You are on page 1of 8

J Am Soc Nephrol 12: 1558 –1565, 2001

DISEASE OF THE MONTH


Eberhard Ritz, Feature Editor

Light Chain Deposition Disease: A Model of


Glomerulosclerosis Defined at the Molecular Level
PIERRE M. RONCO,* MARIE-ALEXANDRA ALYANAKIAN,†
BÉATRICE MOUGENOT,* and PIERRE AUCOUTURIER†
*Renal Division, INSERM Unit 489 and University Pierre et Marie Curie (Paris 6), Hôpital Tenon
(Assistance Publique–Hôpitaux de Paris), and †INSERM Unit 25 and University René Descartes (Paris 5),
Hôpital Necker, Paris, France.

Because the renal plasma flow represents 20% of the total tonovych et al. (5) and confirmed by Randall et al. (6), who
plasma flow and the glomerulus is the renal filtering unit published in 1976 the first description of LC deposition disease
continuously exposed to plasma proteins, the glomerulus is a (LCDD). Although the deposits in AL amyloidosis contain
prominent target for deposition of abnormal proteins or pro- monoclonal LC or LC fragments, the term light-chain deposi-
teins with a peculiar affinity for constituents of the capillary tion disease is used only for the disease that features nonamy-
wall. Among these proteins, monoclonal Ig or their heavy- loid electron-dense granular deposits as described by Randall
chain (HC) or light-chain (LC) subunits are responsible for a et al. (6). Monoclonal HC were found together with LC in the
wide spectrum of glomerular diseases that can be classified tissue deposits from certain patients (6,7), and the term light-
into two categories by electron microscopy (Table 1). The first and heavy-chain deposition disease (LHCDD) (1) was pro-
category is those with organized deposits and includes diseases posed. More recently, deposits containing monoclonal HC in
with fibril formation, mainly amyloidosis, and diseases with the absence of detectable LC were observed in patients who
microtubule formation, including cryoglobulinemic kidney and were affected by otherwise typical Randall’s disease (heavy-
immunotactoid glomerulonephritis. Only amyloid deposits chain deposition disease [HCDD]) (8), and the first series of
stain for Congo red, a tinctorial property related to the four patients was reported in 1999 (9).
␤-pleated sheet organization of amyloid fibrils common to all
forms of amyloid. The second category of diseases is charac- Pathogenesis
terized by nonorganized electron-dense granular deposits. At variance with amyloidosis, lesions in MIDD are related
They are localized in basement membranes in most tissues, clearly to accumulation of extracellular matrices (ECM). The
especially the kidney, and define a disease now called mono- Ig chains that are responsible for this effect display peculiar
clonal Ig deposition disease (MIDD) (1,2), a term that by structural properties and proneness to precipitate in certain
convention excludes Ig-derived amyloidosis. MIDD differs extracellular areas where they most likely stimulate ECM
from amyloidosis by the lack of affinity for Congo red. The production.
distinction between amyloid light-chain (AL) amyloidosis and
MIDD also is justified by the different pathophysiology of
Ig Chain Structure
amyloid that implicates one-dimensional elongation of a
Ig are made up of two pairs of subunits: two HC and two LC,
pseudocrystalline structure and MIDD that involves nonorga-
each of which includes a variable domain implicated in antigen
nized amorphous precipitation of Ig chains (see Pathogenesis
recognition, and a constant region that itself includes one or
section).
several domains and that activates a variety of effector sys-
tems. Binding to antigen and activating effector cells or com-
History plement (i.e., antibody function) is made possible by the asso-
It was known from the late 1950s that nonamyloidotic forms
ciation of two HC and two LC. LC may be of ␬ or ␭ type, with
of glomerular disease resembling diabetic glomerulosclerosis
one constant and one variable domain (termed CL and VL). HC
could occur in multiple myeloma (3,4). The presence of mono-
may be of nine isotypes (␮, ␥1, ␥2, ␥3, ␥4, ␣1, ␣2, ␦, and ␧)
clonal LC in these lesions was recognized in 1973 by An-
that define Ig classes and subclasses: IgM, IgG (including IgG1
to IgG4), IgA (including IgA1 and IgA2), IgD, and IgE. HC
include from the ⫺NH2 to the ⫺COOH terminus one variable
Correspondence to Dr. Pierre Ronco, INSERM U 489, Hôpital Tenon, 4 rue de
domain (VH) from a common repertoire for all isotypes and
la Chine, 75020 Paris, France. Phone: 33-1-56-01-66-39; Fax: 33-1-56-01-69-
99; E-mail: pierre.ronco@tnn.ap-hop-paris.fr three (for IgG, IgA, and IgD) or four (for IgM and IgE)
1046-6673/1207-1558 constant domains, termed CH1 to CH4 (see Figure 1).
Journal of the American Society of Nephrology Normal Ig display a striking structural heterogeneity that is
Copyright © 2001 by the American Society of Nephrology borne essentially by the variable domains and is related to the
J Am Soc Nephrol 12: 1558 –1565, 2001 Light Chain Deposition Disease 1559

Table 1. Glomerular diseases with monoclonal immunoglobulin deposition


Immunoglobulin Deposits Glomerular Disease

Organized
fibrillar Amyloidosis
microtubular Cryoglobulinemia, immunotactoid glomerulonephritis
Nonorganized, granular Monoclonal immunoglobulin deposition disease:
light chain, heavy chain, and light plus heavy
chain deposition disease

diversity of the antigenic determinants that they are supposed ␭ to ␬ ratio seen in amyloidosis. Data on N-terminal sequences
to recognize. These regions of the Ig chains are encoded by of the LC from six consecutive patients with LCDD suggested
numerous gene segments, the rearrangement of which occurs an overrepresentation of the rare V␬IV variability subgroup
during B-cell differentiation and is mandatory for Ig produc- (10). This subgroup features a longer CDR1 loop that contains
tion. Comparison of VH and VL sequences allowed definition some hydrophobic residues. The role of LC variable region
of variability subgroups on the basis of their homologies: HC (VL) in LC deposition is suggested by the fact that amino acid
express six VH subgroups, ␬ chains express four V␬ subgroups, changes in VL were sufficient to promote tissue deposition in
and ␭ chains express six V␭ subgroups. The variability of VH mice that expressed a human LCDD V␬IV chain (11).
and VL is even higher in three small peptidic portions called The primary structures of a few additional LCDD precursors
complementarity determining regions (CDR1, CDR2, and were analyzed at the cDNA (12,13) and protein levels (14). As
CDR3) that form three loops at one end of the domain that in AL amyloidosis, no common structural motif emerged from
constitutes the antibody binding site. these studies. The most remarkable observations were unusual
At variance with normal polyclonal Ig, monoclonal Ig are hydrophobic residues at positions where they either could be
secreted by a single clone of differentiated B cells that expands exposed to the solvent or strongly modify the conformation,
excessively, either in a tumoral context (myeloma, Walden- especially in the CDR (13,14). In particular, molecular mod-
ström’s disease, etc.) or without patent hematologic malig- eling experiments performed on V␬I and V␬IV LC underline
nancy (monoclonal gammopathy of undetermined signifi- the presence of leucine, isoleucine, or tyrosine at positions 27
cance). A monoclonal Ig may be secreted as free LC (or rarely, and/or 31 in all known cases of LCDD. Other nonpolar groups
HC) that displays structural anomalies causing tissue may be exposed on CDR regions, which suggests that hydro-
deposition. phobic interactions are important either in the amorphous pre-
cipitation of LC or in the mechanisms that lead to overproduc-
Abnormal Structures of LC Variable Domains tion of ECM components (15). Of note, in contrast to LCDD
In LCDD, isotype restriction is significant; ␬ chains occur in LC, those involved in AL amyloidosis often present particular
approximately 80% of cases. This contrasts with the increased acidic residues in CDR and might form fibrils through elec-
trostatic interactions.
When pathogenic LC could not be detected in the serum and
urine, which occurred in 15 to 30% of patients, they seemed to
be N-glycosylated in all tested cases (10,16). In vitro biosyn-
thetic labeling experiments on short-term cultures showed that
LC that were absent in the urine actually were secreted by the
bone marrow plasma cells (1,17,18). Thus, together with the
presence of hydrophobic residues, glycosylation might in-
crease the LC propensity to precipitate in tissues and displace
the equilibrium from soluble toward deposited amorphous
forms.

Truncated HC
Twenty-two cases of HCDD have been reported (19 and
reviewed in reference 20). In all patients, a monotypic HC
without LC was detected in the deposits. A deletion of the CH1
domain was found in the deposited or circulating HC in the 10
patients with ␥ HCDD where it was searched for. It also was
Figure 1. Domain structure of an IgG. The light- (LC) and heavy- suggested in a patient with ␣ HCDD (21). Deletions of the
chain domains are shown in gray and white, respectively. VL, light- CH1, hinge, and CH2 domains were found in one case (8).
chain (LC) variable domain; CL, LC constant domain; VH, heavy- The loss of the CH1 domain suggests that secretion of the
chain (HC) variable domain; CH, HC constant domain. free, abnormal HC is required for tissue deposition. Normal
1560 Journal of the American Society of Nephrology J Am Soc Nephrol 12: 1558 –1565, 2001

HC associate posttranslationally with Ig binding protein (BIP) tubular basement membrane thickening (19,25). One patient
in the endoplasmic reticulum. The LC later assemble with HC, (Aucouturier P, Droz D, personal data) had an important LC
and the complex is transferred to the Golgi apparatus for secretion and typical basement membrane deposits by immu-
further processing and secretion. Because the binding site of nofluorescence in the kidney, without histologic alteration, and
BIP is located in the CH1 domain, when a mutant HC lacks the with normal renal functions 2 yr after kidney biopsy. Thus, the
CH1 domain, it fails to associate with BIP and thus may be propensity of a given LC to form deposits does not necessarily
secreted as a free subunit in the circulation. However, CH1 mean that it is pathogenic, and immunofluorescence staining
deletion seems necessary but not sufficient for deposition, and should not be considered a sufficient criterion for pathologic
it is likely that the VH also contributes to tissue deposition, diagnosis of MIDD. As shown by characteristic pathologic
ultrastructural aspects of the deposits, and ECM accumulation. changes and experimental evidence, MIDD lesions are associ-
Indeed, in HC disease, a lymphoproliferative disorder with free ated with local fibrosis.
HC secretion without corresponding tissue deposition, the vari-
able regions are found to be deleted partially or entirely, LC-Induced Mesangial Fibrosis
together with the CH1 domain. In HCDD, the variable regions Results of an in vitro study (26) suggest that pathogenic Ig
are present without major structural alteration, although in the chains may stimulate mesangial cells to secrete ECM compo-
two cases in which they were sequenced (8,22), they contained nents through growth factors, in particular transforming growth
unusual amino acid substitutions that might change the phys- factor-␤ (TGF-␤). LC-induced overproduction of collagen IV,
icochemical properties (e.g., charge, hydrophobicity). In addi- laminin, fibronectin, and tenascin was shown to be maximal at
tion, the structure of an HCDD HC (8) was strikingly similar to 72 h of incubation with mesangial cells (27). Accumulation
that reported in a case of amyloid HC amyloidosis (23), from may be increased by a concomitant inhibition of collagenase
which it differed essentially at the V domain level. It is worth IV, which also is mediated by TGF-␤. None of these effects
noting that in most cases of HCDD, the circulating HC is was found with amyloid LC (27). One way to prevent the
associated with a ␭-type LC. This bias may reflect a preferen- LC-induced fibrosis is to study the detailed mechanism of
tial association of the VH with ␭ VL domains. putative ligand-receptor interactions that may govern the abil-
ity of a given LC to stimulate the ECM synthesis. Other
Deposition Does Not Mean Pathogenicity possibilities are to prevent the secretion of TGF-␤ or to inter-
The finding by Solomon et al. (24) of unexpectedly frequent fere with the TGF-␤ signaling pathway.
(14 of 40) deposition of human monoclonal LC along basement
membranes in a mouse experimental model raises the question Renal Pathology
of the relationship between tissue precipitation and pathogenic Light Microscopy
effects. Human LC that were found deposited along basement Despite clinical manifestations that feature impairment of
membranes in mice were predominantly of the ␭ type (9 of 14), glomerular function in most cases, MIDD should not be con-
contrasting with the striking predominance of ␬ chains in sidered a purely glomerular disease. In fact, tubular lesions
MIDD (24). In addition, LC deposition similar in aspect to may be more conspicuous than the glomerular damage. Tubu-
LCDD by immunofluorescence but with no or only scanty lar lesions are characterized by the deposition of a refractile,
granular electron-dense deposits in the tubular basement mem- eosinophilic, periodic acid-Schiff (PAS)-positive, ribbon-like
brane may occur in the absence of glomerular lesions and material along the tubular basement membrane (Figure 2A).

Figure 2. Light microscopy of an LC nephropathy with predominant tubulointerstitial lesions. (A) Thickening of tubular basement membranes
and diffuse interstitial fibrosis. (B) Strongly periodic acid-Schiff (PAS)-positive deposits delineating the spaces between myocytes in an
interlobular artery. Magnifications: ⫻125 in A (PAS); ⫻312 in B (PAS).
J Am Soc Nephrol 12: 1558 –1565, 2001 Light Chain Deposition Disease 1561

The deposits predominate around the distal tubules, the loops cence was negative despite the presence of large amounts of
of Henle, and, in some instances, the collecting ducts whose granular glomerular deposits by electron microscopy have been
epithelium is flattened and atrophied. Typical myeloma casts reported (31). Local modifications of deposited LC thus might
are only occasionally seen in pure forms of MIDD (see below change their antigenicity (18). In patients without nodular
for association with myeloma cast nephropathy). In advanced lesion (Figure 3A), glomerular staining occurs mainly along
stages, a marked interstitial fibrosis including refractile depos- the basement membrane, but it may involve the mesangium in
its frequently is associated with tubular lesions. some cases. A linear staining usually decorates Bowman’s
Glomerular lesions are much more heterogeneous (28,29). capsule. Deposits frequently are found in vascular walls and
Nodular glomerulosclerosis (NGS) is the most characteristic; it interstitium.
is found in 60 (30) to 100% (19) of patients with LCDD. In patients with HCDD, immunofluorescence with anti-LC
Expansion of the mesangial ECM was observed in all cases of polyclonal and monoclonal antibodies is negative despite typ-
HCDD, with NGS in almost all of them. Mesangial nodules are ical NGS. Monotypic deposits of ␥, ␣, or ␮ HC may be
composed of PAS-positive membrane-like material and often identified. All ␥ subclasses may be observed. Analysis of the
are accompanied by mild mesangial hypercellularity. The cap- kidney biopsy with monoclonal antibodies specific for the
illary loops stretch at the periphery of florid nodules and may constant domains of the ␥ HC allowed identification of a
undergo aneurysmal dilation. Bowman’s capsule may contain a deletion of the CH1 domain in all tested cases. In most cases of
material that is identical to that present in the center of the HCDD, except those with deposits of ␥ 4 that does not activate
nodules. These lesions resemble nodular diabetic glomerulo- complement, complement components could be demonstrated
sclerosis, but some characteristics are distinctive: the distribu- in a granular or pseudolinear pattern.
tion of the nodules is fairly regular in a given glomerulus, the The accessory proteins (serum amyloid P component, apo-
nodules are poorly argyrophilic, and exudative lesions as “fi- lipoprotein E, and ubiquitin) that are associated with AL amy-
brin caps” and extensive hyalinosis of the efferent arterioles are loidosis and other amyloidoses are not present in LCDD
not observed. In occasional cases with prominent endocapillary deposits.
cellularity and mesangial interposition, the glomerular features The composition of glomerular matrix proteins has been
mimic lobular glomerulonephritis. Milder forms of LCDD examined comparatively in NGS associated with LCDD and
simply show an increase in mesangial matrix and sometimes in diabetes mellitus (32). Nodules are made of normal ECM
mesangial cells and a modest thickening of the basement constituents (collagen type IV, laminin, fibronectin) that are
membranes that are abnormally bright and rigid. Glomerular produced in excess and stain weakly for the small proteogly-
lesions may not even be detected by light microscopy but cans, decorin and biglycan (33). In a series of 36 patients with
require ultrastructural examination. These lesions may repre- LC-related renal diseases including AL amyloidosis, cast ne-
sent early stages of glomerular disease or be induced by LC phropathy, fibrillary glomerulopathy, and LCDD, TGF-␤ was
with a weak pathogenic potential. Their diagnosis would be detected only in glomeruli of the 3 patients with LCDD and
unrecognized without the immunostaining results. Arteries, nodular glomerular lesions (34). In the control series, it was
arterioles, and peritubular capillaries all may contain PAS- found essentially in nodular diabetic glomerulosclerosis, which
positive deposits in close contact with their basement mem- may suggest that distinct initial insults to the glomerular mes-
branes (Figure 2B). Deposits do not show the staining charac- angium may trigger similar fibrogenetic pathways.
teristics of amyloid, but they may be associated with Congo
red–positive amyloid deposits in approximately 10% of pa- Electron Microscopy
tients (19). The most characteristic ultrastructural feature is the presence
of finely to coarsely granular electron-dense deposits along the
Immunofluorescence Microscopy outer (interstitial) aspect of the tubular basement membranes.
A key step in the diagnosis of the various forms of MIDD is In the glomerulus, they predominate in a subendothelial posi-
immunofluorescence examination of the kidney. All biopsy tion along the glomerular basement membrane and are located
specimens show evidence of monotypic LC (mostly ␬) and/or mainly along and in the lamina rara interna. They also can be
HC fixation along tubular basement membranes. This criterion found in mesangial nodules, Bowman’s capsule, and the wall
is requested for the diagnosis of MIDD. of small arteries between the myocytes. Nonamyloid fibrils
The tubular deposits stain strongly and predominate along have been reported in a few patients with LCDD or HCDD.
the loops of Henle and the distal tubules, but they also often are
detected along the proximal tubules. In contrast, the pattern of Association with Myeloma Cast Nephropathy
glomerular immunofluorescence displays marked heterogene- The association of monoclonal LC deposits, mostly along
ity. In patients with NGS, deposits of monotypic Ig chains renal tubule membranes, with typical myeloma cast nephrop-
usually are found along the peripheral glomerular basement athy is more frequent than reported initially (see Figure 3, B
membranes and, to a lesser extent, in the nodules themselves. and C). It was found in 23 of 72 (32%) patients with nonamy-
The staining in glomeruli typically is weaker than that ob- loid monoclonal LC deposits in a French series (30) and in 11
served along the tubular basement membranes. This may not of 34 (32%) patients with MIDD in a recent North American
be a function of the actual amount of deposited material, study (19). NGS is, however, infrequent (⬍10%), and some
because several cases in which glomerular immunofluores- ribbon-like tubular basement membranes are seen in fewer than
1562 Journal of the American Society of Nephrology J Am Soc Nephrol 12: 1558 –1565, 2001

half of the patients (30). In addition, one third of the patients do Renal Disease
not have granular-dense deposits by electron microscopy. The Renal involvement is a constant feature of MIDD, and
lack of ECM accumulation in most of these patients who proteinuria (composed mostly of albumin) and renal failure
present with acute renal failure in the setting of a true myeloma often dominate the clinical presentation. In 23 to 53% of the
may be due to insufficient time for the development of fibrosis patients, albuminuria is associated with nephrotic syndrome.
or to a weaker sclerogenic effect of the LC, if any. As dis- However, in approximately 25% of them, it is less than 1 g/d,
cussed previously, the presence of LC deposits along the and these patients exhibit mainly a tubulointerstitial syndrome
tubular basement membrane is not sufficient to make a diag- (Figure 2A). Albuminuria is not correlated with the existence
nosis of MIDD. of NGS, at least initially, and may occur in the absence of
significant glomerular lesions by light microscopy. Hematuria
Clinical Features of LCDD is more frequent (29 to 67%) than one would expect for a
The main clinical features of MIDD are renal and cardiac nephropathy in which cell proliferation usually is modest, with
manifestations, as in AL amyloidosis. Data collected from the a few exceptions. The prevalence of hypertension is variable
largest series as yet published (1,19,29,35–37) show an unex- but must be interpreted according to medical history.
pectedly wide range of affected ages (35 to 80 yr) with a male The high prevalence (⬎90%), early appearance, and severity
gender preponderance. Mean age tends to be higher in patients of renal failure are other salient features of MIDD. In most
with LCDD and myeloma cast nephropathy (67 yr) (19) than in cases, renal function declines rapidly, which is a main reason
those with pure MIDD (51 to 58 yr). for referral.

Figure 3. (A) Immunofluorescence of renal biopsy specimen from a ␬ LC nephropathy without nodular glomerulosclerosis. Bright staining is
shown along tubular basement membranes, and heavy deposits are shown in an arteriolar wall. Glomerular staining involves Bowman’s capsule
and capillary basement membranes as well as mildly increased mesangium. (B) In a patient who presented with overt myeloma and acute renal
failure, direct immunofluorescence of renal biopsy specimen with the use of anti-␬ antibody showed numerous myeloma casts and staining of
most of the tubular basement membranes. (C) In the same patient, examination of a post mortem liver biopsy revealed ␬ deposits along
sinusoids. (D) Renal biopsy from a patient with HC deposition disease. Direct immunofluorescence with fluorescein-conjugated anti-␥ antibody
shows bright staining of mesangial nodular areas and peripheral capillary walls but, in this case, no staining along basement membranes of the
surrounding tubules. Magnification, ⫻312.
J Am Soc Nephrol 12: 1558 –1565, 2001 Light Chain Deposition Disease 1563

Renal features of the 22 patients with HCDD basically are terstitial nephritis or who has echocardiographic findings that
similar to those seen in LCDD and LHCDD (see below). indicate diastolic dysfunction and a monoclonal Ig component
in the serum and/or the urine. The same combination also is
Extrarenal Manifestations seen in AL amyloidosis, but the latter more often is associated
MIDD is a systemic disease, but visceral LC deposits may with the ␭ LC isotype. Because sensitive techniques, including
be totally asymptomatic and found only at autopsy. Liver and immunofixation, fail to identify a monoclonal Ig component in
cardiac involvements are the most common (29). 15 to 30% of patients, renal biopsy plays an essential role in the
Liver deposits were constant in patients whose liver was ex- diagnosis of MIDD and of the associated dysproteinemia.
amined (38) (Figure 3C). They are discrete, confined to sinusoids The definitive diagnosis is made by the immunohistologic
and basement membranes of biliary ductules without associated analysis of tissue from an affected organ, in most cases the
parenchymal lesions, or massive with marked dilation and multi- kidney, with the use of a panel of Ig chain–specific antibodies,
ple ruptures of sinusoids resembling peliosis. Hepatomegaly with including anti-␬ and anti-␭ LC antibodies to stain the noncon-
mild alterations of liver function tests are the most usual symp- gophilic deposits. When the biopsy stains for a single HC
toms, but several patients develop hepatic insufficiency and portal isotype and does not stain for LC isotypes, the diagnosis of
hypertension, and some of them die of hepatic failure (29). HCDD should be suspected (see below).
Cardiac manifestations have been noted in as many as 80% The diagnosis of plasma cell dyscrasia relies on bone mar-
of the reported cases of LCDD, but they must be interpreted row aspiration and bone marrow biopsy with cell morphologic
with caution because of other age-dependent potential causes evaluation and, if necessary, immunophenotyping with anti-␬
of heart disease. Arrhythmias, conduction disturbances, and and anti-␭ antisera to demonstrate monoclonality. Diagnostic
congestive heart failure are seen. Echocardiography and cath- criteria for a multiple myeloma are present in no more than
eterization may reveal diastolic dysfunction and a reduction in 50% of the patients with LCDD.
myocardial compliance similar to that seen in cardiac amyloid.
As in the kidney and the liver, immunofluorescence showed
monotypic LC deposits in the vascular walls and perivascular Variants of LCDD: LHCDD and HCDD
areas of the heart in all autopsy cases (16). A monotypic HC is associated with the monotypic LC in
Deposits also may occur along the nerve fibers and in the approximately 10% of patients with LCDD. Whether HC and
choroid plexus, as well as in the lymph nodes, bone marrow, LC precipitate as independent subunits or as a whole Ig mol-
spleen, pancreas, thyroid gland, submandibular glands, adrenal ecule remains to be established. In one case, we found different
glands, gastrointestinal tract, abdominal vessels, lungs, and patterns of kidney deposition of the HC and LC (Rose C,
skin. They may be responsible for peripheral neuropathy (20% unpublished observation). Anomalies of the HC structure sug-
of the reported cases), gastrointestinal disturbances, pulmonary gesting deletion were demonstrated in LHCDD (7). Clinical
nodules, amyloid-like arthropathy, and sicca syndrome. presentation and pathologic data in patients with LHCDD are
similar to those in LCDD.
Hematologic Disease The first patients with HCDD were reported in 1993 (8).
The most common underlying disease in MIDD is myeloma, Twenty-two cases have been described so far (19 and reviewed
which accounts for 40 to 50% of pure MIDD (1,19,29,35–37) and in reference 20). The clinical and pathologic features of HCDD
⬎90% of LC deposits associated with myeloma cast nephropathy. basically are the same as in LCDD, although several differ-
MIDD and AL amyloidosis are found at postmortem examination ences can be noted. First, lesions of NGS are constant in
in 5 and 10% of myeloma patients, respectively (39). MIDD, like patients with HCDD, whereas only a faint staining of tubular
AL amyloidosis, often is the presenting disease that leads to the basement membranes was seen in some patients (Figure 3D).
discovery of myeloma at an early stage. In some patients who first Second, extrarenal deposits are less frequent in these patients
presented with “common” myeloma and with normal-sized than in those with LCDD. They have been reported in heart, in
monoclonal Ig without kidney disease, LCDD occurred when the synovial tissue, in skin, in striated muscles, in pancreas, around
disease relapsed after chemotherapy, together with Ig structural thyroid follicles, and in Disse spaces of the liver (8 and
abnormalities (7,16). Because melphalan was shown to induce Ig reviewed in reference 20). Third, signs of complement activa-
gene mutations, the disease in these patients might result from the tion with renal complement deposition are present in most
emergence of a variant clone induced by the alkylating agent. patients with ␥1 or ␥3 HCDD (19).
Apart from myeloma, MIDD may complicate Waldenström’s In some patients with HCDD, a monoclonal component
macroglobulinemia and chronic lymphocytic leukemia in rare cannot be detected in serum and urine (8). In other patients, a
cases (17). It often occurs in the absence of a detectable malignant monoclonal IgG1␭ can be found in serum, but no deletion is
process, even after prolonged (⬎10 yr) follow-up. In such “pri- found in the HC (9). Identification of the nephritogenic deleted
mary” forms, a monoclonal bone marrow plasma cell population HC that circulates in low amounts then requires serum frac-
can be documented easily by immunofluorescence examination. tionation followed by Western blotting (9) (Figure 4). This
finding suggests that serum fractionation also should be per-
Diagnostic Investigation formed in patients with LCDD in whom usual immunochemi-
The diagnosis of MIDD must be suspected in any patient cal methods have failed to detect a circulating monoclonal
who has nephrotic syndrome or rapidly progressive tubuloin- component.
1564 Journal of the American Society of Nephrology J Am Soc Nephrol 12: 1558 –1565, 2001

treat patients with steroids plus melphalan or a cytotoxic agent,


irrespective of the accompanying hematologic disease.
Whether appropriate treatment can result in sustained remission
has long remained unclear. Clearance of the LC deposits has been
demonstrated unequivocally in some patients after intensive che-
motherapy with syngeneic bone marrow transplantation or blood
stem cell autografting (41,42). Disappearance of nodular mesan-
gial lesions and LC deposits also was reported after long-term
chemotherapy (43). These observations are of paramount impor-
tance: they demonstrate that fibrotic nodular glomerular lesions
are reversible, and they argue for intensive chemotherapy in
patients with severe visceral involvement.
Kidney transplantation has been performed in a few patients
with MIDD and end-stage renal failure. Recurrence of the
disease usually is observed. Therefore, intensive chemotherapy
should be performed before kidney transplantation.
In conclusion, MIDD is a rare systemic disease that is
characterized by severe renal failure as a result of the deposi-
tion of a monotypic LC and/or HC of Ig. Glomerular lesions
are so similar to diabetic nephropathy that MIDD may serve as
a model for the understanding of this plague of the third
millennium. MIDD indeed is the only sclerotic glomerular
disease in which the offending molecule is defined perfectly.
Figure 4. Immunoblotting study after agarose nondenaturing electro- As in AL amyloidosis, controlled trials are required to define
phoresis of a 5S Sephadex-G200 fraction from the serum of patients the best chemotherapy combination according to clinical pre-
A and B. Blots were revealed with monoclonal antibodies of Ig sentation and severity of renal failure.
domain specificities as indicated. Note that both fractions contained a
monoclonal IgG1␭ exhibiting normal reactivity with anti-␥ constant
domain antibodies (including the two anti-␥CH1 tested) and an iso-
Acknowledgments
We thank Catherine Bazaud for excellent secretarial assistance.
lated ␥ chain of IgG1 subclass (reactivity with NL16) lacking CH1
This work was supported by a grant from the Programme Hospitalier
domain epitopes (arrow). The fraction from the serum of patient A
de Recherche Clinique (AOM 96058).
also included a free ␭ LC (from Moulin et al., J Am Soc Nephrol 10:
526, 1999; reprinted with permission of the publisher).
References
1. Buxbaum JN, Chuba JV, Hellman GC, Solomon A, Gallo GR:
Monoclonal immunoglobulin deposition disease: Light chain and
Outcome and Treatment light and heavy chain deposition diseases and their relation to
The outcome of MIDD remains uncertain, mainly because light chain amyloidosis. Clinical features, immunopathology,
extrarenal deposits of LC can be totally asymptomatic or cause and molecular analysis. Ann Intern Med 112: 455– 464, 1990
severe organ damage that leads to death. Survival from onset of 2. Preud’homme JL, Aucouturier P, Touchard G, Striker L, Kham-
lichi AA, Rocca A, Denoroy L, Cogne M: Monoclonal immu-
symptoms varies from 1 mo to 10 yr, whereas by comparison,
noglobulin deposition disease (Randall type). Relationship with
the prognosis of a related disease, AL amyloidosis, is much
structural abnormalities of immunoglobulin chains. Kidney Int
more homogeneous with a median survival time of 18 mo 46: 965–972, 1994
under chemotherapy (40). Although renal prognosis is poor, 3. Kobernick SD, Whiteside JH: Renal glomeruli in multiple my-
patient survival can be considerable with 70 and 37% 5-yr eloma. Lab Invest 6: 478 – 485, 1957
patient survival and renal survival, respectively (36). The only 4. Sanchez LM, Domz CA: Renal patterns in myeloma. Ann Intern
predictor of renal patient survival seems to be the initial serum Med 52: 44 –54, 1960
creatinine at the time of biopsy, whereas the presence of 5. Antonovych TT, Lin RC, Parrish E, Mostofi K: Light chain deposits
multiple myeloma does not seem to influence renal or patient in multiple myeloma [Abstract]. Lab Invest 30: 370A, 1974
survival (19). Outcomes in terms of renal and patient survival 6. Randall RE, Williamson WC Jr, Mullinax F, Tung MY, Still WJ:
are significantly better in patients with pure MIDD, compared Manifestations of systemic light chain deposition. Am J Med 60:
293–299, 1976
with those who present with myeloma cast nephropathy (19).
7. Preud’homme JL, Morel-Maroger L, Brouet JC, Mihaesco E,
As in AL amyloidosis, treatment should be aimed at reduc- Mery JP, Seligmann M: Synthesis of abnormal heavy and light
ing Ig production. Chemotherapy is logical in patients with MIDD chains in multiple myeloma with visceral deposition of mono-
and myeloma. It is controversial in the absence of overt malig- clonal immunoglobulin. Clin Exp Immunol 42: 545–553, 1980
nancy given the uncertain outcome of LCDD and the absence of 8. Aucouturier P, Khamlichi AA, Touchard G, Justrabo E, Cogne
reliable follow-up criteria, especially in patients without detect- M, Chauffert B, Martin F, Preud’homme JL: Brief report: Heavy-
able M component. However, it has become general practice to chain deposition disease. N Engl J Med 329: 1389 –1393, 1993
J Am Soc Nephrol 12: 1558 –1565, 2001 Light Chain Deposition Disease 1565

9. Moulin B, Deret S, Mariette X, Kourilsky O, Imai H, Dupouet L, G, Ponticelli C, Dordrecht, Kluwer Academic Publishers, 1988,
Marcellin L, Kolb I, Aucouturier P, Brouet JC, Ronco PM, pp 171–181
Mougenot B: Nodular glomerulosclerosis with deposition of 26. Zhu L, Herrera GA, Murphy-Ullrich JE, Huang ZQ, Sanders
monoclonal immunoglobulin heavy chains lacking CH1. J Am PW: Pathogenesis of glomerulosclerosis in light chain deposition
Soc Nephrol 10: 519 –528, 1999 disease. Role for transforming growth factor-␤. Am J Pathol 147:
10. Denoroy L, Déret S, Aucouturier P: Overrepresentation of the 375–385, 1995
V␬IV subgroup in light chain deposition disease. Immunol Lett 27. Herrera GA, Russell WJ, Isaac J, Turbat-Herrera EA, Tagouri
42: 63– 66, 1994 YM, Sanders PW, Picken MM, Dempsey S: Glomerulopathic
11. Khamlichi AA, Rocca A, Touchard G, Aucouturier P, light chain-mesangial cell interactions modulate in vitro extra-
Preud’homme JL, Cogne M: Role of light chain variable region cellular matrix remodeling and reproduce mesangiopathic find-
in myeloma with light chain deposition disease: Evidence from ings documented in vivo. Ultrastruct Pathol 23: 107–126, 1999
an experimental model. Blood 86: 3655–3659, 1995 28. Sanders PW, Herrera GA, Kirk KA, Old CW, Galla JH: Spec-
12. Khamlichi AA, Aucouturier P, Silvain C, Bauwens M, Touchard trum of glomerular and tubulointerstitial renal lesions associated
G, Preud’homme JL, Nau F, Cogne M: Primary structure of a with monotypical immunoglobulin light chain deposition. Lab
monoclonal ␬ chain in myeloma with light chain deposition Invest 64: 527–537, 1991
disease. Clin Exp Immunol 87: 122–126, 1992 29. Ganeval D: Visceral deposition of monoclonal light chains and
13. Rocca A, Khamlichi AA, Aucouturier P, Noel LH, Denoroy L, immunoglobulins: A study of renal and immunopathologic ab-
Preud’homme JL, Cogne M: Primary structure of a variable normalities. Adv Nephrol Necker Hosp 11: 25, 1982
region of the V␬l subgroup (ISE) in light chain deposition 30. Lomé C, Beaufils H, Mougenot B, Noël LH, Droz D: Non-amyloid
disease. Clin Exp Immunol 91: 506 –509, 1993 monoclonal immunoglobulin light-chain (LC) deposits in renal ex-
14. Bellotti V, Stoppini M, Merlini G, Zapponi MC, Meloni ML, tracellular matrix [Abstract]. J Am Soc Nephrol 8: 539A, 1997
Banfi G, Ferri G: Amino acid sequence of k Sci, the Bence Jones 31. Pirani CL, Silva F, D’Agati V, Chander P, Striker L: Renal
protein isolated from a patient with light chain deposition dis- lesions in plasma cell dyscrasias: Ultrastructural observations.
ease. Biochim Biophys Acta 1097: 177–182, 1991 Am J Kidney Dis 10: 208 –221, 1987
15. Deret S, Chomilier J, Huang DB, Preud’homme JL, Stevens FJ, 32. Bruneval P, Foidart JM, Nochy D, Camilleri JP, Bariety J:
Aucouturier P: Molecular modeling of immunoglobulin light Glomerular matrix proteins in nodular glomerulosclerosis in as-
chains implicates hydrophobic residues in non-amyloid light sociation with light chain deposition disease and diabetes melli-
chain deposition disease. Protein Eng 10: 1191–1197, 1997 tus. Hum Pathol 16: 477– 484, 1985
16. Ganeval D, Noël LH, Preud’homme JL, Droz D, Grünfeld JP: 33. Stokes MB, Holler S, Cui Y, Hudkins KL, Eitner F, Fogo A,
Light-chain deposition disease: Its relation with AL-type amy- Alpers CE: Expression of decorin, biglycan, and collagen type I
loidosis. Kidney Int 26: 1–9, 1984 in human renal fibrosing disease. Kidney Int 57: 487– 498, 2000
17. Preud’homme JL, Morel-Maroger L, Brouet JC, Cerf M, Mignon 34. Herrera GA, Shultz JJ, Soong SJ, Sanders PW: Growth factors in
F, Guglielmi P, Seligmann M: Synthesis of abnormal immuno- monoclonal light-chain-related renal diseases. Hum Pathol 25:
globulins in lymphoplasmacytic disorders with visceral light 883– 892, 1994
chain deposition. Am J Med 69: 703–710, 1980 35. Tubbs RR, Gephardt GN, McMahon JT, Hall PM, Valenzuela R,
18. Cogné M, Preud’homme JL, Bauwens M, Touchard G, Aucou- Vidt DG: Light chain nephropathy. Am J Med 71: 263–269, 1981
turier P: Structure of a monoclonal kappa chain of the V␬IV 36. Heilman RL, Velosa JA, Holley KE, Offord KP, Kyle RA: Long-
subgroup in the kidney and plasma cells in light chain deposition term follow-up and response to chemotherapy in patients with
disease. J Clin Invest 87: 2186 –2190, 1991 light-chain deposition disease. Am J Kidney Dis 20: 34 – 41, 1992
19. Lin J, Markowitz GS, Valeri AM, Kambham N, Sherman WH, 37. Pozzi C, Fogazzi GB, Banfi G, Strom EH, Ponticelli C, Locatelli
Appel GB, D’Agati VD: Renal monoclonal immunoglobulin F: Renal disease and patient survival in light chain deposition
deposition disease: The disease spectrum. J Am Soc Nephrol, 12: disease. Clin Nephrol 43: 281–287, 1995
1482–1492, 2001 38. Droz D, Noel LH, Carnot F, Degos F, Ganeval D, Grunfeld JP:
20. Ronco PM, Aucouturier P, Mougenot B: Monoclonal gammopa- Liver involvement in nonamyloid light chain deposits disease.
thies: Multiple myeloma, amyloidosis, and related disorders. In: Lab Invest 50: 683– 689, 1984
Diseases of the Kidney, 7th Ed., edited by Schrier RM, Philadel- 39. Ivanyi B: Frequency of light chain deposition nephropathy rel-
phia, Lippincott Williams & Wilkins, in press ative to renal amyloidosis and Bence Jones cast nephropathy in
21. Cheng IK, Ho SK, Chan DT, Ng WK, Chan KW: Crescentic a necropsy study of patients with myeloma. Arch Pathol Lab
nodular glomerulosclerosis secondary to truncated immunoglobulin Med 114: 986 –987, 1990
alpha heavy chain deposition. Am J Kidney Dis 28: 283–288, 1996 40. Kyle RA: A trial of three regimens for primary amyloidosis:
22. Khamlichi AA, Aucouturier P, Preud’homme JL, Cogné M: Colchicine alone, melphalan and prednisone, and melphalan,
Structure of abnormal heavy chains in human heavy-chain-dep- prednisone, and colchicine. N Engl J Med 336: 1202–1207, 1997
osition disease. Eur J Biochem 229: 54 – 60, 1995 41. Barjon P: Traitement de la maladie par dépôts de chaı̂nes légères
23. Eulitz M, Weiss DT, Solomon A: Immunoglobulin heavy-chain- par greffe de moelle. Nephrologie 13: 24, 1992
associated amyloidosis. Proc Natl Acad Sci USA 87: 6542– 6546, 42. Mariette X, Clauvel JP, Brouet JC: Intensive therapy in AL
1990 amyloidosis and light-chain deposition disease. Ann Intern Med
24. Solomon A, Weiss DT, Kattine AA: Nephrotoxic potential of 123: 553, 1995
Bence Jones proteins. N Engl J Med 324: 1845–1851, 1991 43. Komatsuda A, Wakui H, Ohtani H, Kodama T, Miki K, Imai H,
25. Gallo G, Buxbaum J: Monoclonal immunoglobulin deposition Miura A: Disappearance of nodular mesangial lesions in a patient
disease: Immunopathologic aspects of renal involvement. In: The with light chain nephropathy after long-term chemotherapy. Am
Kidney in Plasma Cell Dyscrasias, edited by Minetti L, D’Amico J Kidney Dis 35: 9E, 2000

You might also like