You are on page 1of 10

Seminar

Glomerulonephritis
S J Chadban, R C Atkins Lancet 2005; 365: 1797–806
Department of Nephrology,
The term glomerulonephritis encompasses a range of immune-mediated disorders that cause inflammation within Monash Medical Centre,
Clayton, Victoria, Australia
the glomerulus and other compartments of the kidney. Studies with animal models have shown the crucial
(S J Chadban FRACP,
interaction between bone-marrow-derived inflammatory cells and cells intrinsic to the kidney that is both Prof R C Atkins FRACP); and
fundamental and unique to the pathogenesis of glomerulonephritis. The mechanisms of interaction between these Renal Medicine and
cells and the mediators of their coordinated response to inflammation are being elucidated. Despite these Transplantation, Royal Prince
Alfred Hospital and University
pathophysiological advances, treatments for glomerulonephritis remain non-specific, hazardous, and only partly
of Sydney, Camperdown, NSW,
successful. Glomerulonephritis therefore remains a common cause of end-stage kidney failure worldwide. Australia (S J Chadban)
Molecule-specific approaches offer hope for more effective and safer treatments in the future. Correspondence to:
Dr S J Chadban, Transplantation,
Glomerulonephritis is a subject of confusion among the major contributors, glomerulonephritis is likely to be Level E9, Royal Prince Alfred
Hospital, Camperdown,
health-care workers. Much of the confusion stems from the cause in a substantial proportion.
NSW 2050, Australia
the nomenclature, which attempts to encapsulate the The epidemiology of more serious and clinically Steve.Chadban@cs.nsw.gov.au
aetiology, pathology, and clinical presentation. The apparent episodes of glomerulonephritis is better defined.
aetiological description refers to primary (aetiology Accurate classification requires a histological diagnosis;
unknown, generally thought to be a manifestation of consequently, the reported prevalence might vary
autoimmunity) or secondary (associated with one of according to local indications for renal biopsy. A study of
several autoimmune, infectious, malignant, or all renal biopsies done in the Australian state of Victoria
metabolic diseases) forms of glomerulonephritis. The during 1995 and 1997 found that 21·5 people per 100 000
pathological description addresses glomerular population per year underwent renal biopsy, yielding a
involvement, cell involvement, and changes in non- yearly incidence of biopsy-proven glomerulonephritis of
cellular components of the glomerulus (panel 1; figure). 12·3 per 100 000. The most commonly diagnosed types of
Classification according to the clinical presentation is glomerulonephritis in adults were IgA nephropathy, focal
the simplest and most effective tool for the clinician and segmental glomerulosclerosis, and vasculitis; those
(panel 2). Although each clinical presentation can be most commonly diagnosed in children were minimal-
associated with several distinct types of glomerulo- change disease, focal and segmental glomerulosclerosis,
nephritis defined by aetiology and pathology, the lupus nephritis, and IgA nephropathy.4 A population-
approach to diagnosis and management is best directed based study spanning 1976–2002, which included 898
by the clinical presentation. Rapidly progressive biopsy-proven cases of glomerulonephritis among
glomerulonephritis is a medical emergency, demanding 412 735 inhabitants of a region of western France,
urgent histological diagnosis and treatment to prevent documented similar incidence, prevalence, and pattern of
renal failure.1 Nephritic and nephrotic presentations disease and an increase in the incidence of crescentic
generally indicate urgent renal biopsy to guide glomerulonephritis over time.5
management and provide prognostic information. Most patients with glomerulonephritis incur chronic
Chronic glomerulonephritis is also an indication for kidney disease, with the attendant risks of premature
diagnostic biopsy and for the initiation of therapy to slow cardiovascular disease6 and progressive kidney failure.
progression of renal impairment and limit the The burden of glomerulonephritis is most clinically
consequences of renal failure. Asymptomatic urinary apparent in its contribution to end-stage kidney failure,
abnormalities require a less urgent approach— which necessitates dialysis or transplantation.
diagnostic tests including renal biopsy in some cases, or Glomerulonephritis is recognised as the second
observation over time in others. commonest cause of end-stage renal failure worldwide. In
Australia, glomerulonephritis is the most common cause,
Epidemiology
Many cases of glomerulonephritis result in mild, Search strategy and selection criteria
asymptomatic illness that is not recognised by the
We searched MEDLINE for papers published in English
patient, is not brought to medical attention, and remains
between 1998 and 2004 with the search term
undiagnosed. The incidence and prevalence of such mild
“glomerulonephritis” and selected those relevant to the
episodes of glomerulonephritis are unknown but could
theme of the Seminar. We also searched reference lists of
be substantial. Population-based screening studies have
papers of interest and accessed key publications over the past
shown that evidence of kidney damage—proteinuria,
50 years. Since there is much research on this topic, the
haematuria, low calculated glomerular filtration rate, or a
reference list reflects the results of these searches, with
combination of these features—is present in 16% of
selection at our discretion rather than a systematic review.
adults in Australia2 and a similar proportion in the USA.3
Although diabetic and hypertensive nephropathies are

www.thelancet.com Vol 365 May 21, 2005 1797


Seminar

occurring types of biopsy-proven glomerulonephritis


Panel 1: Pathological classification causing end-stage renal failure were IgA nephropathy
Glomerular involvement (27%), focal sclerosing (14%), rapidly progressive/
All glomeruli (diffuse) or only some (focal) crescentic glomerulonephritis (10%, including vasculitis
Extent of disease within involved glomeruli: patchy and antiglomerular-basement-membrane disease),
(segmental) or general membranous (5%), and lupus nephritis (4%).7
Glomerulonephritis is more common in the male than
Cell involvement in the female population.4 Lupus nephritis is the major
Increases in cell number (proliferative) exception; the frequency is two or more times higher
Neutrophil accumulation (exudative) among the female than among the male population.4
Cell damage However, since the female to male ratio of systemic
Cell necrosis visible by light microscopy (necrotising) lupus erythematosus is roughly ten, yet that of nephritis
Ultrastructural damage visible only by electronmicroscopy is only two to three, male patients with lupus seem more
(foot-process effacement, membrane thinning) likely to develop nephritis than female patients.
Defined populations are clearly at increased risk of
Changes in non-cellular glomerular components glomerulonephritis. Children in less developed countries,
Matrix accumulation (hyalinosis) or immune deposits and those in impoverished sections of more developed
Site of deposition (mesangial, subendothelial, subepithelial) countries, such as Australian Aboriginals,9 are at
increased risk of infection-associated glomerulonephritis
after streptococcal skin or throat infections. This disease is
leading to 27% of cases of end-stage renal failure in 2001;7 now rare in more developed countries.4,5,10 Individuals with
in the USA, however, glomerulonephritis ranks third chronic infections with viruses, particularly hepatitis B or
behind diabetes and hypertension.8 The propensity for C virus and HIV, are at increased risk of several types of
each type of glomerulonephritis to cause end-stage renal immune-mediated glomerulonephritis.11,12 Predisposition
failure varies, but the types most frequently identified on to glomerulonephritis might also be hereditary, as is the
biopsy are also those most likely to result in kidney case with Alport’s syndrome, in which mutations in the
failure.4 In Australia during 2001, the most frequently COL4A5 (X-linked), COL4A3, or COL4A4 (both
autosomal recessive) genes result in defective synthesis
of collagen type IV with consequent dysfunction and
Mesangial-cell disease
IgA nephropathy, IgM nephropathy, inflammation of the glomerular basement membrane.13,14
mesangioproliferative glomerulonephritis,
class II lupus nephritis,
diabetic nephropathy Panel 2: Classification according to clinical presentation

Mesangial cell Asymptomatic urinary abnormalities


Basement membrane Subnephrotic-range proteinuria, and/or microscopic
Endothelial cell
Epithelial cell
haematuria, not accompanied by renal impairment, oedema,
or hypertension
Epithelial-cell injury Lumen Endothelial-cell injury Nephritic syndrome
Membranous nephropathy, Infection-associated glomerulonephritis,
minimal-change disease, mesangiocapillary glomerulonephritis, Recent onset of haematuria and proteinuria, renal
focal and segmental class III and IV lupus nephritis, impairment, and salt and water retention, causing
glomerulosclerosis, antiglomerular-basement-membrane disease,
vasculitis and cryoglobulinaemia,
hypertension
class V lupus nephritis,
diabetic nephropathy haemolytic uraemic syndrome
Rapidly progressive glomerulonephritis
Progression to renal failure over days to weeks, in most cases
In health, solutes are filtered into the urinary space. The presence of abnormal
in the context of a nephritic presentation, typically associated
amounts of protein or cells suggests glomerular pathology with the pathological finding of extensive glomerular
crescent formation on renal biopsy
Figure: Cellular location of injury during glomerulonephritis Nephrotic syndrome
Mesangial cells are directly exposed to the circulation. Deposition of immune complexes within these cells is Nephrotic-range proteinuria (more than 3·5 g per 1·73 m2 in
typically seen in disorders such as IgA nephropathy; it results in proliferation and expansion of the cells leading to
haematuria, proteinuria, and renal impairment. Epithelial cells, in conjunction with basement membrane, allow
24 h), hypoalbuminaemia, hyperlipidaemia, and oedema, in
filtration of plasma solutes but retard passage of cells and plasma proteins. Disease related to these cells is typified many cases complicated by predisposition to venous
by the presence of subepithelial deposits and flattening of the foot processes that engage the basement thrombosis and bacterial infection
membrane, resulting in disruption of the filtration barrier and proteinuria. Endothelial-cell disease can result from
immune-complex deposition (as occurs in mesangiocapillary glomerulonephritis), antibody attachment to the Chronic glomerulonephritis
basement membrane (Goodpasture’s disease), or trauma and activation of coagulation (haemolytic uraemic Persistent proteinuria with or without haematuria and slowly
syndrome). Endothelial-cell proliferation and necrosis are accompanied by leucocyte accumulation, and rupture of
progressive impairment of renal function
the basement membrane, crescent formation, and disruption of glomerular architecture can develop. A nephritic
or rapidly progressive presentation ensues.

1798 www.thelancet.com Vol 365 May 21, 2005


Seminar

Pathogenesis mediated cytotoxicity,33 by activating infiltrating and


The pathogenesis of glomerulonephritis is complex and intrinsic kidney cells via Fc receptors,16 or by fixing and
remains incompletely understood. Complexity arises from activating complement.34
several factors. First, many types of insult can initiate Goodpasture’s disease is initiated by deposition of IgG
glomerulonephritis; they include endogenous processes, along the glomerular basement membrane (hence anti-
such as autoimmunity, cancer, and ultrastructural glomerular-basement-membrane disease). Antibody
abnormalities within the kidney, and exogenous factors, binding promotes a type II immune response, whereby
including infectious organisms and drugs. Second, there complement and coagulation are activated. An adaptive
is individual variability in the susceptibility to T-cell response ensues, which causes kidney damage
glomerulonephritis, which is likely to have a genetic basis; and is sufficient to perpetuate the disease process.1 The
for example, only a minority of individuals infected with antibodies causing the disease are specific for the
hepatitis C virus develop glomerulonephritis. Third, there non-collagenous domain of type IV collagen present
are complex interactions between soluble factors within the glomerular basement membrane.35 The
(including antibodies, complement, chemokines, presence of this antigen within the alveolar basement
cytokines, and growth factors) and cells (both leucocytes membrane is the reason why many patients with this
and intrinsic kidney cells) that mediate glomerular disease suffer pulmonary haemorrhage in addition to
inflammation. A final cause of complexity is the glomerulonephritis.
progressive nature of kidney disease; it is due to the Antibody can also be deposited from circulation as a
contribution of non-specific factors such as hypertension component of immune complexes or cryoglobulins, thus
and proteinuria that can promote continuing kidney triggering a type III immune response within the
damage, despite resolution of the initial insult. glomerulus. Circulating immune-complex formation
Small-animal models of glomerulonephritis closely and deposition within the glomerulus is seen particularly
replicating features of human glomerulonephritis have during subacute bacterial endocarditis, lupus nephritis,
provided insights into the pathogenesis. The and hepatitis-C-related cryoglobulinaemia.11
reproducibility and uniformity of such animal models Accumulation of isotype-specific, though not antigen-
has permitted a step-by-step dissection of many of the specific, antibody within mesangial cells is the
mechanisms involved. We discuss several key pathognomonic feature of IgA nephropathy. The cause
mechanisms thus identified (table 1)15–32 and relate these of the IgA deposition remains unclear, but it is probably
to their counterparts in human disease. a consequence of either abnormal IgA structure or
abnormal Fc-receptor function in susceptible
Antibody individuals.36 The interaction of antibody with cells, via
Antibody deposition within the kidney seems to be the cell-surface receptors for the constant domain of
initiating event in several forms of naturally occurring immunoglobulin (Fc receptors), is an essential element
and experimentally induced glomerulonephritis. in the NZB/W model of lupus nephritis; mice
Deposited antibody could lead to glomerular genetically deficient in Fc receptors are resistant to the
inflammation by promoting antibody-dependent cell- development of glomerulonephritis.16

Target process Target cells Strategy Effect


Antibody deposition B-cells/plasma cells Blockade of B-lymphocyte stimulator Reduces autoantibodies and kidney damage in lupus model15
Antibody receptors Macrophages, mesangial cells Fc-receptor-deficient mice Inhibits inflammation in response to deposited IgG in lupus model16
Complement activation NA (soluble protein) Complement depletion; anti-C5, soluble receptor Inhibits proteinuria;17 inhibits progression of proteinuria and kidney dysfunction18,19
Coagulation Endothelium Hirudin (thrombin antagonist) Inhibits crescentic nephritis20
Chemotaxis Infiltrating leucocytes, intrinsic kidney Antagonist: met-RANTES Attenuation of proteinuria, infiltrate;21 resolution of proteinuria, infiltrate;22
cells Antibody: MCP1 tubulointerstitial protection23
MCP1-knockout mice
Adhesion Endothelium Interleukin-1-receptor antagonist inhibits Attenuation of leucocyte accumulation and kidney damage in anti-GBM disease24
kidney expression of ICAM1
Proliferation Macrophages, mesangial cells, tubular Antibody to MCSF receptor Inhibits macrophage proliferation;25 increased glomerular hypercellularity and
cells p27Kip-1 knockout mice tubular damage26
Signal transduction Mesangial cells, neutrophils STI-571 PDGF receptor phophorylation inhibitor Prevention of hypercellularity in mesangioproliferative model;27 reduced
Inhibition of MAP kinase neutrophil-dependent proteinuria28
Gene transcription Intrinsic cells, leucocytes NF-B decoy oligodinucleotide Inhibited TNF-induced glomerular inflammation29
Cell depletion T lymphocytes Depleting anti-CD4 Inhibition of proteinuria in a model of membranous nephropathy30,31
Depleting anti-CD8
Effector molecules Macrophages, mesangial cells Nitric oxide synthetase antagonist, L-NAME Diminished monocyte infiltration and glomerular injury32

NA=not applicable; MCP1=macrophage chemotactic protein 1; ICAM1=intercellular adhesion molecule 1; GBM=glomerular basement membrane; MCSF=macrophage-colony-stimulating factor; PDGF=platelet-derived growth
factor; MAP=mitogen-activated protein; NF-B=nuclear factor B; TNF=tumour necrosis factor; L-NAME=NG-nitro-L-arginine methyl ester.

Table 1: Experimental therapies in glomerulonephritis

www.thelancet.com Vol 365 May 21, 2005 1799


Seminar

Antineutrophil cytoplasmic antibodies (ANCA) are evidence of a role for complement in the initiation and
detectable in the serum of most patients with necrotising progression of glomerular disease. Blockade of C5b-9
small-vessel vasculitis affecting the kidney, most of formation in the Heymann nephritis model of human
whom are classified as having Wegener’s granulomatosis membranous nephropathy effectively prevents dis-
or microscopic polyangiitis. ANCA can be directed ease,17,42,43 confirming a role for complement in disease
against proteinase 3, which gives a cytoplasmic initiation. The finding that complement might drive
distribution on indirect immunofluorescence (cANCA), progressive kidney damage in glomerular diseases
or myeloperoxidase, which results in a perinuclear characterised by proteinuria is potentially more
distribution (pANCA). ANCA are useful for diagnostic important. Hsu and Couser44 reviewed evidence in
purposes37 and seem likely to have a pathogenetic role in support of the hypothesis that during proteinuric disease,
vasculitis. In vitro and in vivo, ANCA upregulate complement components enter the tubular lumen via
endothelial-cell expression of adhesion molecules and filtration through damaged glomeruli or local synthesis45
thereby increase interactions with primed monocytes and then become activated, culminating in the generation
neutrophils.38,39 and deposition of C5b-9 along the luminal membrane of
tubular cells, causing chronic tubulointerstitial inflam-
Complement mation and progressive kidney failure. Studies in a non-
The complement cascade is a central component of the immunologically initiated model of glomerular disease
innate immune system. Activation of the cascade can (aminonucleoside nephrosis) in rats unable to assemble
occur in the classic way, after exposure to bacteria, or via the C5b-9 complex provide support for this theory.46
the alternative pathway in response to bacterial Similar results have been achieved by the administration
polysaccharides, damaged cells, or aggregated IgA. In of the soluble, recombinant inhibitor of C5b-9 formation
either case, a self-amplifying signalling cascade is initiated, soluble complement receptor 118 or complement-
which generates chemotactic (C5a) and proinflammatory blocking antibodies such as anti-C5,19 which shows the
(C3a) molecules and ultimately a cytolytic protein complex potential of anticomplement strategies for the
known as C5b-9, or the membrane attack complex. management of human glomerulonephritis.
Activation of the complement cascade is tightly regulated
by the short half-life of activated complement proteins and Coagulation
by a series of endogenous regulatory proteins.34 Intraglomerular activation of the coagulation cascade,
Components of the complement cascade liberated triggered primarily by tissue factor and resulting in
during the process of activation can be detected in tissue, thrombin formation then fibrin deposition, is evident in
serum, or urine and are evidence of complement activation severe forms of human and experimental glomerulo-
and the pathway taken to activation—classic or alternative. nephritis. Local upregulation of tissue factor, augmented
Evidence of complement deposition within glomeruli is a by a transient reduction in local production of its
key feature of many forms of glomerulonephritis, and a endogenous regulator tissue-factor pathway inhibitor
pathogenetic role of complement is implied by several (TFPI), trigger activation of the extrinsic coagulation
observations in human disease. First, deposition of pathway in experimental crescentic glomerulonephritis.
complement products can be readily shown by TFPI antibody exacerbated disease in this model as
immunohistochemistry or immunofluorescence in shown by fibrin deposition, crescent formation,
specific types of glomerulonephritis (eg, lupus nephritis). inflammatory-cell infiltration, and kidney damage; the
Second, the concentration of complement components in reverse effects were achieved by administration
serum reflects disease activity; for example, C3 of recombinant TFPI.47 Intraglomerular coagulation
concentration is commonly low during active lupus clearly has proinflammatory actions in addition to
nephritis. Third, there is urinary excretion of complement haemodynamic consequences. Studies in a mouse
activation products in progressive, proteinuric nephro- model of crescentic glomerulonephritis have shown the
pathies such as membranous glomerulonephritis.40,41 importance of the interaction between thrombin and
Fourth, genetic deficiency of specific complement protease-activated receptor 1 in promoting cellular
components has been associated with glomerulonephritis, infiltration and inflammation, independent of fibrin
such as C2 deficiency in some cases of lupus nephritis. deposition and clot formation.20
Finally, mechanisms causing aberrant activation of
complement have been identified in some glomerular Leucocytes
diseases, such as the C3-activating factor “C3-neph” in A constant feature of glomerulonephritis is the presence
poststreptococcal glomerulonephritis.34 of inflammatory leucocytes within both the glomerular
Indirect evidence obtained from human studies has and the interstitial compartments of the kidney.
been supported by findings in animal models. The Macrophages and T lymphocytes are the dominant cell
administration of complement inhibitors or the use of types present, and the intensity of cellular accumulation,
mice genetically deficient in complement components or particularly within the interstitium rather than the
complement-regulatory molecules has provided strong glomerulus, has repeatedly been found to correlate with

1800 www.thelancet.com Vol 365 May 21, 2005


Seminar

the clinical and pathological severity of disease in animal and protecting rats from the development of anti-
models and in human beings.48,49 glomerular-basement-membrane disease.24
T cells seem to be crucial to the development of most Proliferation of cells, particularly macrophages, is an
forms of glomerulonephritis. T-cell depletion studies in important mechanism by which the inflammatory
Heymann nephritis, a rat model of membranous response within the kidney is amplified. Proliferation has
nephropathy, have shown an absolute requirement for been clearly identified by incorporation of
CD4-positive30 and CD8-positive31 T cells. In some models, bromodeoxyuridine, injected 2 h before the animals were
T cells are sufficient to produce disease, as shown by the killed, by macrophages within inflamed glomeruli, but
transmission of anti-glomerular-basement-membrane not by monocytes in circulation; thus, macrophages
disease to healthy chickens by transfer of T cells isolated within the kidney passed through S-phase of the cell
from chickens with established disease.50 T cells apparently cycle.59 The demonstration of mitotic figures and
initiate, direct, and amplify the cognate immune response expression of proliferating-cell nuclear antigen by
during many types of glomerulonephritis.51 However, macrophages within inflamed glomeruli in human
specific T-cell subsets are also likely to downregulate this biopsy studies confirms that proliferation also contributes
inflammatory response (T-regulatory cells).52 to inflammation in human glomerulonephritis.60
Most of the accumulated inflammatory cells in many Macrophage proliferation within the inflamed kidney is
forms of human and experimental glomerulonephritis are growth-factor dependent, as shown by upregulation of
macrophages. In a model of glomerulonephritis that monocyte-colony-stimulating factor in both experimental
allows leucocyte depletion followed by reconstitution with and human glomerulonephritis61 and by the inhibition of
macrophages alone, macrophages were able to cause renal proliferation and disease with blockade of macrophage-
injury, as shown by proteinuria and mesangial-cell colony-stimulating-factor receptor.25 Less cell-specific
proliferation.53 The mechanisms by which macrophages blockade of proliferation has been achieved through
mediate renal injury remain to be confirmed, but are likely inhibition of cyclin-dependent kinase, also resulting in
to include: promotion of inflammation via release of pro- attenuation of experimental glomerulonephritis.26
inflammatory cytokines such as tumour necrosis factor ; Within the kidney, T cells and macrophages undergo
recruitment of macrophages and other leucocytes via activation, which is triggered by interactions between
release of chemotactic molecules such as macrophage leucocytes and intrinsic renal cells, by interactions between
chemotactic protein 1; cell proliferation by release of cells and matrix, and by cytokines and other soluble factors
growth factors such as macrophage-colony-stimulating present within the local inflammatory milieu. Production
factor; cell death via the release of reactive oxygen species of proinflammatory cytokines, including interleukin 1,
including nitric oxide; and control of fibrosis and repair via tumour necrosis factor , and interferon , is upregulated
release of metalloproteinases and other enzymes.54 in experimental and human disease, and blocking studies
In the setting of glomerulonephritis, cells accumulate have shown protection in several animal models.62
within the kidney as a result of recruitment from the Inflammation within the glomerulus commonly
circulation and proliferation within the kidney. causes functional and structural damage to the kidney.
Recruitment of leucocytes from the circulation is The mechanisms of damage include: direct cell damage
directed by chemokines generated within the inflamed by macrophages, through production of reactive oxygen
kidney.55 Protection against leucocyte accumulation species;63 induction of apoptosis, particularly by CD8-
within the kidney and renal injury has been achieved positive T cells;64 antibody-dependent cell-mediated
through blockade of key macrophage chemokines, such cytotoxicity, as immunoglobulin attached to kidney cells
as macrophage chemotactic protein 122 or its receptor is engaged by Fc receptors expressed by macrophages or
CCR2,21 or T-cell chemokines, such as RANTES.56 other phagocytes;16,33 liberation of enzymes and
Animals genetically deficient in chemokines are metalloproteases causing disruption of basement
resistant to disease induction,23 although results have membrane and other stromal structures leading to
not been uniform, with an unexpected accentuation architectural disruption;65 myofibroblast accumulation
of experimental anti-glomerular-basement-membrane within the tubulointerstitium66 and glomerulus67 and
disease seen in mice deficient in CCR1, a receptor for production of excess matrix components, driven by
RANTES and macrophage inflammatory protein 1.57 growth factors (particularly transforming growth
Chemokines identified as important in the murine factor ), leading to scarring and fibrosis.68
models are clearly active in human disease.58 Once
leucocytes have been attracted into the renal micro- Intrinsic kidney cells
circulation, engagement between adhesion molecules The glomerulus consists of a network of specialised
and their ligands appears necessary for migration of the capillaries, supported by a stroma of mesangial cells,
cells into kidney tissues, as shown by the effectiveness epithelial cells, and matrix, encased within Bowman’s
of blocking the cytokine interleukin 1; this blockade capsule. Each type of these resident glomerular
inhibited upregulation of intercellular adhesion cells has been implicated in the pathogenesis of
molecule 1, thereby preventing leucocyte infiltration glomerulonephritis (figure).

www.thelancet.com Vol 365 May 21, 2005 1801


Seminar

Glomerular epithelial cells or podocytes have foot turn can cause further kidney damage.77 Conversely,
processes that act in concert with the endothelial-cell commonly used treatments for hypertension and
basement membrane to form a filtration barrier or “slit proteinuria, such as blockade of the renin-angiotensin
diaphragm”, which permits the passage of water and system, directly retard the continuing inflammatory and
electrolytes into the glomerular filtrate, while retarding the fibrotic response within the kidney, in addition to their
passage of cells and proteins from the blood. The beneficial effects on blood pressure and proteinuria.78
molecular composition of the slit diaphragm has been
largely unravelled.69 Mutation of the nephrin gene is Management
present in patients with congenital nephrotic syndrome of Glomerulonephritis is a major contributor to the
the Finnish type, and other molecular abnormalities of the escalating health burden associated with chronic kidney
filtration barrier have been associated with other types of disease. Thus, broader implementation of interventions
glomerulonephritis that are characterised by nephrotic shown to be effective in slowing the progression of
syndrome. glomerulonephritis is very important from an
Necrosis, apoptosis, proliferation, and activation of economic perspective.79 From the patient’s perspective,
glomerular endothelial cells are variably seen in types of interventions likely to achieve even small delays in the
glomerulonephritis causing the nephritic syndrome, requirement for dialysis or transplantation are desirable.79
including mesangiocapillary glomerulonephritis, Therapeutic options for human glomerulonephritis
crescentic glomerulonephritis, and haemolytic uraemic applicable to all cases include symptomatic treatment and
syndrome. Antibody deposition followed by complement strategies to delay progression; immunosuppression is
activation has been shown experimentally to induce appropriate for selected cases; and renal replacement
apoptosis.70 Protection from experimental injury could be therapy with dialysis or transplantation is needed for a
provided by vascular endothelial growth factor.71 The sizeable minority of patients. Molecular therapies
mechanisms involved in human pathology remain to be designed to target key mediators of damage hold great
elucidated. promise and are effective in several experimental settings
There is evidence from both human studies and animal but are yet to achieve translation into the clinic (table 1).
models that mesangial-cell proliferation and expansion of Strategies to delay progression provide an important
mesangial areas within glomeruli precede glomerulo- adjunct to therapy for all patients with glomerulo-
sclerosis in many types of glomerulonephritis, particularly nephritis. Regular clinical follow-up,80 blood-pressure
IgA nephropathy, and in other types of glomerular injury, control,77 and the use of an inhibitor of angiotensin-
such as diabetic nephropathy. Activation and proliferation converting enzyme in patients with proteinuria exceeding
of mesangial cells could be triggered by soluble factors 1 g daily,77,81,82 are of proven benefit. Recent findings
including angiotensin, cytokines, and growth factors, such suggest that more complete blockade of the renin-
as platelet-derived growth factor, or deposition of antibody angiotensin system by use of both an angiotensin-
or immune complexes.72 Once activated, mesangial cells converting-enzyme inhibitor and an angiotensin-receptor
promote inflammation via the release of chemokines and antagonist is superior to either agent alone for prevention
cytokines and glomerulosclerosis via matrix production of progressive renal failure among patients with
and transdifferentiation into myofibroblasts.73 proteinuria.83 Dietary protein restriction can limit uraemic
The cells of the tubulointerstitial compartment tend to symptoms and slow progression in selected cases,77,84 but
be neglected, but they add to glomerular injury through the beneficial effects are slight and such dietary restriction
their release of growth factors and cytokines and by their can incur a substantial risk of malnutrition. Treatment of
contribution to fibrosis and scarring.74 In glomerulo- hyperlipidaemia has slowed progression in experimental
nephritis, tubular cells become major producers of models of glomerulonephritis; a major randomised
cytokines and growth factors75 that perpetuate renal controlled trial is under way to address this question in
damage and promote chronicity. Furthermore, trans- human beings, but available trial data are insufficient for
differentiation of tubular cells to myofibroblasts is lipid lowering to be recommended specifically for
important in renal scarring.76 The importance of the renoprotection.85
tubulointerstitium, especially after the establishment of Currently used immunosuppressive therapies for
injury, is highlighted by the finding that the degree of human glomerulonephritis are not uniformly effective
interstitial leucocyte infiltration48,49 and fibrosis are more and are frequently associated with serious side-effects.
powerful predictors of the likelihood of renal failure than Furthermore, because patients with nephropathies such
are the glomerular changes. as membranous nephritis can experience spontaneous
Glomerulonephritis, as is the case with most forms of remission, the decision to initiate immunosuppressive
renal injury, tends to produce progressive loss of kidney therapy is not always straightforward and necessitates
function despite, in many cases, apparent resolution of the consideration of the diagnosis, the patient, his or her
inciting immunological insult. Immune-mediated susceptibility to side-effects, and prognostic factors that
damage to the glomerulus and interstitium commonly might help to determine the risk of progression versus
leads to proteinuria and hypertension, both of which in remission for each case (table 2).83,86–101 Loss of glomerular

1802 www.thelancet.com Vol 365 May 21, 2005


Seminar

filtration rate, heavy proteinuria, presence of Immunosuppressive therapies are of proven benefit in
hypertension, and fibrosis within the interstitium on crescentic glomerulonephritis,1 proliferative lupus
kidney biopsy are adverse prognostic features for most nephritis,95,96 focal and segmental glomerulosclerosis,89
forms of glomerulonephritis. Their presence warrants minimal-change disease,86 membranous nephropathy,91
consideration of aggressive therapy. Advanced age and and in cases of IgA nephropathy with deteriorating renal
frailty of the patient, probability of developing diabetes or function and proteinuria.101 Table 2 presents a summary
worsening control of pre-existing diabetes, risk of of the approach to management of primary
infertility, and general susceptibility to the side-effects of glomerulonephritis with immunosuppressive agents and
immunosuppression should be considered in the choice cites available evidence. To facilitate an evidence-based
to use such therapies and which agents to use. approach to both research and practice, the Cochrane
Corticosteroids are effective in several types of Renal Group maintains a registry of trials in
glomerulonephritis owing to their ability to inhibit activity glomerulonephritis.105
of the transcription factor nuclear factor B and Exciting developments in therapy have been identified
consequently inhibit the proinflammatory effects of in experimental models of glomerulonephritis (table 1).
cytokines known to promote glomerular inflammation However, several differences between experimental
actively, including interleukin 1 and tumour necrosis glomerulonephritis and human disease should be
factor . However, other actions of corticosteroids result in considered in attempts to translate animal results into
well recognised side-effects that limit the usefulness of potential human therapies. There are biological variations
these drugs. Removal of preformed antibodies from between models and animals that can lead to inconsistent
circulation by plasmapheresis is useful in Goodpasture’s responses to therapy. For example, administration of the
disease, but not in lupus nephritis.102 The antiproliferative potentially immunosuppressive cytokine interleukin 10
agents cyclophosphamide, chlorambucil, and azathioprine was protective against crescentic glomerulonephritis in a
are effective in suppressing glomerular inflammation but mouse model106 but not in rats.107 The aetiological agent is
are non-specific in their actions and cause substantial known in most cases of experimental glomerulonephritis
morbidity and mortality. Mycophenolate mofetil preferen- but is rarely identified in human glomerulonephritis.
tially inhibits the type II isoform of inosine mono- Animals with experimental glomerulonephritis are
phosphate dehydrogenase, which is required by leucocytes homogeneous, whereas patients and human glomerulo-
for de-novo purine synthesis. However, in practice nephritis are heterogeneous. Experimental disease is
mycophenolate mofetil is not completely specific for initiated at a time determined by the investigator and runs
leucocytes and, like other antiproliferative agents, it is a predictable course, thereby permitting intervention at
associated with a broad range of side-effects including defined stages including before induction, during disease
susceptibility to infection and cancer.103 Sirolimus, an establishment, or late in the course. Reports of effective
immunosuppressant that retards T-cell passage through therapies commonly involve treatment before disease
the cell cycle, has activity in some forms of glomerulo- induction or early during the establishment phase. By
nephritis but has been associated with increased contrast, the onset of human glomerulonephritis is
proteinuria and acute renal failure in some cases.104 generally unknown, the time course is variable, and the

Type Clinical prognostic features Management strategy


Minimal-change disease 90% of children are steroid responsive; steroid resistance and relapse Oral prednisone 0·5–1·0 mg/kg daily for 6 weeks, then tapered;86
is common among adults. cyclophosphamide or ciclosporin in steroid-resistant or frequently relapsing patients.86,87
Focal and segmental glomerulosclerosis Declining GFR, nephrotic-range proteinuria, and severe nephrotic Oral prednisone 0·5–1·0 mg/kg daily for at least 6 months, may be tapered after first
syndrome are indications for a trial of immunosuppression. 3 months;88 cyclophosphamide is indicated for steroid-resistant cases, with 50% chance of
response; response reduces probability of progression to ESRD by 50%.88,89
Membranous nephropathy 30% of cases progress to ESRD within 10 years, more likely if nephrosis Cyclophosphamide (or chlorambucil) orally combined with prednisolone,90 achieves
is severe or GFR declining. 50% remission rate; steroids alone are ineffective.91
Mesangiocapillary glomerulonephritis Slowly progressive renal failure is common, predicted by the rate of loss Prednisone can be useful in children92 but not in adults;93 anti-CD20 can be effective in
of GFR and degree of proteinuria. cryoglobulinaemia-associated disease.94
Proliferative lupus nephritis (class III and IV) Biopsy diagnosis is essential because clinical features do not clearly Pulse steroids, followed by monthly intravenous cyclophosphamide and daily oral
differentiate between classes of nephritis, and class determines therapy. prednisone for 3–6 months (remission induction), followed by oral azathioprine or
mycophenolate mofetil and reduced-dose prednisone (maintenance).95-98
Rapidly progressive Lung involvement, degree of GFR reduction, and proteinuria are key Plasmapheresis, pulse methylprednisolone, followed by oral cyclophosphamide and
glomerulonephritis—anti-GBM prognostic indices. prednisone are effective.1,99
Rapidly progressive glomerulonephritis— Typically ANCA-positive; extent of GFR reduction and extrarenal Pulse methylprednisone followed by oral cyclophosphamide and prednisone.100
vasculitis involvement predict prognosis.
IgA nephropathy Heavy proteinuria, decreased GFR, and hypertension are adverse Oral prednisone can be effective in retarding progression;101 however, combined
prognostic features. inhibition of ACE and angiotensin-receptor blockade should be first-line therapy.83

GFR=glomerular filtration rate; ESRD=end-stage renal disease; GBM=glomerular basement membrane; ANCA=antineutrophil cytoplasmic antibodies; ACE=angiotensin-converting enzyme.

Table 2: Management of primary glomerulonephritis with immunosuppressive agents

www.thelancet.com Vol 365 May 21, 2005 1803


Seminar

patient typically has established disease before treatment and cell-targeted therapies. The different types of
can be given. Kidney damage tends to be progressive in glomerulonephritis are likely to need disease-specific
the longer term, despite elimination of the original source agents. Differences between experimental and human
of injury. Most animal models are short term only. Finally, glomerulonephritis complicate translation of
the effectiveness of interventions might be specific to the experimental therapies into practice, and primate studies
phase of injury. For example, transforming growth are likely to be needed in many cases. Third, stem-cell-
factor  has protective anti-inflammatory effects within based therapies should be explored; these could include
the glomerulus during the initiation phase of experi- genetic modification to produce protective molecules
mental glomerulonephritis108 but adverse profibrotic within the kidney or to promote regeneration of damaged
effects once disease is established.68 Since human intrinsic kidney cells. The complexity of the kidney will
glomerulonephritis is generally established by the time it make this approach difficult. Finally, there must be
is diagnosed, this growth factor is unlikely to be useful in continuing evidence-based assessment of immune and
treatment of human beings. non-immune therapies to direct rational care of patients.
Despite such limitations, advances have been made in Conflict of interest statement
the immunosuppressive treatment of experimental We declare that we have no conflict of interest.
glomerulonephritis that might find human application. References
Therapies designed to target each step in the pathogenesis 1 Kerr PG, Chadban SJ, Atkins RC. Rapidly progressive
glomerulonephritis. In: Schrier RW, Gottschalk CW, eds. Diseases of
of glomerulonephritis have been tested in experimental the kidney, 7th edn. Philadelphia: Lippincott, Williams and Wilkins,
models and some examples are summarised in table 1. 2001: 1665–90.
Few have found an approved clinical indication to date. 2 Chadban SJ, Briganti E, Kerr PG, Zimmet P, Shaw J, Atkins RC.
Prevalence of kidney damage in the general community: the AusDiab
Preliminary reports of the successful use of a monoclonal Kidney Study. J Am Soc Nephrol 2003; 14: 5131–38.
antibody against CD20 to deplete B lymphocytes, decrease 3 Coresh J, Astor BC, Greene T, Eknoyan G, Levey AS. Prevalence of
antibody production, and thereby reduce proteinuria and chronic kidney disease and decreased kidney function in the adult
US population: Third National Health and Nutrition Examination
kidney dysfunction in cases of human membranous Survey. Am J Kidney Dis 2003; 41: 1–12.
nephropathy109 and cryoglobulinaemia-associated mesan- 4 Briganti EM, Dowling J, Finlay M, et al. The incidence of biopsy-
giocapillary glomerulonephritis93 indicate the potential to proven glomerulonephritis in Australia. Nephrol Dial Transplant
2001; 16: 1364–67.
achieve selective, mechanism-directed immunosup-
5 Simon P, Ramee MP, Boulahrouz R, et al. Epidemiologic data of
pressive therapies in the near future. primary glomerular diseases in western France. Kidney Int 2004; 66:
Ultimately, many patients with glomerulonephritis 905–08.
progress to end-stage renal failure despite currently 6 Sarnak MJ, Levey AS, Schoolwerth AC, et al. Kidney disease as a risk
factor for development of cardiovascular disease. Circulation 2003;
available therapies. For such patients, transplantation 108: 2154–69.
offers the best means by which to achieve the best quality 7 McDonald S, Russ GR. New patients commencing treatment,
and duration of life. However, transplantation provides ANZDATA Registry Report 2002, http://www.anzdata.org.au
(accessed March 8, 2005).
little barrier to several types of glomerulonephritis that 8 Collins AJ, Kasiske B, Herzog C, et al. Excerpts from the United
show a propensity to recur.110 Despite the presence of States Renal Data System 2003 annual data report: atlas of end-
immunosuppression and the antigenic differences of the stage renal disease in the United States. Am J Kidney Dis 2003; 42
(6 suppl 5): S1–26.
allograft, a substantial proportion of individuals with 9 Streeton CL, Hanna JN, Messer RD, et al. An epidemic of acute post-
primary glomerulonephritis will develop recurrence in the streptococcal glomerulonephritis among aboriginal children.
graft. The clinical course of recurrent glomerulonephritis J Paediatr Child Health 1995; 31: 245–48.
might be different from that of the native disease, but 10 Montseny J, Meyrier A, Kleinknecht D, et al. The current spectrum of
infectious glomerulonephritis. Medicine 1995; 74: 63–73.
recurrence causes graft failure in an increasing number of 11 Johnson RJ, Gretch DR, Yamabe H, et al. Membranoproliferative
patients over time. For those with glomerulonephritis who glomerulonephritis associated with hepatitis C virus infection.
receive a transplant, recurrence is the third commonest N Engl J Med 1993; 328: 465–70.
12 Winston JA, Klotman ME, Klotman PE. HIV-associated nephropathy
cause of graft failure, causing graft loss in 8% at 10 years is a late, not early, manifestation of HIV-1 infection. Kidney Int 1999;
after transplantation.111 55: 1036–40.
13 King K, Flinter FA, Nihalani V, Green PM. Unusual deep intronic
mutations in the COL4A5 gene cause X-linked Alport syndrome.
The future Hum Genet 2002; 111: 548–54.
The way forward in the management of 14 Longo I, Porcedda P, Mari F, et al. COL4A3/COL4A4 mutations:
glomerulonephritis must be multifaceted. First, we need from familial haematuria to autosomal-dominant or recessive Alport
syndrome. Kidney Int 2002; 61: 1947–56.
to refine our knowledge of the epidemiology of the
15 Liu W, Szalai A, Zhao L, et al. Control of spontaneous B lymphocyte
disorder; population-based studies could permit the autoimmunity with adenovirus-encoded soluble TACI. Arthritis
identification of risk factors and potential causative agents. Rheum 2004; 50: 1884–96.
To enable progress, uniformity in the classification, 16 Clynes R, Dumitru C, Ravetch JV. Uncoupling of immune complex
formation and kidney damage in autoimmune glomerulonephritis.
diagnosis (including more routine use of renal biopsy), Science 1998; 279: 1052–54.
and reporting of glomerulonephritis will be needed. 17 Baker P, Ochi R, Schulze M, Johnson R, Campbell C, Couser W.
Second, we need to understand the immunopathogenesis Depletion of C6 prevents development of proteinuria in experimental
membranous nephropathy in rats. Am J Pathol 1989; 135: 185–94.
of glomerulonephritis and to generate molecule-targeted

1804 www.thelancet.com Vol 365 May 21, 2005


Seminar

18 Couser W, Johnson R, Young B, Yeh C, Toth C, Rudolph A. The Mallick N. Urinary C3dg and C5b-9 indicate active immune disease
effects of soluble recombinant complement receptor 1 on in human membranous nephropathy. Kidney Int 1991; 41: 933–37.
complement-mediated experimental glomerulonephritis. 42 Cybulsky A, Quigg R, Salant D. The membrane attack complex
J Am Soc Nephrol 1995; 5: 1888–94. in complement-mediated glomerular epithelial cell injury: formation
19 Wang Y, Madri J, Rollins S, Chodera A, Matis L. Amelioration of and stability of C5b-9 and C5b-7 in rat membranous nephropathy.
lupus-like autoimmune disease in NZB/W F1 mice after treatment J Immunol 1986; 137: 1511–16.
with a blocking monoclonal antibody specific for complement 43 Savin V, Johnson R, Couser W. C5b-9 increases albumin permeability
component 5. Proc Natl Acad Sci USA 1996; 93: 8563–68. of isolated glomeruli in vitro. Kidney Int 1994; 46: 382–88.
20 Cunningham MA, Rondeau E, Chen X, Coughlin SR, 44 Hsu S, Couser WG. Chronic progression of tubulointerstitial damage
Holdsworth SR, Tipping PG. Protease-activated receptor 1 mediates in proteinuric renal disease is mediated by complement activation: a
thrombin-dependent, cell mediated renal inflammation in crescentic therapeutic role for complement inhibitors? J Am Soc Nephrol 2003;
glomerulonephritis. J Exp Med 2000; 191: 455–62. 14: S186–91.
21 Zernecke A, Weber KS, Erwig LP, et al. Combinatorial model of 45 Zhou W, Marsh J, Sacks S. Intrarenal synthesis of complement.
chemokine involvement in glomerular monocyte recruitment: role of Kidney Int 2001; 59: 1227–35.
CXC chemokine receptor 2 in infiltration during nephrotoxic 46 Nangaku M, Pippin J, Couser W. Complement membrane attack
nephritis. J Immunol 2001; 166: 5755–62. complex (C5b-9) mediates interstitial disease in experimental
22 Tang WW, Qi M, Warren JS. Monocyte chemoattractant protein 1 nephrotic syndrome. J Am Soc Nephrol 1999; 10: 2323–31.
mediates glomerular macrophage infiltration in anti-GBM Ab GN. 47 Erlich JH, Apostolopoulos J, Wun TC, Kretzmer KK,
Kidney Int 1996; 50: 665–71. Holdsworth SR, Tipping PG. Renal expression of tissue
23 Tesch GH, Schwarting A, Kinoshita K, Lan HY, Rollins BJ, factor pathway inhibitor and evidence for a role in crescentic
Kelley VR. Monocyte chemoattractant protein-1 promotes glomerulonephritis in rabbits. J Clin Invest 1996; 98: 325–35.
macrophage-mediated tubular injury, but not glomerular injury, in 48 Risdon R, Sloper J, de Wardener H. Relations between renal function
nephrotoxic serum nephritis. J Clin Invest. 1999; 103: 73–80. and histological changes found in renal biopsy specimens from
24 Nikolic-Paterson DJ, Lan HY, Hill PA, Vannice JL, Atkins RC. patients with persistent glomerular nephritis. Lancet 1968; 2: 363–66.
Suppression of experimental glomerulonephritis by the interleukin-1 49 Hooke DH, Gee DC, Atkins RC. Leukocyte analysis using
receptor antagonist: inhibition of intercellular adhesion molecule-1 monoclonal antibodies in human glomerulonephritis. Kidney Int
expression. J Am Soc Nephrol 1994; 4: 1695–700. 1987; 31: 964–72.
25 Le Meur Y, Tesch GH, Hill PA, et al. Macrophage accumulation at a 50 Boulton WK, Chandra M, Tyson TM, Kirkpatrick PR, Sadovnic MJ,
site of renal inflammation is dependent on the M-CSF/c-fms Sturgill BC. Transfer of experimental glomerulonephritis in chickens
pathway. J Leukoc Biol 2002; 72: 530–37. by mononuclear cells. Kidney Int 1988; 34: 598–610.
26 Ophascharoensuk V, Fero ML, Hughes J, Roberts JM, Shankland SJ. 51 Tipping PG, Neale TJ, Holdsworth SR. T lymphocyte participation in
The cyclin-dependent kinase inhibitor p27Kip1 safeguards against antibody-induced experimental glomerulonephritis. Kidney Int 1985;
inflammatory injury. Nat Med 1998; 4: 575–80. 27: 530–37.
27 Gilbert R, Kelly DJ, McKay T, et al. Platelet derived growth factor 52 Groux H. Type 1 T-regulatory cells: their role in the control of
receptor inhibition ameliorates experimental mesangial proliferative immune responses. Transplantation 2003 May 15; 75 (9 suppl): 8S–12.
glomerulonephritis. Kidney Int 2001; 59: 1324–32.
53 Ikezumi Y, Hurst L, Masaki T, Atkins RC, Nicolic-Paterson DJ.
28 Stambe C, Atkins RC, Tesch GH, et al. Blockade of p38alpha MAPK Adoptive transfer studies demonstrate that macrophages can induce
ameliorates acute inflammatory renal injury in rat anti-GBM proteinuria and mesangial cell proliferation. Kidney Int 2003; 63:
glomerulonephritis. J Am Soc Nephrol 2003; 14: 338–51. 83–95.
29 Tomita N, Morishita R, Tomita S, et al. Transcription factor decoy for 54 Nicolic-Paterson, Atkins RC. Macrophages in immune renal injury.
NFkappaB inhibits TNF-alpha-induced cytokine and adhesion In: Neilson E, Couser WG, eds. Immunologic renal diseases.
molecule expression in vivo. Gene Ther 2000; 7: 1326–32. Philadelphia: Lippincott-Raven, 2001: 609–32.
30 Quiza CG, Leenaerts PL, Hall BM. The role of T cells in the 55 Perez de Lema G, Maier H, Nieto E, et al. Chemokine expression
mediation of glomerular injury in Heymann’s nephritis in the rat. precedes inflammatory cell infiltration and chemokine receptor and
Int Immunol 1992; 4: 423–32. cytokine expression during the initiation of murine lupus nephritis.
31 Penny MJ, Boyd RA, Hall BM. Permanent CD8+ T cell depletion J Am Soc Nephrol 2001; 12: 1369–82.
prevents proteinuria in active Heymann nephritis. J Exp Med 1998; 56 Chen S, Bacon KB, Li L, et al. In vivo inhibition of CC and CX3C
188: 1775–84. chemokine induced leukocyte infiltration and attenuation of
32 Bremer V, Tojo A, Kimura K, et al. Role of nitric oxide in rat glomerulonephritis in Wistar-Kyoto rats by vMIP-II. J Exp Med 1998;
nephrotoxic nephritis: comparisons between inducible and 188: 193–98.
constitutive nitric oxide synthetase. J Am Soc Nephrol 1997; 8: 1712–21. 57 Topham PS, Csizmadia V, Soler D, et al. Lack of chemokine receptor
33 Boyle JJ. Human macrophages kill human mesangial cells by CCR1 enhances Th1 responses and glomerular injury during
Fas-L-induced apoptosis when triggered by antibody via CD16. nephrotoxic nephritis. J Clin Invest 1999; 104: 1549–57.
Clin Exp Immunol 2004; 137: 529–37. 58 Segerer S, Cui Y, Hudkins KL, et al. Expression of chemokine
34 Nangaku M. Complement regulatory proteins in glomerulonephritis. monocyte chemoattractant protein-1 and its receptor chemokine
Kidney Int 1998; 54: 1419–28. receptor 2 in human creascentic glomerulonephritis.
35 Leinonen A, Netzer KO, Boutaud A, Gunwar S, Hudson BG. J Am Soc Nephrol 2000; 11: 2231–42.
Goodpasture antigen: expression of the full-length alpha3(IV) chain 59 Lan HY, Nikolic-Paterson DJ, Mu W, Atkins RC. Local macrophage
of collagen IV and localization of epitopes exclusively to the proliferation in the progression of glomerular and tubulointerstitial
noncollagenous domain. Kidney Int 1999; 55: 1120–22. injury in rat anti-GBM glomerulonephritis. Kidney Int 1995; 48:
36 Barratt J, Feehally J, Smith AC. Pathogenesis of IgA nephropathy. 753–60.
Semin Nephrol 2004; 24: 197–217. 60 Yang N, Isbel NM, Nikolic-Paterson DJ, et al. Local macrophage
37 Savige J, Gillis D, Benson E, et al. International consensus statement proliferation in human glomerulonephritis. Kidney Int 1998; 54:
in testing and reporting of ANCA. Am J Pathol 1999; 111: 507–13. 143–51.
38 Sibelius U, Hattar K, Schenkel A, et al. Wegener’s granulomatosis: 61 Isbel NM, Nikolic-Paterson DJ, Hill PA, Dowling J, Atkins RC. Local
anti-proteinase 3 antibodies are potent inducers of human endothelial macrophage proliferation correlates with increased renal M-CSF
cell signaling and leakage response. J Exp Med 1998; 187: 497–503. expression in human glomerulonephritis. Nephrol Dial Transplant
39 Xiao H, Heeringa P, Hu P, et al. Antineutrophil cytoplasmic 2001; 16: 1638–47.
autoantibodies specific for myeloperoxidase cause glomerulonephritis 62 Floege J, von Ostendorf T, Wolf G. Growth factors and cytokines. In:
and vasculitis in mice. J Clin Invest 2002; 110: 955–63. Couser W, Neilson EG, eds. Immunologic renal diseases, 2nd edn.
40 Morita Y, Ikeguchi H, Nakamura J, Hotta N, Yuzawa Y, Matsuo S: Philadelphia: Lippincott-Raven, 2001: 415–63.
Complement activation products in the urine from proteinuric 63 Boyce NW, Tipping PG, Holdsworth SR. Glomerular macrophages
patients. J Am Soc Nephrol 2000; 11: 700–07. produce reactive oxygen species in experimental glomerulonephritis.
41 Brenchley P, Coupes B, Short C, O’Donoghue D, Ballardie F, Kidney Int 1989; 35: 778–82.

www.thelancet.com Vol 365 May 21, 2005 1805


Seminar

64 Baker AJ, Mooney A, Hughes J, Lombardi D, Johnson RJ, Savill J. frequently relapsing idiopathic nephrotic syndrome: a multicentre
Mesangial cell apoptosis: the major mechanism for resolution of randomized controlled trial. Nephrol Dial Transplant 1993; 8:
glomerular hypercellularity in experimental mesangial proliferative 1326–32.
nephritis. J Clin Invest 1994; 94: 2105–16. 88 Rydel JJ, Korbet SM, Borok RZ, Schwartz MM. Focal segmental
65 Kaneko Y, Sakatsume M, Xie Y, et al. Macrophage metalloelastase as glomerular sclerosis in adults: presentation, course and response to
a major factor for glomerular injury in anti-glomerular basement therapy. Am J Kidney Dis 1995; 25: 534–42.
membrane nephritis. J Immunol 2003; 170: 3377–85. 89 Burgess E. Management of focal segmental glomerulosclerosis:
66 Hewitson TD, Becker GJ. Interstitial myofibroblasts in IgA evidence-based recommendations. Kidney Int Suppl 1999; 70: S26–32.
glomerulonephritis. Am J Nephrol 1995; 15: 111–17. 90 Ponticelli C, Zucchelli P, Passerini IP, Cesena B. Methylprednisolone
67 Johnson RJ, Floege J, Yoshimura A, Iida H, Couser WG, Alpers CE. plus chlorambucil as compared with methylprednisolone alone for
The activated mesangial cell: a glomerular “myofibroblast”? the treatment of idiopathic membranous nephropathy. N Engl J Med
J Am Soc Nephrol 1992; 2 (10 suppl): S190–97. 1992; 327: 599–603.
68 Border WA, Okuda S, Languino LR, Sporn MB, Ruoslahti E. 91 Muirhead N. Management of idiopathic membranous nephropathy:
Suppression of experimental glomerulonephritis by anti-serum evidence-based recommendations. Kidney Int Suppl 1999; 70: S47–55.
against TGF-beta. Nature 1990; 346: 371–74. 92 Tarshish P, Bernstein J, Tobin JN, Edelmann CM Jr. Treatment of
69 Tryggvason K. Unraveling the mechanisms of glomerular mesangiocapillary glomerulonephritis with alternate-day prednisone:
ultrafiltration: nephrin, a key component of the slit diaphragm. a report of the International Study of Kidney Disease in Children.
J Am Soc Nephrol 1999; 10: 2440–45. Pediatr Nephrol 1992; 6: 123–30.
70 Hughes J, Nangaku M, Alpers CE, Shankland SJ, Couser WG, 93 Roccatello D, Baldovino S, Rossi D, et al. Long-term effects of anti-
Johnson RJ. C5b-9 membrane attack complex mediates endothelial CD20 monoclonal antibody treatment of cryoglobulinaemic
cell apoptosis in experimental glomerulonephritis. Am J Physiol Renal glomerulonephritis. Nephrol Dial Transplant 2004; 19: 3054–61.
Physiol 2000; 278: F747–57. 94 Levin A. Management of membranoproliferative glomerulonephritis:
71 Kim YG, Suga SI, Kang DH, et al. Vascular endothelial growth factor evidence based recommendation. Kidney Int 1999; 55 (suppl 70):
accelerates renal recovery in experimental thrombotic S41–46
microangiopathy. Kidney Int 2000; 58: 2390–99. 95 Boumpas DT, Austin HA, Vaughn EM, et al. Controlled trial of pulse
72 Floege J, Eng E, Young BA, Johnson RJ. Factors involved in the methylprednisolone versus two regimens of pulse cyclophosphamide
regulation of mesangial cell proliferation in vitro and in vivo. Kidney in severe lupus nephritis. Lancet 1992; 340: 741–45.
Int 1993; 39: S47–54. 96 Flanc RS, Roberts MA, Strippoli G, Chadban SJ, Kerr PG, Atkins RC.
73 Johnson RJ, Floege J, Yoshimura A, Iida H, Couser WG, Alpers CE. Treatment of proliferative lupus nephritis: a meta-analysis of
The activated mesangial cell: a glomerular “myofibroblast”? randomized controlled trials. Am J Kidney Dis 2004; 43: 197–208.
J Am Soc Nephrol 1992; 2 (suppl 10): S190–97. 97 Houssiau FA, Vasconcelos C, D’Cruz D, et al. Early response to
74 D’Amico G, Ferrario F, Pia Rastaldi M. Tubolointerstitial damage in immunosuppressive therapy predicts good renal outcome in lupus
glomerular diseases: its role in the progression of renal damage. nephritis: lessons from long-term follow-up of patients in the Euro-
Am J Kidney Dis 1995; 26: 124–32. Lupus Nephritis trial. Arthritis Rheum 2004; 50: 3934–40.
75 Tesch GH, Yang N, Yu H, et al. Intrinsic renal cells are the major 98 Contreras G, Pardo V, Leclercq B, et al. Sequential therapies for
source of IL-1‚ synthesis in normal and diseased kidney. proliferative lupus nephritis. N Engl J Med 2004; 350: 971–80.
Nephrol Dial Transplant 1997; 12: 1109–15. 99 Couser WG. Rapidly progressive glomerulonephritis: classification,
76 Lan HY. Tubular epithelial-myofibroblast transdifferentiation pathogenetic mechanisms and therapy. Am J Kidney Dis 1988; 11:
mechanisms in proximal tubule cells. Curr Opin Nephrol Hypertens 449–64.
2003; 12: 25–29. 100 Nachman PH, Hogan SL, Jeannette JC, Falk RJ. Treatment response
77 Klahr S, Levey AS, Beck GJ, et al. The effects of dietary protein and relapse in antineutrophil cytoplasmic autoantibody-associated
restriction and blood pressure control on the progression of chronic microscopic polyangiitis and glomerulonephritis. J Am Soc Nephrol
renal disease: Modification of Diet in Renal Disease Study Group. 1996; 7: 33–39.
N Engl J Med 1994; 330: 877–84. 101 Pozzi C, Bolasco PG, Fogazzi GB, et al. Corticosteroids in IgA
78 Morrissey JJ, Klahr S. Differential effects of ACE and AT1 receptor nephropathy: a randomised controlled trial. Lancet 1999; 353: 883–87.
inhibition on chemoattractant and adhesion molecule synthesis. 102 Lewis EJ, Hunsicker LG, Lan SP, Rohde RD, Lachin JM. A controlled
Am J Physiol 1998; 274: F580–86. trial of plasmapheresis therapy in severe lupus nephritis.
79 Trivedi HS, Pang MM, Campbell A, Saab P. Slowing the progression N Engl J Med 1992; 326: 1373–79.
of chronic renal failure: economic benefits and patients’ perspectives. 103 Levin ML. Mycophenolate mofetil treatment for primary glomerular
Am J Kidney Dis 2002; 39: 721–29. diseases. Kidney Int 2002; 62: 1475.
80 Bergstrom J, Alvestrand A, Bucht H, Gutierrez A. Progression of 104 Fervenza FC, Fitzpatrick PM, Mertz J, et al. Acute rapamycin
chronic renal failure in man is retarded with more frequent clinical nephrotoxicity in native kidneys of patients with chronic
follow-ups and better blood pressure control. Clin Nephrol 1986; 25: glomerulonephropathies. Nephrol Dial Transplant 2004; 19: 1288–92.
1–6. 105 Cochrane Renal Group. The renal health library; evidence based
81 GISEN Group. Randomised placebo-controlled trial of effect of information for the nephrology community. Issue 1, 2003. Update
ramipril on decline in glomerular filtration rate and risk of terminal software. ISSN 1478–128X.
renal failure in proteinuric, non-diabetic nephropathy. Lancet 1997; 106 Kitching AR, Tipping PG, Huang XR, Mutch DA, Holdsworth SR.
349: 1857–63. Interleukin-4 and interleukin-10 attenuate established crescentic
82 Ruggenenti P, Perna A, Gherardi G, et al. Renal function and glomerulonephritis in mice. Kidney Int 1997; 52: 52–59.
requirement for dialysis in chronic nephropathy patients on long- 107 Chadban SJ, Tesch G, Lan HY, Atkins RC, Nikolic-Paterson D. Effect
term ramipril: REIN follow-up trial. Lancet 1998; 352: 1252–56. of IL-10 treatment on experimental crescentic glomerulonephritis in
83 Nakao N, Yoshimura A, Morita H, et al. Combination treatment of rat. Kidney Int 1997; 51: 1809–17.
angiotensin-II receptor blocker and angiotensin-converting-enzyme 108 Suto TS, Fine LG, Shimizu F, Kitamura M. In vivo transfer of
inhibitor in non-diabetic renal disease (COOPERATE): a randomised engineered macrophages into the glomerulus: endogenous
controlled trial. Lancet 2003; 361: 117–24. TGF-beta-mediated defense against macrophage-induced
84 Locatelli F, Alberti D, Graziani G, et al. Prospective, randomised glomerular cell activation. J Immunol 1997; 159: 2476–83.
multicentre trial of effect of protein restriction on the progression of 109 Remuzzi G, Chiurchiu C, Abbate M, Brusegan V, Bontempelli M,
chronic renal insufficiency. Lancet 1991; 337: 1299–304. Ruggenenti P. Rituximab for idiopathic membranous nephropathy.
85 Wheeler DC. Does lipid-lowering therapy slow progression of Lancet 2002; 360: 923–24.
chronic kidney disease? Am J Kidney Dis 2004; 44: 917–20. 110 Chadban SJ. Glomerulonephritis recurrence in the renal graft.
86 Bargman JM. Management of minimal lesion glomerulonephritis: J Am Soc Nephrol 2001; 12: 394–402.
evidence-based recommendations. Kidney Int Suppl 1999; 70: S3–16. 111 Brigantte E, Russ G, McNeil JJ, Atkins RC, Chadban SJ. Long-term
87 Ponticelli C, Edefonti A, Ghio L, et al. Cyclosporin versus risk of renal allograft loss due to recurrent glomerulonephritis.
cyclophosphamide for patients with steroid-dependent and N Engl J Med 2002; 347: 103–09.

1806 www.thelancet.com Vol 365 May 21, 2005

You might also like