulated, leading to the prevention of glomerulonephritis in experimental SLE.111,113 Other potential therapeutic strategies aim at diminishing the presence of nucleosomes in the circulation or at inducing tolerance to epitopes involved in the generation of pathogenic autoantibodies. Of course, side effects of these strategies will have to be monitored carefully, such as the spleen enlargement observed in mice with GvHD treated with anti-CD11a/CD54 monoclonal antibodies.172 Therefore further studies investigating the mechanisms underlying B-cell activation and the development of pathogenic autoantibodies in experimental

J Lab Clin Med Volume 137, Number 4

lupus nephritis will be necessary before promising systemic interventions at an early stage in the development of renal disease in SLE can be introduced in the clinic. A second experimental but challenging field for the development of novel therapeutic strategies is the manipulation of mediators involved in local glomerular damage. In experimental SLE development of renal disease is attenuated by blocking, for example, the effects of IL-1, TXA2,199 IL-6,106,110 endothelin,262 and nitric oxide.201,202 These therapeutic agents are administered systemically. Whether their therapeutic effects were caused by modulation of systemic or local glomerular mechanisms still remains to be investigated. Newly developed techniques aimed at delivering proteins locally in the glomerulus (eg, by means of gene transfer studies)170,263 will help to further unravel the role of local glomerular inflammatory mediators and may provide us with local therapeutic means.

Peutz-Kootstra et al

14.

15. 16.

17.

18.

19.

20. REFERENCES

1. Mills JA. Medical progress: systemic lupus erythematosus. N Engl J Med 1994;330:1871-9. 2. Cameron JS. Lupus nephritis. J Am Soc Nephrol 1999;10: 413-24. 3. Foster MH. Relevance of systemic lupus erythematosus nephritis animal models to human disease. Semin Nephrol 1999;19:12-24. 4. Boumpas DT, Austin HA, Fessler BJ, Balow JE, Klippel JH, Lockshin MD. Systemic lupus erythematosus: emerging concepts. Part 1: Renal, neuropsychiatric, cardiovascular, pulmonary, and hematologic disease. Ann Intern Med 1995; 122:940-50. 5. Pollak VE, Pirani CL. Lupus nephritis: pathology, pathogenesis, clinicopathological correlations and prognosis. In: Wallace DJ, Hahn BH, editors. Dubois’ lupus erythematosus. Philadelphia: Lea and Febiger; 1993. p. 525-41. 6. Ginzler EM, Diamond HS, Weiner M, Schlesinger M, Fries JF, Wasner C, et al. A multicenter study of outcome in systemic lupus erythomatosus. I. Entry variables as predictors of prognosis. Arthritis Rheum 1982;25:601-11. 7. Churg J, Bernstein J, Glassock RJ. Lupus nephritis. In: Churg J, Bernstein J, Glassock RJ, editors. Renal disease: classification and atlas of glomerular diseases. New York: IgakuShoin; 1995. p. 151-79. 8. Appel GB, Valeri A. The course and treatment of lupus nephritis. Annu Rev Med 1994;45:525-37. 9. Kashgarian M, Hayslett JP. Renal involvement in systemic lupus erythematosus. In: Tisher CG, Brenner BM, editors. Renal pathology with clinical and functional correlations. Philadelphia: JP Lippincott; 1989. p. 380-408. 10. Hahn BH. Animal models of systemic lupus erythematosus. In: Wallace DJ, Hahn BH, editors. Dubois’ lupus erythematosus. Philadelphia: Lea and Ebiger; 1993. p. 157-77. 11. Kelley VE, Boths JB. Interaction of mutant lpr gene with background strain influences renal disease. Clin Immunol Immunopathol 1985;37:220-9. 12. Lambert PH, Dixon FJ. Pathogenesis of the glomerulonephritis of NZB/W mice. J Exp Med 1968;127:507-22. 13. Datta SK. A search for the underlying mechanisms of sys-

21.

22.

23.

24.

25.

26.

27.

28.

29.

30.

253

temic autoimmune disease in the NZBxSWR model. Clin Immunol Immunopathol 1989;51:141-56. Gavalchin J, Datta SK. The NZB x SWR model of lupus nephritis. II. Autoantibodies deposited in renal lesions show a distinctive and restricted idiotype diversity. J Immunol 1987;138:138-48. Howie JB, Helyer BJ. The immunology and pathology of NZB mice. Adv Immunol 1968;9:215-66. Murphy ED, Roths JB. A Y chromosome-associated factor in strain BXSB producing accelerated autoimmunitiy and lymphoproliferation. Arthritis Rheum 1979;22:1188-94. Vidal S, Gelpí C, Rodríguez-Sánchez JL. (SWR x SJL)F1 mice: a new model of lupus-like disease. J Exp Med 1994;179:1429-35. Bruijn JA, Bergijk EC, de Heer E, Fleuren GJ, Hoedemaeker PJ. Induction and progression of experimental lupus nephritis: exploration of a pathogenetic pathway. Kidney Int 1992;41:5-13. Sutmuller M, Ekstijn GL, Ouellette S, de Heer E, Bruijn JA. Non-MHC genes determine the development of lupus nephritis in H-2 identical mouse strains. Clin Exp Immunol 1996;106:265-72. Gelpi C, Rodriguez-Sanchez JL, Martinez MA, Craft J, Hardin JA. Murine graft-versus-host disease: a model for study of mechanisms that generate autoantibodies to ribonucleoproteins. J Immunol 1988;140:4160-6. Florquin S, Abramowicz D, Heer E, Bruijn JA, Doutrelepont JM, Goldman M, et al. Renal immunopathology in murine host-versus-graft disease. Kidney Int 1991;40:852-61. Mendlovic S, Brocke S, Shoenfeld Y, Ben-Bassat M, Meshorer A, Bakimer R, et al. Induction of a systemic lupus erythematosus-like disease in mice by a common human anti-DNA idiotype. Proc Natl Acad Sci U S A 1988;85: 2260-4. Andrews BS, Eisenberg RA, Theofilopoulos AN, Izui S, Wilson CD, McConahey PJ, et al. Spontaneous murine lupuslike syndromes: clinical and immunopathological manifestations in several strains. J Exp Med 1978;148:1198-215. Takahashi S, Nose M, Sasaki J, Yamamoto T, Kyogoku M. IgG3 production in MRL/lpr mice is responsible for development of lupus nephritis. J Immunol 1991;147:515-9. Banks TA, Babakhani F, Poulos BT, Duffy JJ, Kibler R. Characterization of cross-reactive anti-DNA autoantibodies in murine lupus. Immunol Invest 1993;22:229-48. Gleichmann E, van Elven EH, van der Veen JPW. A systemic lupus erythematosus (SLE)-like disease in mice induced by abnormal T-B cell cooperation. Preferential formation of autoantibodies characteristic of SLE. Eur J Immunol 1982; 12:152-9. Elouaai F, Lule J, Benoist H, Appolinaire-Pilipenko S, Atanassov C, Muller S, et al. Autoimmunity to histones, ubiquitin, and ubiquitinated histone H2A in NZB x NZW and MRL-lpr/lpr mice. Anti-histone antibodies are concentrated in glomerular eluates of lupus mice. Nephrol Dial Transplant 1994;9:362-6. Portanova JP, Claman HN, Kotzin BL. Autoimmunization in murine graft-vs-host disease I. Selective production of antibodies to histones and DNA. J Immunol 1985;135:3850-6. Meziere C, Stöckl F, Batsford S, Vogt A, Muller S. Antibodies to DNA, chromatin core particles and histones in mice with graft-versus-host disease and their involvement in glomerular injury. Clin Exp Immunol 1994;98:287-94. Amoura Z, Chabre H, Koutouzov S, Lotton C, Cabrespines A, Bach JF, et al. Nucleosome-restricted antibodies are

J Lab Clin Med April 2001

254 Peutz-Kootstra et al

31.

32.

33.

34.

35.

36.

37.

38.

39.

40.

41.

42.

43.

44.

45.

detected before anti-dsDNA and/or antihistone antibodies in serum of MRL-Mp lpr/lpr and +/+ mice, and are present in kidney eluates of lupus mice with proteinuria. Arthritis Rheum 1994;37:1684-8. Kramers C, Hylkema MN, Van Bruggen MCJ, van de Lagemaat R, Dijkman HB, Assmann KJ, et al. Anti-nucleosome antibodies complexed to nucleosomal antigens show antiDNA reactivity and bind to rat glomerular basement membrane in vivo. J Clin Invest 1994;94:568-77. Eisenberg RA, Tan EM, Dixon FJ. Presence of anti-Sm reactivity in autoimmune mouse strains. Growth Factors 1992;7: 289-96. van Dam AP, Meilof JF, van den Brink HG, Smeenk RJT. Fine specificities of anti-nuclear antibodies in murine models models of graft-versus-host disease. Clin Exp Immunol 1990;81:31-8. Gelpi C, Angeles Martinez M, Vidal S, Targoff IN, Rodriguez-Sanchez JL. Autoantibodies to transfer RNAassociated protein in a murine model of chronic graft versus host disease. J Immunol 1994;152:1989-99. Ishigatsubo Y, Steinberg AD, Klinman DM. Autoantibody production is associated with polyclonal B cell activation in autoimmune mice which express the lpr or gld genes. Eur J Immunol 1988;18:1089-94. Andre-Schwartz J, Datta SK, Shoenfeld Y, Isenberg DA, Stollar BD, Schwartz RS. Binding of cytoskeletal proteins by monoclonal anti-DNA lupus autoantibodies. Clin Immunol Immunopathol 1984;31:261-71. Surh CD, Gershwin ME, Ahmed A. A peripheral and central T cell antigen recognized by a monoclonal thymocytotoxic autoantibody from New Zealand black mice. J Immunol 1987;138:1421-8. D’Andrea DM, Coupaye-Gerard B, Kleyman TR, Foster MH, Madaio MP. Lupus autoantibodies interact directly with distinct glomerular and vascular cell surface antigens. Kidney Int 1996;49:1214-21. Bruijn JA, Hogendoorn PCW, Corver WE, van den Broek LJCM, Hoedemaeker PJ, Fleuren GJ. Pathogenesis of experimental lupus nephritis: a role for anti-basement membrane and anti-tubular brush border antibodies in murine chronic graft-versus-host disease. Clin Exp Immunol 1990;79:115-22. Hogarth MB, Norsworthy PJ, Allen P, Trinder PK, Loos M, Morley BJ, et al. Autoantibodies to the collagenous region of C1q occur in three strains of lupus-prone mice. Clin Exp Immunol 1996;104:241-6. Termaat RM, Brinkman K, Van Gompel F, van den Heuvel LP, Veerkamp JH, Smeenk RJ, et al. Cross-reactivity of monoclonal anti-DNA antibodies with heparan sulfate is mediated via bound DNA/histone complexes. J Autoimmun 1990;3:531-45. Bernstein KA, Di Valerio R, Lefkowith JB. Glomerular binding activity in MRL lpr serum consists of antibodies that bind to a DNA/histone/type IV collagen complex. J Immunol 1995;154:2424-33. Sabbaga J, Line SRP, Potocnjak P, Madaio MP. A murine nephritogenic monoclonal anti-DNA autoantibody binds directly to mouse laminin, the major non-collagenous protein component of the glomerular basement membrane. Eur J Immunol 1989;19:137-43. Foster MH, Sabbaga J, Line SRP, Thompson KS, Barrett KJ, Madaio MP. Molecular analysis of spontaneous nephrotropic anti-laminin antibodies in an autoimmune MRL-lpr/lpr mouse. J Immunol 1993;151:814-24. Termaat RM, Assmann KJM, Van Son JPHF, Dijkman

46. 47.

48.

49. 50.

51.

52.

53.

54.

55.

56.

57.

58.

59.

60.

61.

62.

HBPM, Koene RAP, Berden JHM. Antigen-specificity of antibodies bound to glomeruli of mice with systemic lupus erythematosus-like syndromes. Lab Invest 1993;68:164-73. Theophilopoulos AN, Dixon FJ. Murine models of systemic lupus erythematosus. Adv Immunol 1985;37:269-391. Bruijn JA, van Elven EH, Hogendoorn PCW, Corver WE, Hoedemaeker PJ, Fleuren GJ. Murine chronic graft-versushost disease as a model for lupus nephritis. Am J Pathol 1988;130:639-41. Bergijk EC, Munaut C, Baelde JJ, Prins F, Foidart JM, Hoedemaeker PJ, et al. A histologic study of the extracellular matrix during the development of glomerulosclerosis in murine chronic graft-versus-host disease. Am J Pathol 1992; 140:1147-56. Goldman M, Baran D, Druet P. Polyclonal activation and experimental nephropathies. Kidney Int 1988;34:141-50. Andersson J, Sjoeberg O, Moeller G. Induction of immunoglobulin and antibody synthesis in vitro by lipopolysaccharides. Eur J Immunol 1972;2:349. Rolink AG, Radaszkiewicz T, Melchers F. The autoantigenbinding B cell repertoires of normal and of chronically graftversus-host-diseased mice. J Exp Med 1987;165:1675-87. Aoki I, Aoki A, Otani M, Miyagi Y, Misugi K, Ishii N, et al. A correlation between IgG class antibody production and glomerulonephritis in the murine chronic graft-versus-host reaction. Clin Immunol Immunopathol 1992;63:34-8. Tsao BP, Ebling FM, Roman C, Panosian-Sahakian N, Calame K, Hahn BH. Structural characteristics of the variable regions of immunoglobulin genes encoding a pathogenic autoantibody in murine lupus. J Clin Invest 1990;85: 530-40. Hirose S, Wakiya M, Kawano-Nishi Y, Yi J, Sanokawa R, Taki S, et al. Somatic diversification and affinity maturation of IgM and IgG anti-DNA antibodies in murine lupus. Eur J Immunol 1993;23:2813-20. Pankewycz OG, Migliorini P, Madaio MP. Polyreactive autoantibodies are nephritogenic in murine lupus nephritis. J Immunol 1987;139:3287-94. Gangemi RMR, Singh AK, Barrett KJ. Independently derived IgG anti-DNA autoantibodies from two lupus-prone mouse strains express a VH gene that is not present in most murine strains. J Immunol 1993;151:4660-71. Eliat D, Hochberg M, Tron F, Jacob L, Bach J-F. The VH gene sequences of anti-DNA antibodies in two different strains of lupus-prone mice are highly related. Eur J Immunol 1989;19:1241-6. Katz MS, Foster MH, Madaio MP. Independently derived murine glomerular immune deposit-forming anti-DNA antibodies are encoded by near-identical VH gene sequences. J Clin Invest 1993;91:402-8. Foster MH, Cizman B, Madaio MP. Nephritogenic autoantibodies in systemic lupus erythematosus: immunochemical properties, mechanisms of immune deposition, and genetic origin. Lab Invest 1993;5:494-507. Madaio MP, Shlomchik MJ. Emerging concepts regarding B cells and autoantibodies in murine lupus nephritis: B cells have multiple roles; all antibodies are not equal. J Am Soc Nephrol 1996;7:387-96. Wloch MK, Alexander AL, Pippen AM, Pisetsky DS, Gilkeson GS. Differences in V kappa gene utilization and VH CDR3 sequence among anti-DNA from C3H-lpr mice and lupus mice with nephritis. Eur J Immunol 1996;26: 2225-33. Mohan C, Datta SK. Lupus: key pathogenic mechanisms and

J Lab Clin Med Volume 137, Number 4

contributing factors. Clin Immunol Immunopathol 1995;77: 209-20. 63. Klinman DM, Steinberg AD. Inquiry into murine and human lupus. Immunol Rev 1995;144:157-93. 64. Horwitz DA, Stohl W. T lymphocytes, cytokines, and immune regulation in systemic lupus erythematosus. In: Wallace DJ, Hahn BH, editors. Dubois’ lupus erythematosus. Philadelphia: Lea and Febiger; 1993. p. 83-96. 65. Klinman DM, Steinberg AD. Systemic autoimmune disease arises from polyclonal B cell activation. J Exp Med 1987; 165:1755-60. 66. Klinman DM. Polyclonal B cell activation in lupus-prone mice precedes and predicts the development of autoimmune disease. J Clin Invest 1990;86:1249-54. 67. Watanabe-Fukunaga R, Brannan CI, Copeland NG, Jenkins NA, Nagata S. Lymphoproliferation disorder in mice explained by defects in Fas antigen that mediates apoptosis. Nature 1992;356:314-7. 68. Adachi M, Watanabe-Fukunaga R, Nagata S. Aberrant transcription caused by the insertion of an early transposable element in an intron of the Fas antigen gene of lpr mice. Proc Natl Acad Sci U S A 1993;90:1756-60. 69. Reap EA, Leslie D, Abrahams M, Eisenberg RA, Cohen PL. Apoptosis abnormalities of splenic lymphocytes in autoimmune lpr and gld mice. J Immunol 1995;154:936-43. 70. Bossu P, Singer CG, Andres P, Ettinger R, Marshak-Rothstein A, Abbas AK. Mature CD4+ T lymphocytes from MRL/lpr mice are resistant to receptor-mediated tolerance and apoptosis. J Immunol 1993;151:7233-9. 71. Merino R, Iwamoto M, Fossati L, Izui S. Polyclonal B cell activation arises from different mechanisms in lupus-prone (NZB x NZW) F1 and MRL/MpJ-lpr/lpr mice. J Immunol 1993;151:6509-16. 72. Reininger L, Radaszkiewicz T, Kosco M, Melchers F, Rolink AG. Development of autoimmune disease in SCID mice populated with long-term ‘in vitro’ proliferating (NZBxNZW)F1 pre-B cells. J Exp Med 1992;176: 1343-53. 73. Krakauer R, Waldman TA, Strober W. Loss of suppressor T cells in adult NZB/NZW mice. J Exp Med 1976;144:662-73. 74. Via CS, Shearer GM. T-cell interactions in autoimmunity: insights from a murine model of graft-versus-host disease. Immunol Today 1988;9:207-13. 75. Rus V, Svetic A, Nguyen P, Gause WC, Via CS. Kinetics of Th1 and Th2 cytokine production during the early course of acute and chronic murine graft-versus-host disease: regulatory role of donor CD8+ T cells. J Immunol 1995;155:2396406. 76. Rizzo LV, DeKruyff RH, Umetsu DT. Generation of B cell memory and affinity maturation: Induction with Th1 and Th2 T cell clones. J Immunol 1992;148:3733-9. 77. Snapper CM, Finkelman FD, Paul WE. Regulation of IgG1 and IgE production by interleukin 4. Immunol Rev 1988;102:51-75. 78. van Snick J. Interleukin-6: an overview. Annu Rev Immunol 1990;8:253-78. 79. Shparago N, Zelazowski P, Jin L, McIntyre TM, Stuber E, Pecanha LM, et al. IL-10 selectively regulates murine Ig isotype switching. Int Immunol 1996;8:781-90. 80. Connolly K, Roubinian JR, Wofsy D. Development of murine lupus in CD4-depleted NZB/NZW mice. Sustained inhibition of residual CD4+ T cells is required to suppress autoimmunity. J Immunol 1992;149:3083-8. 81. Rozendaal L, Pals ST, Gleichmann E, Melief CJM. Persis-

Peutz-Kootstra et al

82.

83.

84.

85.

86.

87.

88.

89.

90.

91.

92.

93.

94.

95.

96.

97.

98.

255

tence of allospecific helper T cells is required for maintaining autoantibody formation in lupus-like graft-versushost disease. Clin Exp Immunol 1990;82:527-32. Santoro TJ, Portanova JP, Kotzin BL. The contribution of L3T4+ T cells to lymphoproliferation and autoantibody production in MRL-lpr/lpr mice. J Exp Med 1988;167: 1713-8. Wofsy D, Seaman WE. Successful treatment of autoimmunity in NZB/NZW F1 mice with monoclonal antibody to L3T4. J Exp Med 1985;161:378-91. Peng SL, Madaio MP, Hughes DP, Crispe IN, Owen MJ, Wen L, et al. Murine lupus in the absence of alpha beta T cells. J Immunol 1996;156:4041-9. Mohan C, Adams S, Stanik V, Datta SK. Nucleosome: a major immunogen for pathogenic autoantibody-inducing T cells of lupus. J Exp Med 1993;177:1367-81. de Alboran IM, Gonzalo JA, Kroemer G, Leonardo E, Marcos MAR, Martinez AC. Attenuation of autoimmune disease and lymphocyte accumulation in MRL/lpr mice by treatment with anti-Vβ8 antibodies. Eur J Immunol 1992;22:2153-8. Singh RR, Ebling FM, Sercarz EE, Hahn BH. Immune tolerance to autoantibody-derived peptides delays development of autoimmunity in murine lupus. J Clin Invest 1995; 96:2990-6. Ebling FM, Tsao BP, Singh RR, Sercarz E, Hahn BH. A peptide derived from an autoantibody can stimulate T cells in the (NZB X NZW)F1 mouse model of systemic lupus erythematosus. Arthritis Rheum 1993;36:355-64. Harada M, Lin T, Kurosawa S, Maeda T, Umesue M, Itoh O, et al. Natural killer cells inhibit the development of autoantibody production in (C57BL/6 x DBA/2) F1 hybrid mice injected with DBA/2 spleen cells. Cell Immunol 1995;161:42-9. Seaman WE, Sleisenger M, Eriksson E, Koo GC. Depletion of natural killer cells in mice by a monoclonal antibody to NK1.1. J Immunol 1987;138:4539-44. Karashima A, Taniguchi K, Himeno K, Kawano Y, Toshitani A, Nuomoto K. Does depression of NK activity cause lymphadenopathy in lpr mice? Cell Immunol 1988;115: 484-90. Snapper CM, Yamaguchi H, Moorman MA, Sneed R, Smoot D, Mond JJ. Natural killer cells induce activated murine B cells to secrete Ig. J Immunol 1993;151:525160. Nakajima A, Azuma M, Kodera S, Nuriya S, Terashi A, Abe M, et al. Preferential dependence of autoantibody production in murine lupus on CD86 costimulatory molecule. Eur J Immunol 1995;25:3060-9. Mohan C, Shi Y, Laman JD, Datta SK. Interaction between CD40 and its ligand gp39 in the development of murine lupus nephritis. J Immunol 1995;154:1470-80. Pulendran B, van Driel R, Nossal GJV. Immunological tolerance in germinal centres. Immunol Today 1997;18: 27-32. Elkon KB, Marshak-Rothstein A. B cells in systemic autoimmune disease: recent insights from Fas-deficient mice and men. Curr Opin Immunol 1996;8:852-9. Craft J, Fatenejad S. Self antigens and epitope spreading in systemic autoimmunity. Arthritis Rheum 1998;40: 1374-82. James JA, Harley JB. A model of peptide-induced lupus autoimmune B cell epitope spreading is strain specific and is not H2-restricted in mice. J Immunol 1998; 160:502-8.

256 Peutz-Kootstra et al

99. Handwerger BS, Rus V, Da Silva L, Via CS. The role of cytokines in the immunopathogenesis of lupus. Springer Semin Immunopathol 1994;16:153-80. 100. Horwitz DA, Jacob CO. The cytokine network in the pathogenesis of systemic lupus erythematosus and possible therapeutic implications. Springer Semin Immunopathol 1994;16:181-200. 101. Kelley VR, Wüthrich RP. Cytokines in the pathogenesis of systemic lupus erythematosus. Semin Nephrol 1999;19:57-66. 102. Mosmann TR, Coffman RL. TH1 and TH2 cells: different patterns of lymphokine secretion lead to different functional properties. Annu Rev Immunol 1989;7:145-73. 103. Davidson WF, Calkins C, Hugin A, Giese T, Holmes KL. Cytokine secretion by C3H-lpr and -gld T cells. Hypersecretion of IFN-gamma and tumour necrosis factor-alpha by stimulated CD4+ T cells. J Immunol 1991;146: 4138-48. 104. Tang B, Matsuda T, Akira S, Nagata N, Ikehara S, Hirano T, et al. Age-associated increase in interleukin 6 in MRL/lpr mice. Int Immunol 1991;3:273-8. 105. Huang FP, Feng GJ, Lindop G, Stott DI, Liew FY. The role of interleukin 12 and nitric oxide in the development of spontaneous autoimmune disease in MRL:MP-lpr:lpr mice. J Exp Med 1996;183:1447-59. 106. Kiberd BA. Interleukin-6 receptor blockage ameliorates murine lupus nephritis. J Am Soc Nephrol 1993;4:58-61. 107. Jacob CO. In vivo treatment of (NZB x NZW)F1 lupuslike nephritis with monoclonal antibody to gamma interferon. J Exp Med 1987;166:798-803. 108. Ishida H, Muchamuel T, Sakaguchi S, Andrade S, Menon S, Howard M. Continuous administration of anti-interleukin 10 antibodies delays onset of autoimmunity in NZB/W F1 mice. J Exp Med 1994;179:305-10. 109. Ozmen L, Roman D, Fountoulakis M, Schmid G, Ryffel B, Garotta G. Experimental therapy of systemic lupus erythematosus: the treatment of NZB/W mice with mouse soluble interferon-gamma receptor inhibits the onset of glomerulonephritis. Eur J Immunol 1995;25:6-12. 110. Finck BK, Chan B, Wofsy D. Interleukin 6 promotes murine lupus in NZB/NZW F1 mice. J Clin Invest 1994;94: 585-91. 111. Nakajima A, Hirose S, Yagita H, Okumura K. Roles of IL4 and IL-12 in the development of lupus in NZB/W F1 mice. J Immunol 1997;158:1466-72. 112. de Wit D, van Mechelen M, Zanini C, Doutrelepont JM, Velu T, Gerard C, et al. Preferential activation of Th2 cells in chronic graft-versus-host reaction. J Immunol 1993;150: 361-6. 113. Umland SP, Razac S, Nahrebne DK, Seymour BW. Effects of in vivo administration of interferon (IFN)-γ anti-IFN-γ, or anti-interleukin-4 monoclonal antibodies in chronic autoimmune graft-versus-host disease. Clin Immunol Immunopathol 1992;63:66-73. 114. Couser WG. Mechanisms of glomerular injury in immunecomplex disease. Kidney Int 1985;28:569-83. 115. Andres G, Brentjens JR, Caldwell PRB, Camussi G, Matsuo S. Formation of immune deposits and disease. Lab Invest 1986;55:510-20. 116. Bruijn JA, Hoedemaeker PJ, Fleuren GJ. Pathogenesis of anti-basement membrane glomerulopathy and immunecomplex glomerulonephritis: dichotomy dissolved. Lab Invest 1989;61:480-8. 117. Suzuki N, Harada T, Mizushima Y, Sakane T. Possible pathogenic role of cationic anti-DNA autoantibodies in the development of nephritis in patients with systemic lupus erythematosus. J Immunol 1993;151:1128-36.

J Lab Clin Med April 2001

118. Kootstra CJ, Arkema A, Hogendoorn PCW, Daha MR, Weening JJ, Parwaresch, et al. A novel rat mesangial matrix protein, MMP-50/100, involved in mesangial glomerulopathies. Lab Invest 1995;73:72-9. 119. Sedor JR, Carey SW, Emancipator SN. Immune complexes bind to cultured rat glomerular mesangial cells to stimulate superoxide release: evidence for an Fc receptor. J Immunol 1987;138:3751-7. 120. Germuth FG, Rodriguez E, Lorelle CA, Trump EI, Milano LL, Wise O. Passive immune complex glomerulonephritis in mice: models for various lesions found in human disease. II. Low avidity complexes a n d diffuse proliferative glomerulonephritis with subepithelial deposits. Lab Invest 1979;41: 366-371. 121. Steward MW. Chronic immune complex disease in mice: the role of antibody affinity. Clin Exp Immunol 1979;38: 414-23. 122. Eliat D. Cross-reactions of anti-DNA antibodies and the central dogma of lupus nephritis. Immunol Today 1985;6: 123-7. 123. Rumore PM, Steinman CR. Endogenous circulating DNA in systemic lupus erythematosus: occurrence as multimeric complexes bound to histones. J Clin Invest 1990;86: 69-74. 124. Jacob L, Viard JD, Allenet B, Anin MF, Slama FB, Vandekerckhove J, et al. A monoclonal anti-double-stranded DNA autoantibody binds to a 94-kDa cell-surface protein on various cell types via nucleosomes or a DNA-histone complex. Proc Natl Acad Sci U S A 1989;86:4669-73. 125. Chan TM, Frampton G, Staines NA, Hobby P, Perry GJ, Cameron JS. Different mechanisms by which anti-DNA Moabs bind to human endothelial cells and glomerular mesangial cells. Clin Exp Immunol 1992;88:68-74. 126. Kramers K, Hylkema M, Termaat RM, Brinkman K, Smeenk R, Berden J. Histones in lupus nephritis. Exp Nephrol 1993;1:224-8. 127. Tax WJ, Kramers C, van Bruggen MC, Berden JHM. Apoptosis, nucleosomes, and nephritis in systemic lupus erythematosus. Kidney Int 1995;48:666-73. 128. Schmiedeke TMJ, Stoeckl FW, Weber R, Sugisaki Y, Batsford SR, Vogt A. Histones have high affinity for the glomerular basement membrane. Relevance for immune complex formation in lupus nephritis. J Exp Med 1989; 169:1879-94. 129. Di Valerio R, Bernstein KA, Varghese E, Lefkowith JB. Murine lupus glomerulotropic monoclonal antibodies exhibit differing specificities but bind via a common mechanism. J Immunol 1995;155:2258-68. 130. Zack DJ, Yamamoto K, Wong AL, Stempniak M, French C, Weisbart RH. DNA mimics a self-protein that may be a target for some anti-DNA antibodies in systemic lupus erythematosus. J Immunol 1995;154:1987-94. 131. Bruijn JA, van Leer EHG, Baelde JJ, Corver WE, Hogendoorn PCW, Fleuren GJ. Characterization and in vivo transfer of nephritogenic autoantibodies directed against dipeptidyl peptidase IV and laminin in experimental lupus nephritis. Lab Invest 1990;63:350-9. 132. Kootstra CJ, Veninga A, Baelde JJ, van Eendenburg J, de Heer E, Bruijn JA. Characterization of reactivity of monoclonal antibodies with renal antigens in experimental lupus nephritis. J Lab Clin Immunol 1996;48:201-18. 133. Madaio MP, Carlson J, Cataldo J, Ucci A, Migliorini P, Pankewycz O. Murine monoclonal anti-DNA antibodies bind directly to glomerular antigens and form immune deposits. J Immunol 1987;138:2883-9.

J Lab Clin Med Volume 137, Number 4

134. Casiano CA, Tan EM. Recent developments in the understanding of antinuclear autoantibodies. Int Arch Allergy Immunol 1996;111:308-13. 135. Alarcon-Segovia D, Ruiz-Arguelles A, Llorente L. Broken dogma: penetration of autoantibodies into living cells. Immunol Today 1996;17:163-4. 136. Yanase K, Smith RM, Cizman B, Foster MH, Peachey LD, Jarett L, et al. A subgroup of murine monoclonal antideoxyribonucleic acid antibodies traverse the cytoplasm and enter the nucleus in a time- and temperature-dependent manner. Lab Invest 1994;71:52-60. 137. Madaio MP. The role of autoantibodies in the pathogenesis of lupus nephritis. Semin Nephrol 1999;19:48-56. 138. Salant DJ, Madaio MP, Adler S, Stilmant MM, Couser WG. Altered glomerular permeability induced by F(ab’)2 and Fab’ antibodies to renal tubular epithelial antigen. Kidney Int 1981;21:36-43. 139. Assmann KJ, van Son JP, Dijkman HB, Koene RA. A nephritogenic rat monoclonal antibody to mouse aminopeptidase A. Induction of massive albuminuria after a single intravenous injection. J Exp Med 1992;175:623-35. 140. Aten J, Bosman CB, de Heer E, Hoedemaeker PJ, Weening JJ. Cyclosporin A induces long-term unresponsiveness in mercuric chloride-incuced autoimmune golmerulonephritis. Clin Exp Immunol 1988;73:307-11. 141. Van Velthuysen M-LF, Veninga A, Bruijn JA, de Heer E, Fleuren GJ. Susceptibility for infection-related glomerulopathy depends on non-MHC genes. Kidney Int 1993;43: 623-9. 142. Bergijk EC, Baelde HJ, de Heer E, Bruijn JA. Prevention of glomerulosclerosis by early cyclosporine treatment of experimental lupus nephritis. Kidney Int 1994;46:1663-73. 143. Shlomchik MJ, Madaio MP, Ni D, Trounstein M, Huszar D. The role of B cells in lpr/lpr-induced autoimmunity. J Exp Med 1994;180:1295-306. 144. Salant DJ, Adler S, Darby C, Capparell NJ, Groggel GC, Feintzeig ID, et al. Influence of antigen distribution on the mediation of immunological glomerular injury. Kidney Int 1985;27:938-50. 145. Sacks SH, Zhou WD, Sheerin NS. Complement synthesis in the injured kidney: does it have a role in immune complex glomerulonephritis? J Am Soc Nephrol 1996; 7:2314-9. 146. Timmerman JJ, Van Gijlswijk-Janssen DJ, van der Kooij SW, Van Es LA, Daha MR. Antigen-antibody complexes enhance the production of complement component C3 by human mesangial cells. J Am Soc Nephrol 1997;8: 1257-65. 147. Van den Dobbelsteen MEA, Verhasselt V, Kaashoek JGJ, Timmerman JJ, Schroeijers WE, Verwieij CL, et al. Regulation of C3 and factor H synthesis of human glomerular mesangial cells by IL-1 and interferon-gamma. Clin Exp Immunol 1994;95:173-80. 148. Schonermark M, Deppisch R, Riedasch G, Rother K, Haensch GM. Induction of mediator release from human glomerular mesangial cells by terminal complement components C5b-9. Int Arch Allergy Appl Immunol 1991;96: 331-7. 149. Lovett DH, Haensch G-M, Goppelt M, Resch K, Gemsa D. Activation of glomerular mesangial cells by the terminal membrane attack complex of complement. J Immunol 1987;138:2473-80. 150. Floege J, Alpers CE, Sage EH, Pritzl P, Gorden K, Johnson RJ, et al. Markers of complement-dependent and complement-independent glomerular visceral epithelial cell

Peutz-Kootstra et al

151.

152.

153.

154.

155.

156.

157.

158.

159.

160.

161.

162.

163. 164.

165.

166. 167.

168.

257

injury in vivo: expression of antiadhesive proteins and cytoskeletal changes. Lab Invest 1992;67:486-97. Wagner C, Braunger M, Beer M, Rother K, Hänsch GM. Induction of matrix protein synthesis in human glomerular mesangial cells by the terminal complement complex. Exp Nephrol 1994;2:51-6. Torbohm I, Schonermark M, Wingen AM, Berger B, Rother K, Haensch GM. C5b-8 and C5b-9 modulate the collagen release of human glomerular epithelial cells. Kidney Int 1990;37:1098-104. Passwell J, Schreiner GF, Nonaka M, Beuscher HU, Colten HR. Local extrahepatic expression of complement genes C3, factor B, C2, and C4 is increased in murine lupus nephritis. J Clin Invest 1988;82:1676-84. Lianos EA, Zanglis A. Effects of complement activation on platelet-activating factor and eicosanoid synthesis in rat mesangial cells. J Lab Clin Med 1992;120:459-64. Nassar GM, Badr KF. Role of leukotrienes and lipoxygenases in glomerular injury. Miner Electrolyte Metab 1995;21:262-70. Floege J, Johnson RJ, Alpers CE, Fatemi-Nainie S, Richardson CA, Gordon K, et al. Visceral glomerular epithelial cells can proliferate in vivo and synthesize platelet-derived growth factor B-chain. Am J Pathol 1993; 142:637-50. Brown Z, Strieter RM, Kunkel SL, Neild GN, Westwick J. Cyclosporin (CS) enhances IL-8 and MCP-1 gene expression and peptide formation by human mesangial cells (MC) [abstract]. J Am Soc Nephrol 1992;3:577. Hora K, Satriano JA, Santiago A, Mori T, Stanley ER, Shan Z, et al. Receptors for IgG complexes activate synthesis of monocyte chemoattractant peptide 1 and colony-stimulating factor 1. Proc Natl Acad Sci U S A 1992;89:1745-9. Bloom RD, Florquin S, Singer GG, Brennan DC, Kelley VR. Colony stimulating factor-1 in the induction of lupus nephritis. Kidney Int 1993;43:1000-9. Rovin BH, Yoshiumura T, Tan L. Cytokine-induced production of monocyte chemoattractant protein-1 by cultured human mesangial cells. J Immunol 1992;148:2148-53. Prodjosudjadi W, Gerritsma JSJ, Klar-Mohamed N, Gerritsen AF, Bruijn JA, Daha MR, et al. Production and cytokine-mediated regulation of monocyte chemoattractant protein-1 by human proximal tubular epithelial cells. Kidney Int 1995;48:1477-86. Wolf G, Aberle S, Thaiss F, Nelson PJ, Krensky AM, Neilson EG, et al. TNF alpha induces expression of the chemoattractant cytokine RANTES in cultured mouse mesangial cells. Kidney Int 1993;44:795-804. Benigni A, Remuzzi G. Inflammation and glomerular injury. Springer Semin Immunopathol 1994;16:39-51. Noris M, Remuzzi G. New insights into circulating cellendothelium interactions and their significance for glomerular pathophysiology. Am J Kidney Dis 1995;26: 541-8. Abbate M, Remuzzi G. Glomerulonephritis: from new knowledge on the mechanisms of tissue damage to novel therapies. Curr Opin Nephrol Hypertens 1995;4:374-82. Brady HR. Leukocyte adhesion molecules and kidney diseases. Kidney Int 1994;45:1285-300. Takano T, Brady HR. The endothelium in glomerular inflammation. Curr Opin Nephrol Hypertens 1995;4: 277-86. Wuthrich RP. Vascular cell adhesion molecule-1 (VCAM1) expression in murine lupus nephritis. Kidney Int 1992;42:903-14.

J Lab Clin Med April 2001

258 Peutz-Kootstra et al

169. Wuthrich RP, Jevnikar AM, Takei F, Glimcher LH, Kelley VE. Intercellular adhesion molecule-1 (ICAM-1) expression is upregulated in autoimmune murine lupus nephritis. Am J Pathol 1990;136:441-50. 170. Moore KJ, Wada T, Barbee SD, Kelley VR. Gene transfer of RANTES elicits autoimmune renal injury in MRLFaslpr mice. Kidney Int 1998;53:1631-41. 171. Wüthrich RP, Fan XH, Ritthaler T, Sibalic V, Yu DJ, Loffing J, et al. Enhanced osteopontin expression and macrophage infiltration in MRL-Faslpr mice with lupus nephritis. Autoimmunity 1998;28:139-50. 172. Kootstra CJ, van der Giezen DM, van Krieken JHJM, de Heer E, Bruijn JA. Effective treatment of experimental lupus nephritis by combined administration of anti-CD11a and anti-CD54 antibodies. Clin Exp Immunol 1997;108: 324-32. 173. Rovin BH, Schreiner GF. Cell-mediated immunity in glomerular disease. Annu Rev Med 1991;42:25-33. 174. Van Goor H, Fidler V, Weening JJ, Grond J. Determinants of focal and segmental glomerulosclerosis in the rat after renal ablation: evidence for involvement of macrophages and lipids. Lab Invest 1991;64:754-65. 175. Kawasaki K, Yaoita E, Yamamoto T, Kihara I. Depletion of CD8 positive cells in nephrotoxic serum nephritis of WKY rats. Kidney Int 1992;41:1517-26. 176. Kawasaki K, Yaoita E, Yamamoto T, Tamatani T, Miyasaka M, Kihara I. Antibodies against intercellular adhesion molecule-1 and lymphocyte function-associated antigen-1 prevent glomerular injury in rat experimental crescentic glomerulonephritis. J Immunol 1993;150:1074-83. 177. Nishikawa K, Guo Y-J, Miyasaka M, Tamatani T, Collins AB, Sy MS, et al. Antibodies to intercellular adhesion molecule 1/lymphocyte function-associated antigen 1 prevent crescent formation in rat autoimmune glomerulonephritis. J Exp Med 1993;177:667-77. 178. Kimura M, Nagase M, Hishida A, Honda N. Intramesangial passage of mononuclear phagocytes in murine lupus glomerulonephritis. Am J Pathol 1987;127:149-56. 179. Kootstra CJ, Sutmuller M, Tijsma O, Baelde JJ, de Heer E, Bruijn JA. Glomerular leukocyte infiltration correlates with development of glomerulosclerosis in experimental lupus nephritis. J Pathol 1998;184:219-25. 180. Floege J, Johnson RJ. Cytokines in renal inflammation. Curr Opin Nephrol Hypertens 1993;2:449-57. 181. Sedor JR, Nakazato Y, Konieczkowski M. Interleukin-1 and the mesangial cell. Kidney Int 1992;41:595-9. 182. Atkins RC. Interleukin-1 in crescentic glomerulonephritis. Kidney Int 1995;48:576-86. 183. Roberts AB, McCune BK, Sporn MB. TGF-β regulation of extracellular matrix. Kidney Int 1992;41:557-9. 184. Border WA, Ruoslahti E. Transforming growth factor-β in disease: the dark side of tissue repair. J Clin Invest 1992; 90:1-7. 185. Bruijn JA, Roos A, de Geus B, de Heer E. Transforming growth factor-β and the glomerular extracellular matrix in renal pathology. J Lab Clin Med 1994;123:34-47. 186. Floege J, Kriz W, Schulze M, Susani M, Kerjaschki D, Mooney A, et al. Basic fibroblast growth factor augments podocyte injury and induces glomerulosclerosis in rats with experimental membranous nephropathy. J Clin Invest 1995; 96:2809-19. 187. Abboud HE. Platelet-derived growth factor and mesangial cells. Kidney Int 1992;41:581-3. 188. Johnson R, Iida H, Yoshimura A, Floege J, Bowen-Pope DF. Platelet-derived growth factor: a potentially impor-

189.

190.

191.

192. 193.

194.

195.

196.

197.

198.

199.

200.

201.

202.

203.

tant cytokine in glomerular disease. Kidney Int 1992; 41:590-4. Bruggeman LA, Pellicoro JA, Horigan EA, Klotman PE. Thromboxane and prostacyclin differentially regulate murine extracellular matrix gene expression. Kidney Int 1993;43:1219-25. Studer RK, Craven PA, DeRubertis FR. Thromboxane stimulation of mesangial cell fibronectin synthesis is signalled by protein kinase C and modulated by cGMP. Kidney Int 1994;46:1074-82. Floege J, Eng E, Lindner V, Alpers CE, Young BA, Reidy MA, et al. Rat glomerular mesangial cells synthesize basic fibroblast growth factor. Release, upregulated synthesis, and mitogenicity in mesangial proliferative glomerulonephritis. J Clin Invest 1992;90:2362-9. Coleman DL, Ruef C. Interleukin-6: an autocrine regulator of mesangial cell growth. Kidney Int 1992;41:604-6. Brennan DC, Yui MA, Wuthrich RP, Kelley VE. Tumor necrosis factor and IL-1 in New Zealand Black/White mice: enhanced gene expression and acceleration of renal injury. J Immunol 1989;143:3470-5. Boswell JM, Yui MA, Burt DW, Kelley VE. Increased tumor necrosis factor and IL-1β gene expression in the kidneys of mice with lupus nephritis. J Immunol 1988;141: 3050-4. Chandrasekar B, Troyer DA, Venkatraman JT, Fernandes G. Dietary omega-3 lipids delay the onset and progression of autoimmune lupus nephritis by inhibiting transforming growth factor beta mRNA and protein expression. J Autoimmun 1995;8:381-93. Moll S, Menoud PA, Fulpius T, Pastore Y, Takahashi S, Fossati L, et al. Induction of plasminogen activator inhibitor type 1 in murine lupus-like glomerulonephritis. Kidney Int 1995;48:1459-68. Nakamura T, Ehihara I, Nagaoka I, Tomino Y, Koide H. Renal platelet-derived growth factor gene expression in NZB/W F1 mice with lupus and ddY mice with IgA nephropathy. Clin Immunol Immunopathol 1992;173:171-81. Kootstra CJ, Tijsma O, Baelde JJ, Schoenberger S, de Heer E, Bruijn JA. TGF-β independent development of glomerulosclerosis in murine chronic graft versus host disease (GvHD)? [abstract]. J Am Soc Nephrol 1996. Salvati P, Lamberti E, Ferrario R, Ferrario RG, Scampini G, Pugliese F, et al. Long-term thromboxane-synthase inhibition prolongs survival in murine lupus nephritis. Kidney Int 1995;47:1168-75. Nakamura T, Ebihara I, Tomino Y, Koide H. Effect of a specific endothelin A receptor antagonist on murine lupus nephritis. Kidney Int 1995;47:481-9. Weinberg JB, Granger DL, Pisetsky DS, Seldin MF, Misukonis MA, Mason SN, et al. The role of nitric oxide in the pathogenesis of spontaneous murine autoimmune disease: increased nitric oxide production and nitric oxide synthase expression in MRL-lpr/lpr mice, and reduction of spontaneous glomerulonephritis and arthritis by orally administered NG-monomethyl-L-arginine. J Exp Med 1994;179:651-60. Hortelano S, Diaz-Guerra MJM, Gonzalez-Garcia A, Leonardo E, Gamallo C, Bosca L, et al. Linomide administration to mice attenuates the induction of nitric oxide synthase elicited by lipopolysaccharide-activated macrophages and prevents nephritis in MRL/Mp-lpr/lpr mice. J Immunol 1997;158:1402-8. Fan PY, Ruiz P, Pisetsky DS, Spurney RF. The effects of short-term treatment with the prostaglandin E1 (PGE1)

J Lab Clin Med Volume 137, Number 4

204.

205.

206.

207.

208.

209. 210.

211.

212.

213.

214.

215.

216. 217.

218.

219.

220.

analog misoprostol on inflammatory mediator production in murine lupus nephritis. Clin Immunol Immunopathol 1995;75:125-30. Schalkwijk CG, De Vet E, Pfeilschifter J, Van den Bosch H. Interleukin-1 beta and transforming growth factor-β2 enhance cytosolic high-molecular-mass phospholipase A2 activity and induce prostaglandin E2 formation in rat mesangial cells. Eur J Biochem 1992;210:169-76. Levine JS, Pugh BJ, Hartwell D, Fitzpatrick JM, MarshakRothstein A, Beller DI. Interleukin-1 dysregulation is an intrinsic defect in macrophages from MRL autoimmuneprone mice. Eur J Immunol 1993;23:2951-8. Lebedeva TV, Singh AK. Increased responsiveness of B cells in the murine MRL/lpr model of lupus nephritis to interleukin-1 beta. J Am Soc Nephrol 1995;5:1530-4. Eddy AA. Experimental insights into the tubulointerstitial disease accompanying primary glomerular lesions J Am Soc Nephrol 1994;5:1273-87. D’Amico G, Ferrario F, Rastaldi MP. Tubulointerstitial damage in glomerular diseases: its role in the progression of renal damage. Am J Kidney Dis 1995;26:124-32. El Nahas AM. Pathways to renal fibrosis. Exp Nephrol 1995;3:71-5. Pichler R, Giachelli C, Young B, Alpers CE, Couser WG, Johnson RJ. The pathogenesis of tubulointerstitial disease associated with glomerulonephritis: the glomerular cytokine theory. Miner Electrolyte Metab 1995;21:317-27. Risdon RA, Sloper JC, de Wardener HE. Relationship between renal function and histological changes found in renal biopsy specimens from patients with persistent glomerular nephritis. Lancet 1968;17:363-6. Moyer CF, Strandberg JD, Reinisch CL. Systemic mononuclear-cell vasculitis in MRL/Mp-lpr/lpr mice. Am J Pathol 1987;127:229-42. Nose M, Nishimura M, Ito MR, Itoh J, Shibata T, Sugisaki T. Arteritis in a novel congenic strain of mice derived from MRL/lpr lupus mice—genetic dissociation from glomerulonephritis and limited autoantibody production. Am J Pathol 1996;149:1763-9. Staszak C, Harbeck RJ. Mononuclear-cell pulmonary vasculitis in NZB/W mice. I. Histopathologic evaluation of spontaneously occurring pulmonary infiltrates. Am J Pathol 1985;120:99-105. Berden JHM, Hang LM, McConahey PJ, Dixon FJ. Analysis of vascular lesions in murine SLE. J Immunol 1983;130: 1699-705. Juliano RL, Haskill S. Signal transduction from the extracellular matrix. J Cell Biol 1993;120:577-85. Lin CQ, Bissell MJ. Multi-faceted regulation of cell differentiation by extracellular matrix. FASEB J 1993;7: 737-43. Ekblom M, Klein G, Mugrauer G, Fecker L, Deutzmann R, Timpl R, et al. Transient and locally restricted expression of laminin A chain mRNA by developing epithelial cells during kidney organogenesis. Cell 1990;60:337-46. Abrahamson DR, St John PL. Loss of laminin epitopes during glomerular basement membrane assembly in developing mouse kidneys. J Histochem Cytochem 1992;40: 1943-53. McCarthy KJ, Bynum K, St John PL, Abrahamson DR, Couchman JR. Basement membrane proteoglycans in glomerular morphogenesis: chondroitin sulfate proteoglycan is temporally and spatially restricted during development. J Histochem Cytochem 1993;41:401-14.

Peutz-Kootstra et al

259

221. Miner JH, Sanes JR. Collagen IV α3, α4, and α5 chains in rodent basal laminae: sequence, distribution, association with laminins, and developmental switches. J Cell Biol 1994;127:879-91. 222. Baricos WH. Chronic renal disease: do metalloproteinase inhibitors have a demonstrable role in extracellular matrix accumulation? Curr Opin Nephrol Hypertens 1995;4: 365-8. 223. Bruijn JA, Kootstra CJ, Sutmuller M, van Vliet A, Bergijk EC, de Heer E. Matrix and adhesion molecules in kidney pathology: recent observations. J Lab Clin Med 1997;130: 357-64. 224. Bergijk EC, de Heer E, Hoedemaeker PJ, Bruijn JA. A reappraisal of immune-mediated glomerulosclerosis. Kidney Int 1996;49:605-11. 225. Van Bruggen MCJ, Kramers K, Hylkema MN, van den Born J, Bakker MA, Assmann KJ, et al. Decrease of heparan sulfate staining in the glomerular basement membrane in murine lupus nephritis. Am J Pathol 1995;146: 753-63. 226. Munaut C, Bergijk EC, Baelde JJ, Noël A, Foidart JM, Bruijn JA. A molecular biologic study of extracellular matrix components during the development of glomerulosclerosis in murine chronic graft-versus-host disease. Lab Invest 1992;67:580-7. 227. Kootstra CJ, Bergijk EC, Veninga A, de Heer E, Abrahamson DR, Bruijn JA. Qualitative alterations in laminin expression in experimental lupus nephritis. Am J Pathol 1995;147:476-88. 228. Bergijk EC, Baelde HJ, Kootstra CJ, de Heer E, Killen PD, Bruijn JA. Cloning of the mouse fibronectin V-region and variation of its splicing pattern in experimental immune complex glomerulonephritis. J Pathol 1996;178:462-8. 229. Peutz-Kootstra CJ, Hansen K, Heer de E, Abrass CK, Bruijn JA. Differential expression of laminin chains and anti-laminin autoantibodies in experimental lupus nephritis. J Pathol 2000;192:404-12. 230. Beck K, Hunter I, Engel J. Structure and function of laminin: anatomy of a multidomain glycoprotein. FASEB J 1990;4:148-60. 231. Engel J. Laminins and other strange proteins. Biochemistry 1992;31:10643-51. 232. van Leer EHG, Bruijn JA, Prins FA, Hoedemaeker PJ, de Heer E. Redistribution of glomerular dipeptidyl peptidase type IV in experimental lupus nephritis. Demonstration of decreased enzyme activity at the ultrastructural level. Lab Invest 1993;68:550-6. 233. Treurniet RA, Bergijk EC, Baelde JJ, de Heer E, Hoedemaeker PJ, Bruijn JA. Gender-related influences on the development of chronic graft-versus-host disease-induced experimental lupus nephritis. Clin Exp Immunol 1993;91: 442-8. 234. Aten J, Chand A, Claessen N, Weening JJ. The B cell repertoire in mercuric chloride-induced systemic autoimmunity in DZB rats [abstract]. J Am Soc Nephrol 1993;4:593. 235. Jevnikar AM, Singer GG, Brennan DC, Xu HW, Kelley VR. Dexamethasone prevents autoimmune nephritis and reduces renal expression of Ia but not costimulatory signals. Am J Pathol 1992;141:743-51. 236. Singer GG, Yokoyama H, Bloom RD, Jevnikar AM, Nabavi N, Kelley VR. Stimulated renal tubular epithelial cells induce anergy in CD4 + T cells. Kidney Int 1993;44: 1030-5. 237. Kelley VR, Diaz-Gallo C, Jevnikar AM, Singer GG. Renal

260 Peutz-Kootstra et al

238.

239.

240.

241.

242. 243.

244. 245.

246.

247.

248.

249.

250.

tubular epithelial and T cell interactions in autoimmune renal disease. Kidney Int 1993;43(Suppl 39):S108-15. Kelley VR, Singer GG. The antigen presentation function of renal tubular epithelial cells. Exp Nephrol 1993;1: 102-11. Brennan DC, Jevnikar AM, Takei F, Rubin-Kelley VE. Mesangial cell accessory functions: mediation by intracellular adhesion molecule-1. Kidney Int 1990;38:1039-46. Mendrick DL, Kelly DM, Rennke HG. Antigen processing and presentation by glomerular visceral epithelium in vitro. Kidney Int 1991;39:71-8. Kaliyaperumal A, Michaels MA, Datta SK. Antigenspecific therapy of murine lupus nephritis using nucleosomal peptides: tolerance spreading impairs pathogenic function of autoimmune T and B cells. J Immunol 1999; 162:5775-83. Tsokos GC. Lymphocyte abnormalities in human lupus. Clin Immunol Immunopathol 1992;63:7-9. Klinman DM. B-cell abnormalities characteristic of systemic lupus erythematosus. In: Wallace DJ, Hahn BH, editors. Dubois’ lupus erythematosus. Philadelphia: Lea and Febiger; 1993. p. 77-82. Harley JB, James JA. Autoepitopes in lupus. J Lab Clin Med 1995;126:509-16. Winfield JB, Fernsten P, Czyzyk J, Wang E, Marchalonis J. Antibodies to CD45 and other cell membrane antigens in systemic lupus erythematosus. Springer Semin Immunopathol 1994;16:201-10. Atta MS, Powell RJ, Hopkinson ND, Todd I. Human antifibronectin antibodies in systemic lupus erythematosus: occurrence and antigenic specificity. Clin Exp Immunol 1994;96:20-5. Shibata S, Sasaki T, Harpel P, Fillit H. Autoantibodies to vascular heparan sulfate proteoglycan in systemic lupus erythematosus react with endothelial cells and inhibit the formation of thrombin-antithrombin III complexes. Clin Immunol Immunopathol 1994;70:114-23. Garcia Lerma JG, Moneo I, Ortiz De Landazuri MO, Sequi Navarro J. Comparison of the anti-laminin antibody response in patients with systemic lupus erythematosus (SLE) and parasitic diseases (filariasis). Clin Immunol Immunopathol 1995;76:19-31. Markovic-Lipovski J, Muller CA, Risler T, Bohle A, Muller GA. Association of glomerular and interstitial mononuclear leukocytes with different forms of glomerulonephritis. Nephrol Dial Transplant 1990;5:10-7. Bruijn JA, Dinklo NJCM. Distinct patterns of expression of ICAM-1, VCAM-1, and ELAM-1 in renal disease. Lab Invest 1993;69:329-35.

J Lab Clin Med April 2001

251. Yokoyama H, Takabatake T, Takaeda M, Wada T, Naito T, Ikeda K, et al. Up-regulated MHC-class II expression and gamma-IFN and soluble IL-2R in lupus nephritis. Kidney Int 1992;42:755-63. 252. Yamamoto T, Noble NA, Cohen AH, et al. Expression of transforming growth factor-beta isoforms in human glomerular disease. Kidney Int 1996;49:461-9. 253. Yamamoto T, Watanabe T, Ikegaya N, Fujigaki Y, Matsui K, Masaoka H, et al. Expression of types I, II, III TGF-beta receptors in human glomerulonephritis. J Am Soc Nephrol 1998;9:2253-61. 254. Ito Y, Aten J, Bende RJ, Oemar BS, Rabelink TJ, Weening JJ, et al. Expression of connective tissue growth factor in human renal fibrosis. Kidney Int 1998;53:853-61. 255. Furusu A, Miyazaki M, Koji T, Abe K, Ozono Y, Harada T, et al. Involvement of IL-4 in human glomerulonephritis: an in situ hybridization study of IL-4 mRNA and IL-4 receptor mRNA. J Am Soc Nephrol 1997;8:730-41. 256. Furusu A, Miyazaki M, Abe K, Tsukasaki S, Shioshita K, Sasaki O, et al. Expression of endothelial and inducible nitric oxide synthase in human glomerulonephritis. Kidney Int 1998;53:1760-8. 257. Striker L, Killen PD, Chi E, Striker GE. The composition of glomerulosclerosis I. Studies in focal sclerosis, crescentic glomerulonephritis, and membranoproliferative glomerulonephritis. Lab Invest 1984;51:181-92. 258. Kuhara T, Kagami S, Kuroda Y. Expression of beta1-integrins on activated mesangial cells in human glomerulonephritis. J Am Soc Nephrol 1997;8:1679-87. 259. Van den Born J, Van den Heuvel LPWJ, Bakker MAH, Veerkamp JH, Assmann KJ, Weening JJ, et al. Distribution of GBM heparan sulfate proteoglycan core protein and side chains in human glomerular diseases. Kidney Int 1993; 43:454-63. 260. Striker LJ. Modern renal biopsy interpretation: can we predict glomerulosclerosis? Semin Nephrol 1993;13:508-15. 261. Donadio JV Jr, Glassock RJ. Immunosuppressive drug therapy in lupus nephritis. Am J Kidney Dis 1993;21: 239-50. 262. Nakamura T, Ebihara I, Tomino Y, Koide H. Effect of a specific endothelin A receptor antagonist on murine lupus nephritis. Kidney Int 1995;47:481-9. 263. Kitamura M, Taylor S, Unwin R, Burton S, Shimizu F, Fine LG. Gene transfer into the rat renal glomerulus via a mesangial cell vector: site-specific delivery, in situ amplification, and sustained expression of an exogenous gene in vivo. J Clin Invest 1994;94:497-505.