Skip to main content

Renal involvement in autoimmune connective tissue diseases

Abstract

Connective tissue diseases (CTDs) are a heterogeneous group of disorders that share certain clinical presentations and a disturbed immunoregulation, leading to autoantibody production. Subclinical or overt renal manifestations are frequently observed and complicate the clinical course of CTDs. Alterations of kidney function in Sjögren syndrome, systemic scleroderma (SSc), auto-immune myopathies (dermatomyositis and polymyositis), systemic lupus erythematosus (SLE), antiphospholipid syndrome nephropathy (APSN) as well as rheumatoid arthritis (RA) are frequently present and physicians should be aware of that.

In SLE, renal prognosis significantly improved based on specific classification and treatment strategies adjusted to kidney biopsy findings. Patients with scleroderma renal crisis (SRC), which is usually characterized by severe hypertension, progressive decline of renal function and thrombotic microangiopathy, show a significant benefit of early angiotensin-converting-enzyme (ACE) inhibitor use in particular and strict blood pressure control in general. Treatment of the underlying autoimmune disorder or discontinuation of specific therapeutic agents improves kidney function in most patients with Sjögren syndrome, auto-immune myopathies, APSN and RA.

In this review we focus on impairment of renal function in relation to underlying disease or adverse drug effects and implications on treatment decisions.

Peer Review reports

Background

Impairment of renal function is present to some extent in many connective tissue diseases (CTDs) with variable occurrence in Sjögren syndrome [1, 2], roughly 5% in systemic scleroderma (SSc) [3], rarely in inflammatory auto-immune myopathies, a prevalence of approximately 50% in systemic lupus erythematosus (SLE) [4], and rare occurrence in antiphospholipid syndrome [5] and rheumatoid arthritis (RA). Apart from that, kidney involvement can be of significant prognostic value and often entails specific therapeutic implications.

Lymphocytic infiltration, leading to acute or chronic tubulointerstitial nephritis, is the predominant renal pathology in Sjögren syndrome [2, 6, 7]. Scleroderma renal crisis (SRC) is a severe, potentially life-threatening complication in scleroderma and is, in most cases, accompanied by malignant hypertension, overexpression of pro-inflammatory cytokines and rapid decline of renal function [810]. In rare cases patients present with normotensive SRC, which is associated with a poorer prognosis and a prompter need for dialysis [1113]. Early commencement of angiotensin-converting-enzyme (ACE)-inhibitors and other antihypertensive drugs is mandatory in the management of SRC. Rhabdomyolysis with acute tubular necrosis or glomerular disorders, including minimal change disease, membranous nephropathy, IgA nephropathy or diffuse proliferative glomerulonephritis, has been reported in patients with auto-immune myopathies [14, 15].

Lupus nephritis is one of the most severe organ manifestations of the disease and, depending on biopsy findings, needs aggressive immunosuppressive therapy. The histopathologic classification of lupus nephritis guides therapeutic interventions with the aim to reduce proteinuria and preserve kidney function. Renal manifestation in primary and secondary antiphospholipid syndrome (APS) is a well-described complication, frequently leading to arterial hypertension and occasionally impairment of renal function [5, 16]. Patients with RA are at an increased risk of developing secondary amyloidosis due to long-lasting chronic inflammation as well as mesangial glomerulonephritis and membranous nephropathy related to specific drugs [17]. Table 1 summarizes specific kidney biopsy findings in the context of CTDs.

Table 1 Overview of kidney biopsy findings in patients with connective tissue diseases

Review

Sjögren syndrome

Introduction

Primary Sjögren syndrome (PSS) is an autoimmune disorder of hitherto unknown origin which is characterized by polyclonal B-cell activation as well as lymphocytic infiltration of the exocrine glands, resulting in keratoconjunctivitis sicca (dry eyes disease) and/or xerostomia (dry mouth disease) [18]. In addition, extraglandular manifestations of PSS can affect organ systems, such as the lungs, blood vessels, skin, the gastrointestinal tract, central and peripheral nervous system, muscular skeletal apparatus and the kidney [19, 20]. Affected patients are at increased risk of developing non-Hodgkin’s lymphoma, in particular lymphomas of B-cell origin [21]. Secondary Sjögren syndrome is associated with other rheumatic disorders, such as RA, SLE, SSc and others. Renal disease with PSS is reported to occur in 4.2% [1] to 67% [2] of patients. The variation is considered to be associated with the different diagnostic criteria used, different study designs and small cohorts examined as well as selection bias.

Histopathology/kidney involvement

Acute or chronic tubulointerstitial nephritis (TIN) with defects in tubular function is the predominant lesion in biopsy-proven renal involvement [2, 6]. Distal (type I) renal tubular acidosis (RTA) is the most common clinical finding, leading to mild symptoms but also to potentially life-threatening complications, such as hypokalemic paralysis [22]. Albeit considered to be rare, proximal (type II) RTA has been reported in some cases [6, 23]. Moreover, glomerular disease, such as cryoglobulinemic membrano-proliferative glomerulonephritis, focal segmental glomerulosclerosis (FSGS), mesangial proliferative glomerulonephritis, membranous nephropathy and minimal change disease have been reported [2, 6, 7, 24]. A single case of long-lasting TIN in a patient with PSS led to a secondary (AA) amyloidosis with, consequently, renal failure and nephrotic syndrome [18]. Interestingly, in one study SSA/Ro, SSB/La and rheumatoid factor, as well as hypergammaglobulinemia, were detected in all subjects with biopsy-proven renal involvement [6]. In another cohort, all patients with distal RTA had positive anti-nuclear antibodies and either SSA or SSB antibodies were detected in 85.7% of them [25].

Therapy

Treatment with glucocorticoids should be initiated as first line therapy in patients with PSS and renal involvement since a good response to early treatment has been reported [6, 26]. In addition, long-term bicarbonate and/or electrolyte supplementation should be commenced in a majority of patients to prevent life-threatening complications [26]. Besides corticosteroids, alternative immunosuppressive therapies (hydroxychloroquine, rituximab, cyclophosphamide) should be prescribed based on kidney biopsy findings as well as comorbidities. It was shown that renal function maintained or improved during a median follow-up period of 38 months after treatment with immunosuppressive drugs [6].

Conclusion

Corticosteroids are a mainstay in the treatment of TIN. Further histologic findings require specifically-tailored immunosuppression and most importantly, supplementation of bicarbonate and/or electrolytes, when indicated, should be commenced.

Scleroderma renal crisis

Introduction

SSc is a CTD characterized by deposition and overproduction of extracellular matrix proteins and collagen, resulting in tissue fibrosis and, subsequently, tissue dysfunction. Affected organs and tissues include the skin, gastrointestinal tract, heart, lungs and kidneys. Involvement of the vascular system generally results in the development of Raynaud’s phenomenon early in the disease course. Consecutively, severe clinical manifestations of vascular dysfunction can be observed in some patients leading to pulmonary fibrosis and pulmonary artery hypertension, esophageal motility dysfunction, watermelon stomach, cardiac involvement, as well as scleroderma renal crisis (SRC) [2729]. Epithelial to mesenchymal transition (EMT), a condition that conveys a phenotypic conversion from differentiated epithelial cells to matrix-producing fibroblasts and myofibroblasts, is recognized as a crucial part of the development of tissue fibrogenesis [30, 31]. Several growth factors, such as transforming growth factor ß (TGFß) [8], connective tissue growth factor (CTGF) [9], as well as other mediators, such as endothelin-1 [10], are involved in tissue remodelling. SRC occurs in roughly 5% of patients with SSc [3]. Several risk factors with a predictive value were established: duration of SSc onset of less than four years, higher incidence of progressive skin thickening prior to renal involvement, new development of anemia and cardiac involvement (pericardial effusion or congestive heart failure) [32]. Detection of anti-RNA polymerase III antibodies displays a strong risk marker for the presence of SRC, whereas the presence of anti-topoisomerase and anti-centromere antibodies in scleroderma indicates a favorable disease course [33]. In addition, a case control study revealed a significant positive association between long-lasting high-dose corticosteroid treatment (≥15 g/d) and the onset of SRC [34]. On average, in 10% of patients, SRC occurs in the absence of hypertension. Normotensive renal crisis was more frequently present in patients treated with high doses of corticosteroids and in patients with redundant microangiopathic hemolytic anemia and thrombocytopenia in consequence to the underlying disease [11]. In addition, normotensive renal failure in SSc was associated with a higher mortality rate and an earlier need for dialysis treatment [1113]. Blood pressure levels greater than 150/90 mmHg were observed in almost 90% of patients experiencing SRC. Hypertensive SRC is accompanied by clinical signs of malignant hypertension with left ventricular failure, hypertensive encephalopathy and arrhythmia [12].

Histopathology/kidney involvement

Diagnosis of SRC is confirmed by renal biopsy, which shows a thrombotic microangiopathic process, particularly affecting small vessels. Vascular changes are accompanied by thrombosis, accumulation of myxoid material and later in the disease course, development of onion-skin lesions and/or fibrointimal sclerosis [13]. Furthermore, one has to keep in mind that ANCA-associated vasculitis is a rare complication of SSc and in general presents with antibodies directed against myeloperoxidase and p-ANCA [35].

Therapy

Early use of ACE inhibitors, on the basis of most experience in particular captopril, is undoubtedly a cornerstone in the management of hypertensive SRC. Immediate use with a progressive increase of ACE inhibitor dosage, even in the presence of deteriorating kidney function, is considered to prevent or even reverse renal failure [36, 37]. Additional antihypertensive therapy (calcium channel blockers, alpha/beta-adrenoreceptor antagonists and/or minoxidil) is mandatory when blood pressure is insufficiently controlled [28, 36]. Recent findings suggest that dialysis was required in more than 50% of patients either in case of volume overload together with renal deterioration or to control blood pressure due to therapy-resistant hypertension [3, 12]. Discontinuation of dialysis treatment could be accomplished in 16 to 55% patients with SRC [3, 37]. After a stable disease course with continuous dialysis, renal transplantation should be considered when contraindications are ruled out. In a cohort of 260 patients with SSc who underwent kidney transplantation, the overall five-year graft survival rate was 56.7%. Among those, the recurrence of disease after transplantation was 6.7% in a report of the United Network of Organ Sharing (UNOS) [38]. Based on the finding that cyclosporine A (CSA) may be responsible for acute renal failure in patients with SSc [39], calcineurin inhibitors are not generally recommended as immunosuppressants after kidney transplantation.

Conclusion

Renal involvement in SSc is often accompanied by progressive renal failure and rapid initiation of therapeutic interventions is mandatory. Blood pressure control, in particular with ACE-inhibitors and additional antihypertensive medication, is essential. If blood pressure is not adjustable or the patient shows signs of fluid overload, dialysis should be considered early in the disease course. Discontinuation of dialysis was reported in some patients. In patients with chronic hemodialysis and renal transplantation, a calcineurin-inhibitor free immunosuppressive regimen might be chosen.

Dermatomyositis and polymyositis

Introduction

Auto-immune myopathies, namely dermatomyositis (DM) and polymyositis (PM), share common clinical features, such as proximal muscle weakness, muscle inflammation, presence of autoantibodies, elevated muscle enzymes, electromyographic alterations and extra muscular manifestations. Despite clinical similarities, both differ regarding muscle biopsy findings and DM is associated with cutaneous involvement. The presence of a heliotrope rash, which is characterized by a violaceous skin discoloration around the eyes, and Gottron’s sign (erythematous papules with involvement of joints) are pathognomonic for DM [40, 41]. Both entities are associated with concurrent incidence of neoplasms. In large cohorts, malignancies were detected in 9.4 to 32% of patients in DM and in 4.4 to 17% in PM patients [4245] with a predominance of adenocarcinomas [45].

Histopathology/kidney involvement

Two types of renal involvement have been described in patients with PM/DM. First, rhabdomyolysis with release of myoglobin can lead to acute tubular necrosis with deterioration of renal function [14, 15]. Second, several reports revealed the occurrence of chronic glomerulonephritis in patients with PM/DM [14, 4649]. In PM, mesangial proliferative glomerulonephritis represents the leading glomerular lesion [46, 50, 51]. Moreover, other biopsy specimens showed lipoid nephrosis with FSGS [52], membranous nephropathy [53] and crescentic glomerulonephritis with FSGS [54]. In contrast, the predominant finding in DM with renal involvement is membranous nephropathy [5557]. Nevertheless, both mesangial proliferative glomerulonephritis [58] and diffuse proliferative glomerulonephritis [49] have been reported in single case reports.

Therapy

High-dose oral corticosteroids are the cornerstone of DM/PM therapy. Moreover, the addition of immunosuppressive drugs, such as azathioprine (AZA) or cyclophosphamide (CYC), as well as anti-malaria medication in DM and methotrexate, CYC, intravenous immunoglobulins and CSA in PM has been reported to improve the renal outcome in DM/PM [14, 49, 5558]. In contrast to these reports, one patient progressed to end-stage renal disease despite immunosuppressive treatment [59]. Follow-up of the patients with DM revealed a high mortality rate due to cancer or multi-organ failure, while mortality in PM was high due to acute rhabdomyolysis followed by severe hyperkalemia and metabolic acidosis in a case report [14].

Conclusion

Management of patients with auto-immune myopathies and renal involvement require special caution, because disease-related mortality due to rhabdomyolysis and hyperkalemia is greatly feared. Special therapeutic intervention with immunosuppression should be tailored to the underlying histology. In most cases, corticosteroids might be effective as one therapeutic component.

Systemic lupus erythematosus

Introduction

SLE depicts a remarkable complex autoimmune disease with considerable heterogeneity in clinical manifestations and disease course. Classification of SLE was last edited by the American College of Rheumatology (ACR) in 1997 [60] (Table 2). Earlier diagnosis, more intensive treatment regimens and diverse alternative strategies and possibilities to treat co-morbidities have contributed to improvement of prognosis [61]. Negative predictive factors with respect to survival include male gender, positive lupus anticoagulant, glomerulonephritis and “severe” onset of SLE [62]. The incidence is much higher in young woman and the prevalence is two- to four-fold greater in non-Caucasian populations [63]. Genetic, environmental and hormonal factors have been identified as possible risk factors for developing SLE [64, 65].

Table 2 Revised criteria of the American College of Rheumatology

Autoantibodies are directed against various nuclear antigens, in particular against chromatin components, such as nucleosomes, histones, anti-nuclear antibodies (ANA), double-stranded DNA antibodies (dsDNA) and ribonucleoproteins. Recently, it was suggested that the nucleosome might be the driving autoantigen in SLE. This hypothesis is supported by the finding that glomerular deposition of anti-dsDNA antibodies in lupus nephritis is mediated by nucleosomes [66, 67]. The kidneys are a major source of autoantibody-producing plasma cells in lupus nephritis and these differentiated plasma cells are frequently observed in patients with severe renal involvement (mainly classes III through V), potentially acting in amplifying the renal disease course [68]. Additional autoantibodies include anti-Smith (Sm) antibodies with a high specificity for SLE, while SSA and SSB are present in other CTDs as well [69]. Complement levels are frequently reduced in patients with active disease. Genetic complete complement deficiencies can resemble a SLE-like disease [70]. Levels of complement C3 and C4 correlate with the overall disease activity. Patients with active lupus nephritis had significantly lower levels of C3 and C4 compared to patients with inactive lupus nephritis [71]. Serum C3 has generally higher sensitivity than serum C4, but both tests have only modest specificity for active lupus nephritis [72]. Assessment of the relationship between serum levels of C3 or C4 and renal flares revealed that C4 is critical for initiating a renal flare, while C3 activation is involved in the actual tissue damage [73]. Antibodies directed against C1q were detected in all patients with active nephritis in a large cohort [74]. Moreover, anti-C1q antibodies showed the strongest association with proteinuria among potential biomarkers and were significantly correlated with Renal Activity Score [75]. However, contradictory to these reports, in a cohort of 126 patients, anti-C1q antibodies were not significantly associated with active lupus nephritis [76].

Histopathology/kidney involvement

Involvement of the kidney in the natural history of disease is present in a majority of patients and is supposed to appear in almost 50% in the first year of diagnosis [4]. Recent findings even suggest a higher incidence, since a considerable proportion of patients with SLE have silent lupus nephritis. Diagnosis in the latter group was significantly earlier compared to the overt lupus nephritis group and urinary sediment as well as renal function tests were normal [77]. Renal biopsy findings are categorized according to the current classification of lupus nephritis, which was published on behalf of the International Society of Nephrology (ISN)/Renal Pathology Society (RPS) [78] (Table 3). However, one should be aware that other glomerular changes, such as collapsing glomerulopathy [79], IgA nephropathy [80], FSGS, IgM nephropathy [81], minimal change disease [82]/glomerular podocytopathy [83] can occur as well and alterations in kidney function due to rhabdomyolysis with acute kidney failure [84], as well as type I and IV RTA [85], have also been reported.

Table 3 Revised classification lupus nephritis according to the International Society of Nephrology/Renal Pathology Society (ISN/RPS) 2003[78]

Therapy

In general, use of ACE inhibitors significantly reduced the development of proteinuria and/or biopsy-proven lupus nephritis and was associated with a decreased risk of disease activity [86]. Concomitant use of antimalarial drugs (chloroquine and hydroxychloroquine) at diagnosis of lupus nephritis reduced the risk of progression to end-stage renal failure and frequency of hypertension [87].

Specific treatment follows the class of lupus nephritis, which is defined by the revised ISN criteria. Class I and class II require no therapy directed at the kidney in consequence of good long-term renal outcome [88]. In contrast, high-dose steroid therapy rapidly resolved nephrotic syndrome in a majority of SLE patients with minimal change disease either in the absence or with underlying class II lupus nephritis based on renal biopsy findings [82, 83]. Immunosuppressive treatment is required in the management of class III (focal), class IV (diffuse) and class V (membranous nephropathy) lupus nephritis and usually consists of high dose glucocorticoid therapy along with intravenous CYC or mycophenolate mofetil (MMF) as induction therapy. The Euro Lupus Nephritis Trial compared low dose CYC (fortnightly, at a fixed dose of 500 mg, with a cumulative dose of 3 g) with the previously established high dose CYC (NIH) regimen (mean cumulative dose 8.5 g). Both strata were followed by AZA as remission-maintaining treatment. Renal outcome was similar in both treatment arms, but the low dose CYC group had fewer severe infections, even though the difference was not statistically significant [89]. Reports from this trial with a 10-year duration of follow-up confirmed the efficacy of the Euro Lupus regimen [90]. Since a majority of the subjects were white in this trial, these results might be less applicable to other ethnicities. In a more diversified cohort (>50% blacks) MMF (mean daily dosage 2.68 g) has been reported to be superior as induction therapy when compared to monthly CYC (0.5 g to 1 g/m2) in patients with class III through V lupus nephritis [91]. A further large, multi-center trial in a balanced cohort with respect to ethnicities designed to show superiority of MMF (mean daily dosage 2.47 g) to CYC failed to meet the primary end point. Both treatment arms achieved virtually identical rates of complete and partial remission. Furthermore, no significant difference with regard to severe adverse events or infections was reported [92]. Response to MMF as induction treatment in pure class V (membranous nephropathy) lupus nephritis in patients with diverse racial background appeared to show no difference in comparison to CYC [93]. Patients (>60% black) with class V lupus nephritis showed a better response regarding induction of remission after CSA when compared to CYC, whereas relapse of nephrotic syndrome occurred more frequently in patients with prior CSA therapy [94]. In a small cohort, multi-target therapy (MMF and tacrolimus) in patients with class IV and class V lupus nephritis revealed a higher rate of complete remission with a good tolerability when compared to intravenous CYC [95].

Following induction therapy, long-term immunosuppression is mandatory to avoid severe flares and to maintain stabilization of disease activity. Thus, immunosuppressants with a favorable safety profile and good efficacy are mandatory. MMF and AZA are deemed suitable and have shown efficacy in maintaining remission of lupus nephritis [96]. Equivalence of MMF and AZA was reported in the MAINTAIN Nephritis Trial, even though a trend towards fewer renal flares in the MMF group (19% vs. 25% in the AZA group) was reported [97]. More recently, in a larger trial, MMF was superior to AZA with respect to maintaining a renal response and preventing relapse in patients with lupus nephritis [98].

B-cell depleting therapy with anti-CD20 antibody rituximab (RTX) proved efficient in patients with active SLE including patients with lupus nephritis, who were nonresponsive to standard immunosuppressive therapy [99]. In proof of the efficacy of RTX treatment in moderately to severely active SLE and lupus nephritis, two large multi-center trials were conducted. The EXPLORER trial (moderate to severe active SLE) demonstrated no difference in primary/secondary end points between RTX and placebo. In a subgroup analysis a beneficial effect of RTX was observed in the African-American/Hispanic subgroup [100]. In patients with proliferative lupus nephritis and background immunosuppression (MMF) no difference was noted when RTX was added with regard to safety and efficacy (LUNAR trial) [101] even though opportunistic infections are reported to be rather common in SLE patients related to RTX treatment [102]. Enthusiasm was also dampened by reports on the development of progressive multifocal leukoencephalopathy in SLE patients following treatment with RTX [103].

Novel approaches with focus on targeted therapy have been developed and are currently being evaluated in clinical trials. Circulating B-lymphocyte stimulator (BLyS) is elevated in SLE, and titers correlate with increased disease activity and elevated dsDNA antibody concentrations [104]. Patients with serologically active SLE responded significantly better to belimumab, an antibody that binds to BLyS and inhibits its biological activity, plus standard of care (SOC) than to SOC alone [105]. The efficacy of belimumab was further corroborated in two large phase III trials, BLISS 52 [106] and BLISS 76 [107]. In both trials, belimumab met its primary efficacy end point and was consequently approved by the FDA in the treatment of SLE with the exception of severe active lupus nephritis or central nervous system lupus. Further investigations addressed to evaluate the role in active lupus nephritis are necessary.

Promising results have been obtained in a phase II trial for epratuzumab, a humanized anti-CD22 antibody [108, 109]. Atacicept, a soluble receptor fusion protein, neutralizes the activity of BLyS and a proliferation-inducing ligand (APRIL) and their heterotrimers [110]. In a phase I trial, atacicept was well tolerated and demonstrated a dose-dependent reduction of immunoglobulin levels and mature/total B cell numbers [111]. However, in patients with active lupus nephritis, a phase II trial was terminated due to an increased number of infections [109]. Further trials assessing the efficacy and safety are currently ongoing.

In patients with highly active lupus nephritis with failure of conventional therapy, short-term as well as prolonged immuno-adsorption led to a significant reduction in proteinuria and to sustained remission rates [112]. Autologous stem cell transplantation achieved sustained clinical remissions in patients refractory to conventional immunosuppressive treatment, even though this clinical benefit was associated with increased mortality rates in most studies conducted so far [113]. Intravenous immunoglobulins have shown benefits in patients nonresponsive to other therapies and as a steroid-sparing agent [114].

Conclusion

The histopathologic classification of lupus nephritis still guides the therapy. In proliferative lupus nephritis (III and IV), CYC and MMF have shown almost identical therapeutic responses as induction therapy in large trials. CSA might be an alternative to these immunosuppressive agents in pure class V lupus nephritis. In patients not responding to initial treatment, multi-target therapy might be an effective alternative. MMF seems to be superior to AZA in maintaining remission. The role of RTX in the treatment of lupus nephritis has to be further elucidated, as well as the significance of novel therapeutic approaches in the therapy of lupus nephritis.

Kidney disease in antiphospholipid syndrome

Introduction

Antiphospholipid syndrome (APS) is defined by the association of vascular thrombosis potentially affecting all segments of the vascular bed, complications during pregnancy (including unexplained consecutive spontaneous abortions, premature births because of severe preeclampsia, eclampsia or placental insufficiency or unexplained death before the 10th week of gestation), and the presence of antiphospholipid antibodies (aPL), namely anticardiolipin antibodies (aCL) and lupus anticoagulant (LAC) [115]. The APS is classified as primary APS in the absence of associated autoimmune disease, whereas secondary APS is found alongside other autoimmune disorders [116].

Histopathology/kidney involvement

Renal manifestations in the context of APS may result from thrombosis occurring at any location in the renal vasculature. Renal artery stenosis (RAS) is a common complication of APS, leading to renovascular hypertension [117]. In a retrospective study, patients with APS, RAS and hypertension receiving oral anticoagulation with a target trough International Normalized Ratio (INR) >3.0 had better blood pressure control and renal function remained stable or improved, while in patients with an INR <3.0 renal function significantly deteriorated and blood pressure was poorly controlled [118]. Arterial hypertension is a well-documented complication of APS. In a series of patients with primary APS, a large proportion of patients presented with hypertension, which was attributed to biopsy-proven vascular nephropathy [119]. Kleinknecht et al. reported that all patients had severe hypertension and renal insufficiency in a small cohort of patients with secondary APS due to SLE [120]. Thrombosis of the renal vein and inferior vena cava usually presents with nephrotic-range proteinuria in primary and secondary APS [121], especially in those with circulating LAC [122]. APSN refers to kidney damage caused by intrarenal vascular damage and may be acute, in case of the presence of thrombotic microangiopathy, and/or chronic, in the case of arteriosclerosis, fibrous intimal hyperplasia and focal cortical atrophy [119, 123]. Thrombotic microangiopathy is characterized by distinctive microscopic and ultrastructural changes and clinical presentation commonly includes hypertension, mild to nephrotic-range proteinuria and renal impairment [119, 123]. Tektonidou et al. examined kidney biopsies obtained from patients with SLE with or without presence of aPL. APSN was detected in almost 40% with aPL, compared with only 4.3% of patients without aPL [16]. Fakhouri et al. examined 29 kidney biopsies of patients with APS [124]. In nine of these biopsies predominant pathological features distinct from ASPN were noted: membranous nephropathy (three cases), minimal-change disease/focal segmental glomerulosclerosis (three cases), mesangial c3 nephropathy (two cases), and pauci-immune crescentic glomerulonephritis (one case). Furthermore, a case of fibrillary glomerulonephritis in a patient with APS was published recently [125]. Interestingly, the presence of aPL in patients undergoing renal transplantation significantly increases the risk of renal vascular thrombosis and graft failure [126, 127].

Therapy

Blood pressure control is the key intervention in the treatment of APS-related renal involvement. Adequate anticoagulation (if evidence of microthrombi is present) has shown encouraging results in small cohorts and may prevent progression to end-stage renal disease [128]. Evidence supporting immunosuppressive therapy in these patients is limited to case series [125, 129] and is not routinely recommended in APS-related renal manifestations. Contrasting, patients with catastrophic APS, which is characterized by severe multiple organ dysfunction in consequence of diffuse small vessel ischemia and thromboses predominantly affecting the parenchymal organs, usually receive a combination therapy, including anticoagulation, steroids, intravenous immunoglobulins and plasmapheresis, but despite this aggressive approach mortality is still high [130].

Conclusion

Blood pressure control is mandatory in patients with APSN. The role of anticoagulation with a target through INR above 3.0 in patients with APSN and microthrombi to prevent kidney function deterioration has to be elucidated in further, larger studies.

Rheumatoid arthritis

Introduction

RA is characterized by persistent synovial, systemic inflammation and autoantibodies (particularly to rheumatoid factor and citrullinated peptides). Genetic as well as environmental factors contribute to the risk of developing RA [131]. Renal involvement is relatively common in patients with RA.

Histopathology/kidney involvement

A study of renal biopsy specimens indicated that mesangial glomerulonephritis is the predominant histopathologic finding in RA, followed by amyloidosis, membranous nephropathy, focal proliferative glomerulonephritis, minimal-change nephropathy and acute interstitial nephritis [17]. Development of membranous nephropathy is related either to therapy with disease modifying antirheumatic drugs (DMARDs), in particular gold thiomalate, D-penicillamine and bucillamine [132], and anti-TNF alpha therapy, such as etanercept and adalimumab [133, 134], or rarely occurs concomitant with RA [135]. Secondary AA amyloidosis was prevalent in 5.8% of patients with RA and was accompanied by a shortened life expectancy [136]. Deposition of amyloid in renal tissue correlated significantly with parameters of renal function [132], while a lack of amyloid deposition in the glomerulus may characterize subjects with stable renal function [137]. Mesangial glomerulonephritis is probably related to RA itself, since its occurrence was associated with higher titers of rheumatoid factor (RF) when compared with RA patients without nephropathy. Deposition of mesangial IgA correlated with the duration of RA and elevated serum IgA levels, whereas mesangial IgM deposition was correlated with serum levels of IgM class RF [138]. In addition, single reports reveal the presence of FSGS [139] and fibrillary glomerulonephritis [140] in RA patients. Anti-TNF alpha therapy can be causative for the development of necrotizing crescentic glomerulonephritis and proliferative lupus nephritis [141, 142]. Besides the renal side effects of gold salts, D-penicillamine and bucillamine, CSA as another DMARD has a serious potential for renal toxicity, which is manifested primarily in a decline in creatinine clearance [143].

Therapy

Improvement of clinical and laboratory parameters was achieved in most cases after drug withdrawal and in case of necessary initiation of immunosuppression [133, 134, 142, 143]. In patients with amyloid deposition, etanercept treatment reduced proteinuria as well as serum amyloid A. Furthermore, it entailed a decrease in serum creatinine in patients with creatinine values <2.0 mg/dl at the onset of amyloidosis [144].

Conclusion

Therapy related deterioration of kidney function has to be excluded in patients with RA. In addition, the persisting inflammation can lead to deposition of amyloid. Thus, adequate therapy to reduce disease activity may be effective in preventing this late-onset complication. Specific therapeutic interventions should be tailored to the underlying histologic kidney involvement.

Conclusion and future directions

Renal involvement is frequently present in CTDs and has variable phenotypes. Since there is a steady increase of knowledge regarding the pathophysiology behind auto-immune disorders, more specific therapeutic approaches have been developed and are currently in clinical trials.

Acute or chronic TIN is the predominant kidney biopsy finding in Sjögren syndrome. Kidney function normalizes in most cases after corticosteroids are initiated [2, 6]. In addition, several glomerular lesion patterns have been described in Sjögren syndrome.

Results from hematopoietic stem cell transplantation (HSCT) in SSc are promising. Current studies, namely the SCOT and ASTIS trials, have completed patient recruitment and the first results are expected soon [145]. The ASSIST trial clearly depicted the efficacy of HSCT in patients with scleroderma, since all 10 patients in the HSCT-group improved when compared to none in the CYC–treated cohort [146]. In addition, endothelin receptor antagonists in combination with dual blockade of the renin-angiotensin-aldosterone system (RAAS) significantly reduced proteinuria and stabilized the serum creatinine level after an initial increase in a patient with SRC [147]. Despite efficacy in patients with pulmonary arterial hypertension in SSc [148], trials with the aim to show benefits of endothelin receptor antagonists in SRC have yet to be conducted.

Diverse glomerular alterations and rhabdomyolysis have been reported in patients with auto-immune myopathies. Guided therapy with the aim to treat the underlying disease improves kidney function in most cases.

In SLE, new therapeutic approaches have gained attention. One of these novel agents is belimumab, an inhibitor of serum BLyS, which was recently approved by the FDA for treatment of SLE with the exception of active lupus nephritis and central nervous system involvement. A randomized, controlled trial with inclusion of active lupus nephritis is currently being designed. Furthermore, BLyS inhibition may also be effective in the treatment of PSS, since patients with Sjögren syndrome exhibit increased BLyS levels [149]. In patients with SLE, B-cell depleting therapy with RTX was effective in a larger cohort including patients with lupus nephritis [99], and efficacy was furthermore confirmed in a recent meta-analysis evaluating patients with refractory lupus nephritis [150]. However, RTX failed to show superiority in two large phase III trials with patients either presenting without renal involvement (EXPLORER) or with renal involvement (LUNAR) [100, 101] even though a post hoc analysis of the EXPLORER trial indicated that RTX-treated patients achieved lower disease activity without a subsequent severe disease flare when compared to those treated with placebo [151]. Persistent B-cell presence was associated with no clinical response following RTX treatment [152]. In addition, physicians should be aware of severe infectious complications following RTX treatment in SLE patients [102, 103]. Despite other strategies, such as immunoglobulin administration, immuno-adsorption and stem cell transplantation [112114], RTX is nevertheless one alternative in refractory SLE [99].

APS-related renal manifestation potentially affects any segment of the vascular bed and is commonly accompanied by arterial hypertension. Blood pressure control is crucial, whereas the role and the target level of oral anticoagulation needs to be further elucidated. Chronic inflammation, as well as drug related adverse effects, is causative of kidney involvement in RA. Etanercept has shown encouraging results in reduction of serum amyloid A in amyloidosis and patients with a baseline serum creatinine below 2 mg/dl tended to show a benefit following TNF-alpha inhibition [144].

Based on studies in non-diabetic nephropathy, patients with renal involvement in CTDs should receive RAAS blocking agents once proteinuria is >1 g/day [149, 150]. Renal function needs to be monitored as well as serum potassium levels and blood pressure. In chronic kidney disease in the pre-dialysis state the lowering of LDL-cholesterol safely reduced the risk of major atherosclerotic events [153]. Accelerated atherosclerosis is a common finding in patients with chronic inflammation and in CTDs in particular [154]. Thus, modification of the risk factors contributing to the evolution of cardiovascular disease is crucial in these patients. Moreover, adherence to therapeutic advice may be an underestimated problem, since a recent study indicated that only one-quarter of patients with SLE had an adherence rate ≥80% [155]. In addition, counselling against smoking should be mandatory in patients with SLE and RA [156].

In summary, renal manifestations of CTDs are frequent. Renal biopsy to ensure diagnosis is necessary in most patients presenting with deterioration of renal function, increase of proteinuria or signs of nephritic syndrome (summarized in Table 4). An interdisciplinary approach to optimize treatment is the aim for patients with CTDs.

Table 4 Suggested kidney biopsy indications in connective tissue diseases

Abbreviations

AA:

Amyloid A

ACE:

Angiotensin-converting-enzyme

aCL:

Anticardiolipin antibodies

ACR:

American college of rheumatology

ANA:

Anti-nuclear antibodies

aPL:

Antiphospholipid antibodies

APRIL:

A proliferation-inducing ligand

APS:

Antiphospholipid syndrome

APSN:

Antiphospholipid syndrome nephropathy

AZA:

Azathioprine

BLyS:

B-lymphocyte stimulator

CSA:

Cyclosporine A

CTD:

Connective tissue disease

CTGF:

Connective tissue growth factor

CYC:

Cyclophosphamide

DM:

Dermatomyositis

DMARD:

Disease modifying antirheumatic drug

dsDNA:

Double-stranded DNA antibodies

EMT:

Epithelial to mesenchymal transition

FDA:

Food and drug administration

FSGS:

Focal segmental glomerulosclerosis

HSCT:

Hematopoietic stem cell transplantation

INR:

International normalized ratio

ISN:

International society of nephrology

LAC:

Lupus anticoagulant

LDL:

Low-density lipoprotein

MMF:

Mycophenolate mofetil

PM:

Polymyositis

PSS:

Primary sjögren syndrome

RA:

Rheumatoid arthritis

RAAS:

Renin-angiotensin-aldosterone system

RAS:

Renal artery stenosis

RF:

Rheumatoid factor

RPS:

Renal pathology society

RTA:

Renal tubular acidosis

RTX:

Rituximab

SLE:

Systemic lupus erythematosus

Sm:

Smith

SRC:

Scleroderma renal crisis

SOC:

Standard of care

SSc:

Systemic scleroderma

TGFß:

Transforming growth factor ß

TIN:

Tubulointerstitial nephritis

TNF:

Tumor-necrosis factor

UNOS:

United network of organ sharing.

References

  1. Goules A, Masouridi S, Tzioufas AG, Ioannidis JP, Skopouli FN, Moutsopoulos HM: Clinically significant and biopsy-documented renal involvement in primary Sjogren syndrome. Medicine (Baltimore). 2000, 79: 241-249.

    CAS  Google Scholar 

  2. Bossini N, Savoldi S, Franceschini F, Mombelloni S, Baronio M, Cavazzana I, Viola BF, Valzorio B, Mazzucchelli C, Cattaneo R, Scolari F, Maiorca R: Clinical and morphological features of kidney involvement in primary Sjogren's syndrome. Nephrol Dial Transplant. 2001, 16: 2328-2336.

    CAS  PubMed  Google Scholar 

  3. Penn H, Howie AJ, Kingdon EJ, Bunn CC, Stratton RJ, Black CM, Burns A, Denton CP: Scleroderma renal crisis: patient characteristics and long-term outcomes. QJM. 2007, 100: 485-494.

    CAS  PubMed  Google Scholar 

  4. Seshan SV, Jennette JC: Renal disease in systemic lupus erythematosus with emphasis on classification of lupus glomerulonephritis: advances and implications. Arch Pathol Lab Med. 2009, 133: 233-248.

    PubMed  Google Scholar 

  5. Sinico RA, Cavazzana I, Nuzzo M, Vianelli M, Napodano P, Scaini P, Tincani A: Renal involvement in primary antiphospholipid syndrome: retrospective analysis of 160 patients. Clin J Am Soc Nephrol. 2010, 5: 1211-1217.

    CAS  PubMed  PubMed Central  Google Scholar 

  6. Maripuri S, Grande JP, Osborn TG, Fervenza FC, Matteson EL, Donadio JV, Hogan MC: Renal involvement in primary Sjogren's syndrome: a clinicopathologic study. Clin J Am Soc Nephrol. 2009, 4: 1423-1431.

    PubMed  PubMed Central  Google Scholar 

  7. Ren H, Wang WM, Chen XN, Zhang W, Pan XX, Wang XL, Lin Y, Zhang S, Chen N: Renal involvement and followup of 130 patients with primary Sjogren's syndrome. J Rheumatol. 2008, 35: 278-284.

    CAS  PubMed  Google Scholar 

  8. Kawakami T, Ihn H, Xu W, Smith E, LeRoy C, Trojanowska M: Increased expression of TGF-beta receptors by scleroderma fibroblasts: evidence for contribution of autocrine TGF-beta signaling to scleroderma phenotype. J Invest Dermatol. 1998, 110: 47-51.

    CAS  PubMed  Google Scholar 

  9. Igarashi A, Nashiro K, Kikuchi K, Sato S, Ihn H, Grotendorst GR, Takehara K: Significant correlation between connective tissue growth factor gene expression and skin sclerosis in tissue sections from patients with systemic sclerosis. J Invest Dermatol. 1995, 105: 280-284.

    CAS  PubMed  Google Scholar 

  10. Mouthon L, Mehrenberger M, Teixeira L, Fakhouri F, Berezne A, Guillevin L, Noel LH: Endothelin-1 expression in scleroderma renal crisis. Hum Pathol. 2011, 42: 95-102.

    CAS  PubMed  Google Scholar 

  11. Helfrich DJ, Banner B, Steen VD, Medsger TA: Normotensive renal failure in systemic sclerosis. Arthritis Rheum. 1989, 32: 1128-1134.

    CAS  PubMed  Google Scholar 

  12. Teixeira L, Mouthon L, Mahr A, Berezne A, Agard C, Mehrenberger M, Noel LH, Trolliet P, Frances C, Cabane J, Guillevin L: Mortality and risk factors of scleroderma renal crisis: a French retrospective study of 50 patients. Ann Rheum Dis. 2008, 67: 110-116.

    CAS  PubMed  Google Scholar 

  13. Batal I, Domsic RT, Medsger TA, Bastacky S: Scleroderma renal crisis: a pathology perspective. Int J Rheumatol. 2010, 2010: 543704.

    PubMed  PubMed Central  Google Scholar 

  14. Yen TH, Lai PC, Chen CC, Hsueh S, Huang JY: Renal involvement in patients with polymyositis and dermatomyositis. Int J Clin Pract. 2005, 59: 188-193.

    PubMed  Google Scholar 

  15. Joshi D, Kumar N, Rai A: Dermatomyositis presenting with rhabdomyolysis and acute renal failure; an uncommon manifestation. Ann Indian Acad Neurol. 2009, 12: 45-47.

    PubMed  PubMed Central  Google Scholar 

  16. Tektonidou MG, Sotsiou F, Nakopoulou L, Vlachoyiannopoulos PG, Moutsopoulos HM: Antiphospholipid syndrome nephropathy in patients with systemic lupus erythematosus and antiphospholipid antibodies: prevalence, clinical associations, and long-term outcome. Arthritis Rheum. 2004, 50: 2569-2579.

    PubMed  Google Scholar 

  17. Helin HJ, Korpela MM, Mustonen JT, Pasternack AI: Renal biopsy findings and clinicopathologic correlations in rheumatoid arthritis. Arthritis Rheum. 1995, 38: 242-247.

    CAS  PubMed  Google Scholar 

  18. Ooms V, Decupere M, Lerut E, Vanrenterghem Y, Kuypers DR: Secondary renal amyloidosis due to long-standing tubulointerstitial nephritis in a patient with Sjogren syndrome. Am J Kidney Dis. 2005, 46: e75-e80.

    PubMed  Google Scholar 

  19. Roguedas AM, Youinou P, Lemasson G, Pennec YL, Misery L: Primary Gougerot-Sjogren syndrome: a dermatological approach. J Eur Acad Dermatol Venereol. 2006, 20: 243-247.

    PubMed  Google Scholar 

  20. Manthorpe R, Asmussen K, Oxholm P: Primary Sjogren's syndrome: diagnostic criteria, clinical features, and disease activity. J Rheumatol Suppl. 1997, 50: 8-11.

    CAS  PubMed  Google Scholar 

  21. Rehman HU: Sjogren's syndrome. Yonsei Med J. 2003, 44: 947-954.

    PubMed  Google Scholar 

  22. Yilmaz H, Kaya M, Ozbek M, Ureten K, Safa Yildirim I: Hypokalemic periodic paralysis in Sjogren's syndrome secondary to distal renal tubular acidosis. Rheumatol Int. 2012, Epub ahead of print

    Google Scholar 

  23. Kobayashi T, Muto S, Nemoto J, Miyata Y, Ishiharajima S, Hironaka M, Asano Y, Kusano E: Fanconi's syndrome and distal (type 1) renal tubular acidosis in a patient with primary Sjogren's syndrome with monoclonal gammopathy of undetermined significance. Clin Nephrol. 2006, 65: 427-432.

    CAS  PubMed  Google Scholar 

  24. Yang ML, Kuo MC, Ou TT, Chen HC: Primary Sjogren's syndrome with minimal change disease–a case report. Kaohsiung J Med Sci. 2011, 27: 190-194.

    PubMed  Google Scholar 

  25. Aasarod K, Haga HJ, Berg KJ, Hammerstrom J, Jorstad S: Renal involvement in primary Sjogren's syndrome. QJM. 2000, 93: 297-304.

    CAS  PubMed  Google Scholar 

  26. Pessler F, Emery H, Dai L, Wu YM, Monash B, Cron RQ, Pradhan M: The spectrum of renal tubular acidosis in paediatric Sjogren syndrome. Rheumatology (Oxford). 2006, 45: 85-91.

    CAS  Google Scholar 

  27. Hunzelmann N, Krieg T: Scleroderma: from pathophysiology to novel therapeutic approaches. Exp Dermatol. 2010, 19: 393-400.

    CAS  PubMed  Google Scholar 

  28. Strange G, Nash P: The manifestations of vasculopathy in systemic sclerosis and its evidence-based therapy. Int J Rheum Dis. 2009, 12: 192-206.

    PubMed  Google Scholar 

  29. Walker UA, Tyndall A, Czirjak L, Denton C, Farge-Bancel D, Kowal-Bielecka O, Muller-Ladner U, Bocelli-Tyndall C, Matucci-Cerinic M: Clinical risk assessment of organ manifestations in systemic sclerosis: a report from the EULAR scleroderma trials and research group database. Ann Rheum Dis. 2007, 66: 754-763.

    CAS  PubMed  PubMed Central  Google Scholar 

  30. Liu Y: New insights into epithelial-mesenchymal transition in kidney fibrosis. J Am Soc Nephrol. 2010, 21: 212-222.

    CAS  PubMed  Google Scholar 

  31. Postlethwaite AE, Shigemitsu H, Kanangat S: Cellular origins of fibroblasts: possible implications for organ fibrosis in systemic sclerosis. Curr Opin Rheumatol. 2004, 16: 733-738.

    PubMed  Google Scholar 

  32. Steen VD, Medsger TA, Osial TA, Ziegler GL, Shapiro AP, Rodnan GP: Factors predicting development of renal involvement in progressive systemic sclerosis. Am J Med. 1984, 76: 779-786.

    CAS  PubMed  Google Scholar 

  33. Nguyen B, Mayes MD, Arnett FC, Del Junco D, Reveille JD, Gonzalez EB, Draeger HT, Perry M, Hendiani A, Anand KK, Assassi S: HLA-DRB1*0407 and *1304 are risk factors for scleroderma renal crisis. Arthritis Rheum. 2011, 63: 530-534.

    PubMed  PubMed Central  Google Scholar 

  34. Steen VD, Medsger TA: Case–control study of corticosteroids and other drugs that either precipitate or protect from the development of scleroderma renal crisis. Arthritis Rheum. 1998, 41: 1613-1619.

    CAS  PubMed  Google Scholar 

  35. Arad U, Balbir-Gurman A, Doenyas-Barak K, Amit-Vazina M, Caspi D, Elkayam O: Anti-neutrophil antibody associated vasculitis in systemic sclerosis. Semin Arthritis Rheum. 2011, 41: 223-229.

    CAS  PubMed  Google Scholar 

  36. Bussone G, Berezne A, Pestre V, Guillevin L, Mouthon L: The scleroderma kidney: progress in risk factors, therapy, and prevention. Curr Rheumatol Rep. 2011, 13: 37-43.

    PubMed  Google Scholar 

  37. Steen VD, Medsger TA: Long-term outcomes of scleroderma renal crisis. Ann Intern Med. 2000, 133: 600-603.

    CAS  PubMed  Google Scholar 

  38. Pham PT, Pham PC, Danovitch GM, Gritsch HA, Singer J, Wallace WD, Hayashi R, Wilkinson AH: Predictors and risk factors for recurrent scleroderma renal crisis in the kidney allograft: case report and review of the literature. Am J Transplant. 2005, 5: 2565-2569.

    PubMed  Google Scholar 

  39. Denton CP, Sweny P, Abdulla A, Black CM: Acute renal failure occurring in scleroderma treated with cyclosporin A: a report of three cases. Br J Rheumatol. 1994, 33: 90-92.

    CAS  PubMed  Google Scholar 

  40. Mammen AL: Dermatomyositis and polymyositis: clinical presentation, autoantibodies, and pathogenesis. Ann N Y Acad Sci. 2010, 1184: 134-153.

    CAS  PubMed  Google Scholar 

  41. Clarke JT, Werth VP: Rheumatic manifestations of skin disease. Curr Opin Rheumatol. 2010, 22: 78-84.

    CAS  PubMed  PubMed Central  Google Scholar 

  42. Kuo CF, See LC, Yu KH, Chou IJ, Chang HC, Chiou MJ, Luo SF: Incidence, cancer risk and mortality of dermatomyositis and polymyositis in Taiwan: a nationwide population study. Br J Dermatol. 2011, 165: 1273-1279.

    PubMed  Google Scholar 

  43. Chen YJ, Wu CY, Huang YL, Wang CB, Shen JL, Chang YT: Cancer risks of dermatomyositis and polymyositis: a nationwide cohort study in Taiwan. Arthritis Res Ther. 2010, 12: R70.

    PubMed  PubMed Central  Google Scholar 

  44. Stockton D, Doherty VR, Brewster DH: Risk of cancer in patients with dermatomyositis or polymyositis, and follow-up implications: a Scottish population-based cohort study. Br J Cancer. 2001, 85: 41-45.

    CAS  PubMed  PubMed Central  Google Scholar 

  45. Hill CL, Zhang Y, Sigurgeirsson B, Pukkala E, Mellemkjaer L, Airio A, Evans SR, Felson DT: Frequency of specific cancer types in dermatomyositis and polymyositis: a population-based study. Lancet. 2001, 357: 96-100.

    CAS  PubMed  Google Scholar 

  46. Frost NA, Morand EF, Hall CL, Maddison PJ, Bhalla AK: Idiopathic polymyositis complicated by arthritis and mesangial proliferative glomerulonephritis: case report and review of the literature. Br J Rheumatol. 1993, 32: 929-931.

    CAS  PubMed  Google Scholar 

  47. Valenzuela OF, Reiser IW, Porush JG: Idiopathic polymyositis and glomerulonephritis. J Nephrol. 2001, 14: 120-124.

    CAS  PubMed  Google Scholar 

  48. Takizawa Y, Kanda H, Sato K, Kawahata K, Yamaguchi A, Uozaki H, Shimizu J, Tsuji S, Misaki Y, Yamamoto K: Polymyositis associated with focal mesangial proliferative glomerulonephritis with depositions of immune complexes. Clin Rheumatol. 2007, 26: 792-796.

    PubMed  Google Scholar 

  49. Xie Q, Liu Y, Liu G, Yang N, Yin G: Diffuse proliferative glomerulonephritis associated with dermatomyositis with nephrotic syndrome. Rheumatol Int. 2010, 30: 821-825.

    PubMed  Google Scholar 

  50. Dyck RF, Katz A, Gordon DA, Johnson M, Shainhouse Z, Cardella CJ, Bear RA: Glomerulonephritis associated with polymyositis. J Rheumatol. 1979, 6: 336-344.

    CAS  PubMed  Google Scholar 

  51. Carleton H, Pitts W, Davidson W, Roth P: Muscle disease associated with renal failure. Arch Intern Med. 1977, 137: 939-941.

    CAS  PubMed  Google Scholar 

  52. Moutsopoulos H, Fye KH: Letter: Lipoid nephrosis and focal glomerulosclerosis in a patient with polymyositis. Lancet. 1975, 1: 1039.

    CAS  PubMed  Google Scholar 

  53. Hara I, Kurata N, Hyozu K, Tamao H: A case of polymyositis complicated with membranous nephritis. Nippon Naika Gakkai Zasshi. 1991, 80: 108-109.

    CAS  PubMed  Google Scholar 

  54. Tsunemi M, Ishimura E, Tsumura K, Shoji S, Sugimura T, Nishizawa Y, Morii H: A case of crescentic glomerulonephritis associated with polymyositis. Nephron. 1993, 64: 488-489.

    CAS  PubMed  Google Scholar 

  55. Fukui H, Kimura T, Nakabayashi K, Nagasawa T: Dermatomyositis associated with immune-complex type nephritis induced by tubular epithelial antigen. Nihon Jinzo Gakkai Shi. 1976, 18: 523-533.

    CAS  PubMed  Google Scholar 

  56. Moriyama T, Uruta Y, Yamaguchi H, Fukuzaki M, Uchida Y, Yasumoto Y, Yamashita W, Harada R, Ohsaki K, Nakajima A: A case of immune-complex type glomerulonephritis associated with dermatomyositis. Nippon Naika Gakkai Zasshi. 1989, 78: 994-995.

    CAS  PubMed  Google Scholar 

  57. Makino H, Hirata K, Matsuda M, Amano T, Ota Z: Membranous nephropathy developing during the course of dermatomyositis. J Rheumatol. 1994, 21: 1377-1378.

    CAS  PubMed  Google Scholar 

  58. Yen TH, Huang JY, Chen CY: Unexpected IgA nephropathy during the treatment of a young woman with idiopathic dermatomyositis: case report and review of the literature. J Nephrol. 2003, 16: 148-153.

    PubMed  Google Scholar 

  59. Kamata K, Kobayashi Y, Shigematsu H, Saito T: Childhood type polymyositis and rapidly progressive glomerulonephritis. Acta Pathol Jpn. 1982, 32: 801-806.

    CAS  PubMed  Google Scholar 

  60. Hochberg MC: Updating the american college of rheumatology revised criteria for the classification of systemic lupus erythematosus. Arthritis Rheum. 1997, 40: 1725.

    CAS  PubMed  Google Scholar 

  61. Cervera R, Khamashta MA, Font J, Sebastiani GD, Gil A, Lavilla P, Mejia JC, Aydintug AO, Chwalinska-Sadowska H, de Ramon E, Fernandez-Nebro A, Galeazzi M, Valen M, Mathieu A, Houssiau F, Caro N, Alba P, Ramos-Casals M, Ingelmo M, Hughes GR: Morbidity and mortality in systemic lupus erythematosus during a 10-year period: a comparison of early and late manifestations in a cohort of 1,000 patients. Medicine (Baltimore). 2003, 82: 299-308.

    Google Scholar 

  62. Doria A, Iaccarino L, Ghirardello A, Zampieri S, Arienti S, Sarzi-Puttini P, Atzeni F, Piccoli A, Todesco S: Long-term prognosis and causes of death in systemic lupus erythematosus. Am J Med. 2006, 119: 700-706.

    PubMed  Google Scholar 

  63. Pons-Estel GJ, Alarcon GS, Scofield L, Reinlib L, Cooper GS: Understanding the epidemiology and progression of systemic lupus erythematosus. Semin Arthritis Rheum. 2010, 39: 257-268.

    PubMed  Google Scholar 

  64. Rubtsov AV, Rubtsova K, Kappler JW, Marrack P: Genetic and hormonal factors in female-biased autoimmunity. Autoimmun Rev. 2010, 9: 494-498.

    CAS  PubMed  PubMed Central  Google Scholar 

  65. Cooper GS, Wither J, Bernatsky S, Claudio JO, Clarke A, Rioux JD, Fortin PR: Occupational and environmental exposures and risk of systemic lupus erythematosus: silica, sunlight, solvents. Rheumatology (Oxford). 2010, 49: 2172-2180.

    CAS  Google Scholar 

  66. van Bavel CC, Fenton KA, Rekvig OP, van der Vlag J, Berden JH: Glomerular targets of nephritogenic autoantibodies in systemic lupus erythematosus. Arthritis Rheum. 2008, 58: 1892-1899.

    CAS  PubMed  Google Scholar 

  67. Dieker JW, van der Vlag J, Berden JH: Triggers for anti-chromatin autoantibody production in SLE. Lupus. 2002, 11: 856-864.

    CAS  PubMed  Google Scholar 

  68. Espeli M, Bokers S, Giannico G, Dickinson HA, Bardsley V, Fogo AB, Smith KG: Local renal autoantibody production in lupus nephritis. J Am Soc Nephrol. 2011, 22: 296-305.

    PubMed  PubMed Central  Google Scholar 

  69. Egner W: The use of laboratory tests in the diagnosis of SLE. J Clin Pathol. 2000, 53: 424-432.

    CAS  PubMed  PubMed Central  Google Scholar 

  70. Aggarwal R, Sestak AL, D'Sousa A, Dillon SP, Namjou B, Scofield RH: Complete complement deficiency in a large cohort of familial systemic lupus erythematosus. Lupus. 2010, 19: 52-57.

    CAS  PubMed  Google Scholar 

  71. Julkunen H, Ekblom-Kullberg S, Miettinen A: Nonrenal and renal activity of systemic lupus erythematosus: a comparison of two anti-C1q and five anti-dsDNA assays and complement C3 and C4. Rheumatol Int. 2012, 32: 2445-2451.

    CAS  PubMed  Google Scholar 

  72. Bertsias GK, Tektonidou M, Amoura Z, Aringer M, Bajema I, Berden JH, Boletis J, Cervera R, Dorner T, Doria A, Ferrario F, Floege J, Houssiau F, Ioannidis JP, Isenberg DA, Kallenberg CG, Lightstone L, Marks SD, Martini A, Moroni G, Neumann I, Praga M, Schneider M, Starra A, Tesar V, Vasconcelos C, van Vollenhoven RF, Zakharova H, Haubitz M, Gordon C: Joint european league against rheumatism and european renal association-european dialysis and transplant association (EULAR/ERA-EDTA) recommendations for the management of adult and paediatric lupus nephritis. Ann Rheum Dis. 2012, 71: 1771-1782.

    CAS  PubMed  PubMed Central  Google Scholar 

  73. Birmingham DJ, Irshaid F, Nagaraja HN, Zou X, Tsao BP, Wu H, Yu CY, Hebert LA, Rovin BH: The complex nature of serum C3 and C4 as biomarkers of lupus renal flare. Lupus. 2010, 19: 1272-1280.

    CAS  PubMed  PubMed Central  Google Scholar 

  74. Oelzner P, Deliyska B, Funfstuck R, Hein G, Herrmann D, Stein G: Anti-C1q antibodies and antiendothelial cell antibodies in systemic lupus erythematosus - relationship with disease activity and renal involvement. Clin Rheumatol. 2003, 22: 271-278.

    CAS  PubMed  Google Scholar 

  75. Akhter E, Burlingame RW, Seaman AL, Magder L, Petri M: Anti-C1q antibodies have higher correlation with flares of lupus nephritis than other serum markers. Lupus. 2011, 20: 1267-1274.

    CAS  PubMed  Google Scholar 

  76. Katsumata Y, Miyake K, Kawaguchi Y, Okamoto Y, Kawamoto M, Gono T, Baba S, Hara M, Yamanaka H: Anti-C1q antibodies are associated with systemic lupus erythematosus global activity but not specifically with nephritis: a controlled study of 126 consecutive patients. Arthritis Rheum. 2011, 63: 2436-2444.

    CAS  PubMed  Google Scholar 

  77. Zabaleta-Lanz ME, Munoz LE, Tapanes FJ, Vargas-Arenas RE, Daboin I, Barrios Y, Pinto JA, Bianco NE: Further description of early clinically silent lupus nephritis. Lupus. 2006, 15: 845-851.

    CAS  PubMed  Google Scholar 

  78. Weening JJ, D'Agati VD, Schwartz MM, Seshan SV, Alpers CE, Appel GB, Balow JE, Bruijn JA, Cook T, Ferrario F, Fogo AB, Ginzler EM, Hebert L, Hill G, Hill P, Jennette JC, Kong NC, Lesavre P, Lockshin M, Looi LM, Makino H, Moura LA, Nagata M: The classification of glomerulonephritis in systemic lupus erythematosus revisited. J Am Soc Nephrol. 2004, 15: 241-250.

    PubMed  Google Scholar 

  79. Gupta R, Sharma A, Bhowmik D, Gupta S, Agarwal S, Dinda A: Collapsing glomerulopathy occurring in HIV-negative patients with systemic lupus erythematosus: report of three cases and brief review of the literature. Lupus. 2011, 20: 866-870.

    CAS  PubMed  Google Scholar 

  80. Mac-Moune Lai F, Li EK, Tang NL, Li PK, Lui SF, Lai KN: IgA nephropathy: a rare lesion in systemic lupus erythematosus. Mod Pathol. 1995, 8: 5-10.

    CAS  PubMed  Google Scholar 

  81. Baranowska-Daca E, Choi YJ, Barrios R, Nassar G, Suki WN, Truong LD: Nonlupus nephritides in patients with systemic lupus erythematosus: a comprehensive clinicopathologic study and review of the literature. Hum Pathol. 2001, 32: 1125-1135.

    CAS  PubMed  Google Scholar 

  82. Dube GK, Markowitz GS, Radhakrishnan J, Appel GB, D'Agati VD: Minimal change disease in systemic lupus erythematosus. Clin Nephrol. 2002, 57: 120-126.

    CAS  PubMed  Google Scholar 

  83. Kraft SW, Schwartz MM, Korbet SM, Lewis EJ: Glomerular podocytopathy in patients with systemic lupus erythematosus. J Am Soc Nephrol. 2005, 16: 175-179.

    PubMed  Google Scholar 

  84. de Carvalho JF, da Mota LM, Bonfa E: Fatal rhabdomyolysis in systemic lupus erythematosus. Rheumatol Int. 2011, 31: 1243-1245.

    PubMed  Google Scholar 

  85. Li SL, Liou LB, Fang JT, Tsai WP: Symptomatic renal tubular acidosis (RTA) in patients with systemic lupus erythematosus: an analysis of six cases with new association of type 4 RTA. Rheumatology (Oxford). 2005, 44: 1176-1180.

    CAS  Google Scholar 

  86. Duran-Barragan S, McGwin G, Vila LM, Reveille JD, Alarcon GS: Angiotensin-converting enzyme inhibitors delay the occurrence of renal involvement and are associated with a decreased risk of disease activity in patients with systemic lupus erythematosus–results from LUMINA (LIX): a multiethnic US cohort. Rheumatology (Oxford). 2008, 47: 1093-1096.

    CAS  Google Scholar 

  87. Siso A, Ramos-Casals M, Bove A, Brito-Zeron P, Soria N, Munoz S, Testi A, Plaza J, Sentis J, Coca A: Previous antimalarial therapy in patients diagnosed with lupus nephritis: influence on outcomes and survival. Lupus. 2008, 17: 281-288.

    CAS  PubMed  Google Scholar 

  88. Bomback AS, Appel GB: Updates on the treatment of lupus nephritis. J Am Soc Nephrol. 2010, 21: 2028-2035.

    CAS  PubMed  Google Scholar 

  89. Houssiau FA, Vasconcelos C, D'Cruz D, Sebastiani GD, Garrido Ed Ede R, Danieli MG, Abramovicz D, Blockmans D, Mathieu A, Direskeneli H, Galeazzi M, Gul A, Levy Y, Petera P, Popovic R, Petrovic R, Sinico RA, Cattaneo R, Font J, Depresseux G, Cosyns JP, Cervera R: Immunosuppressive therapy in lupus nephritis: the euro-lupus nephritis trial, a randomized trial of low-dose versus high-dose intravenous cyclophosphamide. Arthritis Rheum. 2002, 46: 2121-2131.

    CAS  PubMed  Google Scholar 

  90. Houssiau FA, Vasconcelos C, D'Cruz D, Sebastiani GD, de Ramon Garrido E, Danieli MG, Abramovicz D, Blockmans D, Cauli A, Direskeneli H, Galeazzi M, Gul A, Levy Y, Petera P, Popovic R, Petrovic R, Sinico RA, Cattaneo R, Font J, Depresseux G, Cosyns JP, Cervera R: The 10-year follow-up data of the euro-lupus nephritis trial comparing low-dose and high-dose intravenous cyclophosphamide. Ann Rheum Dis. 2010, 69: 61-64.

    CAS  PubMed  Google Scholar 

  91. Ginzler EM, Dooley MA, Aranow C, Kim MY, Buyon J, Merrill JT, Petri M, Gilkeson GS, Wallace DJ, Weisman MH, Appel GB: Mycophenolate mofetil or intravenous cyclophosphamide for lupus nephritis. N Engl J Med. 2005, 353: 2219-2228.

    CAS  PubMed  Google Scholar 

  92. Appel GB, Contreras G, Dooley MA, Ginzler EM, Isenberg D, Jayne D, Li LS, Mysler E, Sanchez-Guerrero J, Solomons N, Wofsy D: Mycophenolate mofetil versus cyclophosphamide for induction treatment of lupus nephritis. J Am Soc Nephrol. 2009, 20: 1103-1112.

    CAS  PubMed  PubMed Central  Google Scholar 

  93. Radhakrishnan J, Moutzouris DA, Ginzler EM, Solomons N, Siempos II, Appel GB: Mycophenolate mofetil and intravenous cyclophosphamide are similar as induction therapy for class V lupus nephritis. Kidney Int. 2010, 77: 152-160.

    CAS  PubMed  Google Scholar 

  94. Austin HA, Illei GG, Braun MJ, Balow JE: Randomized, controlled trial of prednisone, cyclophosphamide, and cyclosporine in lupus membranous nephropathy. J Am Soc Nephrol. 2009, 20: 901-911.

    CAS  PubMed  PubMed Central  Google Scholar 

  95. Bao H, Liu ZH, Xie HL, Hu WX, Zhang HT, Li LS: Successful treatment of class V + IV lupus nephritis with multitarget therapy. J Am Soc Nephrol. 2008, 19: 2001-2010.

    CAS  PubMed  PubMed Central  Google Scholar 

  96. Contreras G, Tozman E, Nahar N, Metz D: Maintenance therapies for proliferative lupus nephritis: mycophenolate mofetil, azathioprine and intravenous cyclophosphamide. Lupus. 2005, 14: s33-s38.

    CAS  PubMed  Google Scholar 

  97. Houssiau FA, D'Cruz D, Sangle S, Remy P, Vasconcelos C, Petrovic R, Fiehn C, de Ramon Garrido E, Gilboe IM, Tektonidou M, Blockmans D, Ravelingien I, le Guern V, Depresseux G, Guillevin L, Cervera R: Azathioprine versus mycophenolate mofetil for long-term immunosuppression in lupus nephritis: results from the MAINTAIN nephritis trial. Ann Rheum Dis. 2010, 69: 2083-2089.

    CAS  PubMed  PubMed Central  Google Scholar 

  98. Dooley MA, Jayne D, Ginzler EM, Isenberg D, Olsen NJ, Wofsy D, Eitner F, Appel GB, Contreras G, Lisk L, Solomons N: Mycophenolate versus azathioprine as maintenance therapy for lupus nephritis. N Engl J Med. 2011, 365: 1886-1895.

    CAS  PubMed  Google Scholar 

  99. Lu TY, Ng KP, Cambridge G, Leandro MJ, Edwards JC, Ehrenstein M, Isenberg DA: A retrospective seven-year analysis of the use of B cell depletion therapy in systemic lupus erythematosus at university college london hospital: the first fifty patients. Arthritis Rheum. 2009, 61: 482-487.

    PubMed  Google Scholar 

  100. Merrill JT, Neuwelt CM, Wallace DJ, Shanahan JC, Latinis KM, Oates JC, Utset TO, Gordon C, Isenberg DA, Hsieh HJ, Zhang D, Brunetta PG: Efficacy and safety of rituximab in moderately-to-severely active systemic lupus erythematosus: the randomized, double-blind, phase II/III systemic lupus erythematosus evaluation of rituximab trial. Arthritis Rheum. 2010, 62: 222-233.

    CAS  PubMed  PubMed Central  Google Scholar 

  101. Rovin BH, Furie R, Latinis K, Looney RJ, Fervenza FC, Sanchez-Guerrero J, Maciuca R, Zhang D, Garg JP, Brunetta P, Appel G: Efficacy and safety of rituximab in patients with active proliferative lupus nephritis: The lupus nephritis assessment with rituximab (LUNAR) study. Arthritis Rheum. 2012, 64: 1215-1226.

    CAS  PubMed  Google Scholar 

  102. Tony HP, Burmester G, Schulze-Koops H, Grunke M, Henes J, Kotter I, Haas J, Unger L, Lovric S, Haubitz M, Fischer-Betz R, Chehab G, Rubbert-Roth A, Specker C, Weinerth J, Holle J, Muller-Ladner U, Konig R, Fiehn C, Burgwinkel P, Budde K, Sorensen H, Meurer M, Aringer M, Kieseier B, Erfurt-Berge C, Sticherling M, Veelken R, Ziemann U, Strutz F: Safety and clinical outcomes of rituximab therapy in patients with different autoimmune diseases: experience from a national registry (GRAID). Arthritis Res Ther. 2011, 13: R75.

    CAS  PubMed  PubMed Central  Google Scholar 

  103. Molloy ES, Calabrese LH: Progressive multifocal leukoencephalopathy: a national estimate of frequency in systemic lupus erythematosus and other rheumatic diseases. Arthritis Rheum. 2009, 60: 3761-3765.

    PubMed  Google Scholar 

  104. Petri M, Stohl W, Chatham W, McCune WJ, Chevrier M, Ryel J, Recta V, Zhong J, Freimuth W: Association of plasma B lymphocyte stimulator levels and disease activity in systemic lupus erythematosus. Arthritis Rheum. 2008, 58: 2453-2459.

    CAS  PubMed  Google Scholar 

  105. Wallace DJ, Stohl W, Furie RA, Lisse JR, McKay JD, Merrill JT, Petri MA, Ginzler EM, Chatham WW, McCune WJ, Fernandez V, Chevrier MR, Zhong ZJ, Freimuth WW: A phase II, randomized, double-blind, placebo-controlled, dose-ranging study of belimumab in patients with active systemic lupus erythematosus. Arthritis Rheum. 2009, 61: 1168-1178.

    CAS  PubMed  PubMed Central  Google Scholar 

  106. Navarra SV, Guzman RM, Gallacher AE, Hall S, Levy RA, Jimenez RE, Li EK, Thomas M, Kim HY, Leon MG, Tanasescu C, Nasonov E, Lan JL, Pineda L, Zhong ZJ, Freimuth W, Petri MA: Efficacy and safety of belimumab in patients with active systemic lupus erythematosus: a randomised, placebo-controlled, phase 3 trial. Lancet. 2011, 377: 721-731.

    CAS  PubMed  Google Scholar 

  107. Furie R, Petri M, Zamani O, Cervera R, Wallace DJ, Tegzova D, Sanchez-Guerrero J, Schwarting A, Merrill JT, Chatham WW, Stohl W, Ginzler EM, Hough DR, Zhong ZJ, Freimuth W, van Vollenhoven RF: A phase III, randomized, placebo-controlled study of belimumab, a monoclonal antibody that inhibits B lymphocyte stimulator, in patients with systemic lupus erythematosus. Arthritis Rheum. 2011, 63: 3918-3930.

    CAS  PubMed  Google Scholar 

  108. Dorner T, Kaufmann J, Wegener WA, Teoh N, Goldenberg DM, Burmester GR: Initial clinical trial of epratuzumab (humanized anti-CD22 antibody) for immunotherapy of systemic lupus erythematosus. Arthritis Res Ther. 2006, 8: R74.

    PubMed  PubMed Central  Google Scholar 

  109. Yildirim-Toruner C, Diamond B: Current and novel therapeutics in the treatment of systemic lupus erythematosus. J Allergy Clin Immunol. 2011, 127: 303-312. quiz 313–304

    CAS  PubMed  PubMed Central  Google Scholar 

  110. Dillon SR, Harder B, Lewis KB, Moore MD, Liu H, Bukowski TR, Hamacher NB, Lantry MM, Maurer M, Krejsa CM, Ellsworth JL, Pederson S, Elkon KB, Wener MH, Dall'Era M, Gross JA: B-lymphocyte stimulator/a proliferation-inducing ligand heterotrimers are elevated in the sera of patients with autoimmune disease and are neutralized by atacicept and B-cell maturation antigen-immunoglobulin. Arthritis Res Ther. 2010, 12: R48.

    PubMed  PubMed Central  Google Scholar 

  111. Dall'Era M, Chakravarty E, Wallace D, Genovese M, Weisman M, Kavanaugh A, Kalunian K, Dhar P, Vincent E, Pena-Rossi C, Wofsy D: Reduced B lymphocyte and immunoglobulin levels after atacicept treatment in patients with systemic lupus erythematosus: results of a multicenter, phase Ib, double-blind, placebo-controlled, dose-escalating trial. Arthritis Rheum. 2007, 56: 4142-4150.

    PubMed  Google Scholar 

  112. Stummvoll GH, Schmaldienst S, Smolen JS, Derfler K, Biesenbach P: Lupus nephritis: prolonged immunoadsorption (IAS) reduces proteinuria and stabilizes global disease activity. Nephrol Dial Transplant. 2012, 27: 618-626.

    CAS  PubMed  Google Scholar 

  113. Illei GG, Cervera R, Burt RK, Doria A, Hiepe F, Jayne D, Pavletic S, Martin T, Marmont A, Saccardi R, Voskuyl AE, Farge D: Current state and future directions of autologous hematopoietic stem cell transplantation in systemic lupus erythematosus. Ann Rheum Dis. 2011, 70: 2071-2074.

    PubMed  PubMed Central  Google Scholar 

  114. Toubi E, Kessel A, Shoenfeld Y: High-dose intravenous immunoglobulins: an option in the treatment of systemic lupus erythematosus. Hum Immunol. 2005, 66: 395-402.

    CAS  PubMed  Google Scholar 

  115. Wilson WA, Gharavi AE, Koike T, Lockshin MD, Branch DW, Piette JC, Brey R, Derksen R, Harris EN, Hughes GR, Triplett DA, Khamashta MA: International consensus statement on preliminary classification criteria for definite antiphospholipid syndrome: report of an international workshop. Arthritis Rheum. 1999, 42: 1309-1311.

    CAS  PubMed  Google Scholar 

  116. Gigante A, Gasperini ML, Cianci R, Barbano B, Giannakakis K, Di Donato D, Fuiano G, Amoroso A: Antiphospholipid antibodies and renal involvement. Am J Nephrol. 2009, 30: 405-412.

    CAS  PubMed  Google Scholar 

  117. Sangle SR, D'Cruz DP, Jan W, Karim MY, Khamashta MA, Abbs IC, Hughes GR: Renal artery stenosis in the antiphospholipid (Hughes) syndrome and hypertension. Ann Rheum Dis. 2003, 62: 999-1002.

    CAS  PubMed  PubMed Central  Google Scholar 

  118. Sangle SR, D'Cruz DP, Abbs IC, Khamashta MA, Hughes GR: Renal artery stenosis in hypertensive patients with antiphospholipid (Hughes) syndrome: outcome following anticoagulation. Rheumatology (Oxford). 2005, 44: 372-377.

    CAS  Google Scholar 

  119. Nochy D, Daugas E, Droz D, Beaufils H, Grunfeld JP, Piette JC, Bariety J, Hill G: The intrarenal vascular lesions associated with primary antiphospholipid syndrome. J Am Soc Nephrol. 1999, 10: 507-518.

    CAS  PubMed  Google Scholar 

  120. Kleinknecht D, Bobrie G, Meyer O, Noel LH, Callard P, Ramdane M: Recurrent thrombosis and renal vascular disease in patients with a lupus anticoagulant. Nephrol Dial Transplant. 1989, 4: 854-858.

    CAS  PubMed  Google Scholar 

  121. Uthman I, Khamashta M: Antiphospholipid syndrome and the kidneys. Semin Arthritis Rheum. 2006, 35: 360-367.

    CAS  PubMed  Google Scholar 

  122. Mintz G, Acevedo-Vazquez E, Gutierrez-Espinosa G, Avelar-Garnica F: Renal vein thrombosis and inferior vena cava thrombosis in systemic lupus erythematosus. Frequency and risk factors. Arthritis Rheum. 1984, 27: 539-544.

    CAS  PubMed  Google Scholar 

  123. Amigo MC: Kidney disease in antiphospholipid syndrome. Rheum Dis Clin North Am. 2006, 32: 509-522.

    PubMed  Google Scholar 

  124. Fakhouri F, Noel LH, Zuber J, Beaufils H, Martinez F, Lebon P, Papo T, Chauveau D, Bletry O, Grunfeld JP, Piette JC, Lesavre P: The expanding spectrum of renal diseases associated with antiphospholipid syndrome. Am J Kidney Dis. 2003, 41: 1205-1211.

    PubMed  Google Scholar 

  125. Javaid MM, Denley H, Tagboto S: Fibrillary glomerulonephritis with small fibrils in a patient with the antiphospholipid antibody syndrome successfully treated with immunosuppressive therapy. BMC Nephrol. 2007, 8: 7.

    PubMed  PubMed Central  Google Scholar 

  126. Wagenknecht DR, Fastenau DR, Torry RJ, Becker DG, LeFor WM, Carter CB, Haag BW, McIntyre JA: Risk of early renal allograft failure is increased for patients with antiphospholipid antibodies. Transpl Int. 2000, 13: S78-S81.

    PubMed  Google Scholar 

  127. Wagenknecht DR, Becker DG, LeFor WM, McIntyre JA: Antiphospholipid antibodies are a risk factor for early renal allograft failure. Transplantation. 1999, 68: 241-246.

    CAS  PubMed  Google Scholar 

  128. Dayal NA, Isenberg DA: Endstage renal failure in primary antiphospholipid syndrome–case report and review of literature. Rheumatology (Oxford). 2003, 42: 1128-1129.

    CAS  Google Scholar 

  129. Korkmaz C, Kabukcuoglu S, Isiksoy S, Yalcin AU: Renal involvement in primary antiphospholipid syndrome and its response to immunosuppressive therapy. Lupus. 2003, 12: 760-765.

    CAS  PubMed  Google Scholar 

  130. Erkan D, Lockshin MD: New approaches for managing antiphospholipid syndrome. Nat Clin Pract Rheumatol. 2009, 5: 160-170.

    PubMed  Google Scholar 

  131. Scott DL, Wolfe F, Huizinga TW: Rheumatoid arthritis. Lancet. 2010, 376: 1094-1108.

    PubMed  Google Scholar 

  132. Kuroda T, Tanabe N, Kobayashi D, Wada Y, Murakami S, Nakano M, Narita I: Significant association between renal function and area of amyloid deposition in kidney biopsy specimens in reactive amyloidosis associated with rheumatoid arthritis. Rheumatol Int. 2012, 32: 3155-3162.

    CAS  PubMed  Google Scholar 

  133. Giordano A, Cencioni L, Salvo DP, Berrettini M: Membranous nephropathy secondary to rheumatoid arthritis occurring during anti-TNFalpha therapy and responsive to second-line treatment with rituximab. G Ital Nefrol. 2011, 28: 214-218.

    PubMed  Google Scholar 

  134. Maruotti N, Corrado A, Gaudio A, Cantatore FP: Membranous nephropathy in rheumatoid arthritis: a case report. Clin Exp Rheumatol. 2009, 27: 840-842.

    CAS  PubMed  Google Scholar 

  135. Honkanen E, Tornroth T, Pettersson E, Skrifvars B: Membranous glomerulonephritis in rheumatoid arthritis not related to gold or D-penicillamine therapy: a report of four cases and review of the literature. Clin Nephrol. 1987, 27: 87-93.

    CAS  PubMed  Google Scholar 

  136. Myllykangas-Luosujarvi R, Aho K, Kautiainen H, Hakala M: Amyloidosis in a nationwide series of 1666 subjects with rheumatoid arthritis who died during 1989 in Finland. Rheumatology (Oxford). 1999, 38: 499-503.

    CAS  Google Scholar 

  137. Uda H, Yokota A, Kobayashi K, Miyake T, Fushimi H, Maeda A, Saiki O: Two distinct clinical courses of renal involvement in rheumatoid patients with AA amyloidosis. J Rheumatol. 2006, 33: 1482-1487.

    PubMed  Google Scholar 

  138. Korpela M, Mustonen J, Teppo AM, Helin H, Pasternack A: Mesangial glomerulonephritis as an extra-articular manifestation of rheumatoid arthritis. Br J Rheumatol. 1997, 36: 1189-1195.

    CAS  PubMed  Google Scholar 

  139. Gedalia A, Mendez EA, Craver R, Vehaskari M, Espinoza LR: Renal involvement in juvenile rheumatoid arthritis: report of two cases. Clin Rheumatol. 2001, 20: 153-156.

    CAS  PubMed  Google Scholar 

  140. Yun YS, Song HC, Lee K, Choi EJ, Kim YS, Min JK, Kim YK: Fibrillary glomerulonephritis in rheumatoid arthritis. Nephrology (Carlton). 2010, 15: 266-267.

    Google Scholar 

  141. Kaneko K, Nanki T, Hosoya T, Mizoguchi F, Miyasaka N: Etanercept-induced necrotizing crescentic glomerulonephritis in two patients with rheumatoid arthritis. Mod Rheumatol. 2010, 20: 632-636.

    PubMed  Google Scholar 

  142. Stokes MB, Foster K, Markowitz GS, Ebrahimi F, Hines W, Kaufman D, Moore B, Wolde D, D'Agati VD: Development of glomerulonephritis during anti-TNF-alpha therapy for rheumatoid arthritis. Nephrol Dial Transplant. 2005, 20: 1400-1406.

    CAS  PubMed  Google Scholar 

  143. Schiff MH, Whelton A: Renal toxicity associated with disease-modifying antirheumatic drugs used for the treatment of rheumatoid arthritis. Semin Arthritis Rheum. 2000, 30: 196-208.

    CAS  PubMed  Google Scholar 

  144. Nakamura T, Higashi S, Tomoda K, Tsukano M, Shono M: Etanercept can induce resolution of renal deterioration in patients with amyloid A amyloidosis secondary to rheumatoid arthritis. Clin Rheumatol. 2010, 29: 1395-1401.

    PubMed  Google Scholar 

  145. Tyndall A: Successes and failures of stem cell transplantation in autoimmune diseases. Hematology Am Soc Hematol Educ Program. 2011, 2011: 280-284.

    PubMed  Google Scholar 

  146. Burt RK, Shah SJ, Dill K, Grant T, Gheorghiade M, Schroeder J, Craig R, Hirano I, Marshall K, Ruderman E, Jovanovic B, Milanetti F, Jain S, Boyce K, Morgan A, Carr J, Barr W: Autologous non-myeloablative haemopoietic stem-cell transplantation compared with pulse cyclophosphamide once per month for systemic sclerosis (ASSIST): an open-label, randomised phase 2 trial. Lancet. 2011, 378: 498-506.

    CAS  PubMed  Google Scholar 

  147. Dhaun N, MacIntyre IM, Bellamy CO, Kluth DC: Endothelin receptor antagonism and renin inhibition as treatment options for scleroderma kidney. Am J Kidney Dis. 2009, 54: 726-731.

    CAS  PubMed  Google Scholar 

  148. Sfikakis PP, Papamichael C, Stamatelopoulos KS, Tousoulis D, Fragiadaki KG, Katsichti P, Stefanadis C, Mavrikakis M: Improvement of vascular endothelial function using the oral endothelin receptor antagonist bosentan in patients with systemic sclerosis. Arthritis Rheum. 2007, 56: 1985-1993.

    CAS  PubMed  Google Scholar 

  149. Vadacca M, Margiotta D, Sambataro D, Buzzulini F, Lo Vullo M, Rigon A, Afeltra A: BAFF/APRIL pathway in Sjogren syndrome and systemic lupus erythematosus: relationship with chronic inflammation and disease activity. Reumatismo. 2010, 62: 259-265.

    CAS  PubMed  Google Scholar 

  150. Weidenbusch M, Rommele C, Schrottle A, Anders HJ: Beyond the LUNAR trial. Efficacy of rituximab in refractory lupus nephritis. Nephrol Dial Transplant. 2013, 28: 106-111.

    CAS  PubMed  Google Scholar 

  151. Merrill J, Buyon J, Furie R, Latinis K, Gordon C, Hsieh HJ, Brunetta P: Assessment of flares in lupus patients enrolled in a phase II/III study of rituximab (EXPLORER). Lupus. 2011, 20: 709-716.

    CAS  PubMed  Google Scholar 

  152. Vital EM, Dass S, Buch MH, Henshaw K, Pease CT, Martin MF, Ponchel F, Rawstron AC, Emery P: B cell biomarkers of rituximab responses in systemic lupus erythematosus. Arthritis Rheum. 2011, 63: 3038-3047.

    CAS  PubMed  Google Scholar 

  153. Baigent C, Landray MJ, Reith C, Emberson J, Wheeler DC, Tomson C, Wanner C, Krane V, Cass A, Craig J, Neal B, Jiang L, Hooi LS, Levin A, Agodoa L, Gaziano M, Kasiske B, Walker R, Massy ZA, Feldt-Rasmussen B, Krairittichai U, Ophascharoensuk V, Fellstrom B, Holdaas H, Tesar V, Wiecek A, Grobbee D, de Zeeuw D, Gronhagen-Riska C, Dasgupta T: The effects of lowering LDL cholesterol with simvastatin plus ezetimibe in patients with chronic kidney disease (study of heart and renal protection): a randomised placebo-controlled trial. Lancet. 2011, 377: 2181-2192.

    CAS  PubMed  PubMed Central  Google Scholar 

  154. Malaviya AP, Hall FC: Targeting CVD risk in chronic connective tissue disease. Practitioner. 2012, 256: 21-26.

    PubMed  Google Scholar 

  155. Marengo M, Waimann C, de Achaval S, Zhang H, Garcia Gonzalez A, Richardson MN, Reveille JD, Suarez-Almazor ME: Measuring therapeutic adherence in systemic lupus erythematosus with electronic monitoring. Lupus. 2012, 21: 1158-1165.

    CAS  PubMed  Google Scholar 

  156. Klareskog L, Padyukov L, Alfredsson L: Smoking as a trigger for inflammatory rheumatic diseases. Curr Opin Rheumatol. 2007, 19: 49-54.

    CAS  PubMed  Google Scholar 

Pre-publication history

Download references

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Gert Mayer.

Additional information

Competing interests

The authors declare that they have no competing interests.

Authors’ contributions

AK performed the literature search and wrote the manuscript. GM critically reviewed the manuscript. Both authors approved the final version of the manuscript.

Rights and permissions

This article is published under license to BioMed Central Ltd. This is an Open Access article distributed under the terms of the Creative Commons Attribution License (http://creativecommons.org/licenses/by/2.0), which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited.

Reprints and permissions

About this article

Cite this article

Kronbichler, A., Mayer, G. Renal involvement in autoimmune connective tissue diseases. BMC Med 11, 95 (2013). https://doi.org/10.1186/1741-7015-11-95

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/1741-7015-11-95

Keywords