Nature medicine



Yüklə 85,66 Kb.
Pdf görüntüsü
tarix18.05.2018
ölçüsü85,66 Kb.
#44586


NATURE MEDICINE • VOLUME 7 • NUMBER 3 • MARCH 2001

365


A RT I C L E S  

Myasthenia gravis (MG) is an antibody-mediated autoimmune

disease of the neuromuscular junction. In approximately 80% of

patients, auto-antibodies to the muscle nicotinic acetylcholine

receptor (AChR) are present

1

. These antibodies cause loss of

AChR numbers and function, and lead to failure of neuromuscu-

lar transmission with muscle weakness

2

. The pathogenic mecha-

nisms acting in the 20% of patients with generalized MG who

are seronegative for AChR-antibodies (AChR-Ab)

3

have not been

elucidated, but there is evidence that they also have an anti-

body-mediated disorder

4,5

, with the antibodies directed towards

another, previously unidentified muscle-surface–membrane tar-

get

6–8

. Here we show that 70% of AChR-Ab–seronegative MG pa-

tients, but not AChR-Ab–seropositive MG patients, have serum

auto-antibodies against the muscle-specific receptor tyrosine ki-

nase, MuSK. MuSK mediates the agrin-induced clustering of

AChRs during synapse formation, and is also expressed at the

mature neuromuscular junction

9–12

. The MuSK antibodies were

specific for the extracellular domains of MuSK expressed in

transfected COS7 cells and strongly inhibited MuSK function in

cultured myotubes. Our results indicate the involvement of

MuSK antibodies in the pathogenesis of AChR-Ab–seronegative

MG, thus defining two immunologically distinct forms of the

disease. Measurement of MuSK antibodies will substantially aid

diagnosis and clinical management.

We first tested coded plasmas from AChR-Ab–seronegative MG

patients and healthy individuals using COS7 cells transfected

with rat MuSK constructs (Fig. 1a). IgG from all five AChR-

Ab–seronegative MG plasmas (Fig. 1b, AChR-Ab–neg/MuSK), but

not from the three healthy control plasmas, bound to MuSK ag-

gregates on the cell surface at dilutions of up to 1:1000. The pat-

tern of immunoreactivity was indistinguishable from that

observed with polyclonal antibodies raised against recombinant

MuSK in rabbits

13

. Each of the AChR-Ab–seronegative MG plas-



mas recognized the extracellular domains of MuSK, as we ob-

served no immunoreactivity with COS7 cells expressing the

transmembrane and cytoplasmic domains only (Fig. 1b, AChR-

Ab–neg/MuSK

Ig1-4).


Immunoprecipitation experiments confirmed that the IgG an-

tibodies in AChR-Ab–seronegative MG plasmas recognized the

native MuSK protein. As a source of MuSK, we used detergent ex-

tracts from either MuSK-expressing COS7 cells or mouse C2C12

myotubes that express functional MuSK

10,11,13


. In each case, after

the plasma incubations, we precipitated the IgG antibodies with

immobilized G protein and ran the precipitates on an SDS-PAGE.

We used a rabbit serum against MuSK as a positive control.

Antibodies from the AChR-Ab–seronegative plasmas immuno-

precipitated bands of 110 kD that were identified as MuSK by

binding of a specific antibody against MuSK (Fig 1c). MuSK was

not immunoprecipitated by healthy control plasmas, AChR-

Ab–seropositive plasmas or from an AChR-Ab–seronegative

plasma that had been pre-absorbed with the extracellular do-

main of MuSK (Ig1-4).

Currently, AChR-Ab–seronegative MG is thought to represent

about 10–20% of all MG patients

3

, but the true prevalence is dif-



ficult to assess because of differences in patient ascertainment

and referral to specialist centers. Most AChR-Ab–seropositive

MG patients present in adult life, often after the age of 50 years

14

,



but children with MG are frequently AChR-Ab–seronegative

3

. To



establish an assay that could be used for future diagnosis and epi-

demiological studies, we tested sera and plasmas in an ELISA

using plates coated with fragments of the extracellular domains

of MuSK, expressed in secreted form from COS7 cells. We calcu-

lated a cut-off (0.08 optical density units [OD]) on the basis of

the mean 

±

3 s.d. of the values with healthy control plasmas or



sera. Raised levels of IgG antibodies to MuSK were found in

17/24 samples from patients with AChR-Ab–seronegative gener-

alised MG, including the 2 children who had presented before

the age of 10 years. Titers were borderline or in the control range

in 19 AChR-Ab–seropositive MG patients without thymoma,

and in 20 with thymoma, as well as in 33 sera from patients with

other neurological disorders (Fig. 2a). The lack of detectable

MuSK antibodies in MG patients with thymoma is significant be-

cause these patients frequently have antibodies against a range

of muscle proteins

15

. Detection of MuSK antibodies will provide a



new and much needed tool for the diagnosis of AChR-

Ab–seronegative MG in children and in adults and facilitate epi-

demiological studies. It will also strengthen the case for

immunosuppressive therapy and provide a means of monitoring

its effectiveness. Importantly, the thymus gland usually appears

normal in AChR-Ab–seronegative MG (ref. 16), indicating that

thymectomy (often used therapeutically in AChR-Ab–seroposi-

tive MG) may not be appropriate in those cases with MuSK anti-

bodies. Also, the presence of MuSK antibodies should help to

exclude a thymoma.

We noted a broad correlation between optical density values

Auto-antibodies to the receptor tyrosine kinase MuSK in

patients with myasthenia gravis without acetylcholine

receptor antibodies

W

ERNER


H

OCH


1

, J


OHN

M

C



C

ONVILLE


2

, S


IGRUN

H

ELMS



1

, J


OHN

N

EWSOM



-D

AVIS


2

A



RTHUR

M

ELMS



3

& A


NGELA

V

INCENT



2

1

Max Planck Institute for Developmental Biology, Tübingen, Germany

2

Neurosciences Group, Institute of Molecular Medicine, John Radcliffe Hospital, Oxford, UK

3

Department of Neurology, University of Tübingen, Tübingen, Germany

Correspondence should be addressed to W.H.; email: werner.hoch@tuebingen.mpg.de

or A.V.; email: angela.vincent@imm.ox.ac.uk

©

2001 Nature Pub

lishing Gr

oup  http://medicine

.nature

.com

© 2001 Nature Publishing Group  http://medicine.nature.com


366

NATURE MEDICINE • VOLUME 7 • NUMBER 3 • MARCH 2001

A RT I C L E S

for IgG binding to MuSK and IgG binding to the human TE671

cell line measured in an earlier study

8

(r



2

= 0.72; < 0.001; =

12), indicating that the previously defined, but unidentified,

cell-surface antigen on TE671 cells might be MuSK. The patients

who were negative for both MuSK and AChR antibodies were not

clinically distinguishable from those who had MuSK antibodies,

but 4 of 7 MuSK-negative samples were from patients who had

received immunosuppressive treatments, compared with only 2

of 17 of the MuSK-positive samples. This indicates that the cur-

rent assay may lack sensitivity for patients who are already im-

munosuppressed. Further analysis showed that most of the

MuSK antibodies (Fig. 2b) were directed against the amino-ter-

minal sequences (construct Ig1-2 in Fig. 1a) rather than the

membrane-proximal half (construct Ig3-4 in Fig. 1a). Overall,

there is 94% homology between rat MuSK and the published

human sequence

9

, and they differ by only two amino acids in



the Ig1-2 construct. Nonetheless, use of human MuSK, as well as

further refinement of the assay, may increase the sensitivity of

the test. In addition, as we did not detect IgM antibodies to

MuSK (data not shown), the target for the putative non-IgG anti-

bodies in some patients

6

, including two of the MuSK-Ab–nega-



tive patients studied here, requires further investigation.

These data indicate that the MuSK antibodies are directed

against extracellular determinants and could therefore be di-

rectly involved in the pathogenesis of the disease. To determine

whether the antibodies can interfere with MuSK function, we ex-

amined agrin-induced AChR clustering in myotubes derived

from the mouse cell line, C2C12. In the absence of agrin (Fig. 3a,

upper panels), very few spontaneous clusters of AChRs were

found in the presence of healthy control plasma, whereas MuSK-

Ab–positive MG plasma induced some AChR aggregates along

the surface of the myotubes. Similar results have previously been

reported for rabbit antibodies raised against purified MuSK (ref.

13). When agrin was added together with the plasmas (Fig. 3a,

lower panels), the marked agrin-induced clustering which oc-

curred in the presence of healthy control plasma was not seen in

the presence of MuSK-Ab–positive MG plasma, indicating that

the MuSK antibodies had inhibited the agrin-induced AChR clus-

tering. A quantitative analysis of the data revealed that both the

clustering (Fig. 3b), and the inhibitory activity (Fig. 3c), were

found with MuSK-Ab–positive plasmas or IgG, but not with the

one MuSK-Ab–seronegative preparation or the healthy controls

tested. Moreover, both the clustering (Fig. 3b) and inhibitory

(Fig. 3c) activity of an AChR-Ab–negative plasma were prevented

by absorption with soluble MuSK, demonstrating that the MuSK

antibodies were responsible for these effects. By contrast, plas-

mas from two patients with AChR-Ab–seropositive MG (Fig. 3c),

and the non-IgG fractions of MuSK-Ab–positive MG patients

a

b

c

Fig. 1

Antibodies from AChR-Ab–seronegative MG patients bind to MuSK.



a, Domain structure of the MuSK molecule and schematic view of constructs

used in Fig. 1and and Fig. 2. b, AChR-Ab–negative MG IgGs bound to

COS7 cells expressing full-length MuSK (AChR-Ab–neg/MuSK), whereas

healthy control IgG did not (Healthy control/MuSK). MuSK immunoreactivity

appeared as a speckled pattern, similar to that seen previously with rabbit

anti-MuSK antibodies

13

. Non-transfected cells in the same field, demonstrated



by phase contrast microscopy (arrows), showed non-specific binding only.

No specific binding of AChR-Ab–seronegative MG IgG to cells expressing

MuSK lacking the extracellular domains (AChR-Ab–neg/MuSK 

Ig1-4) was



detected. c, IgG from 2 AChR-Ab–seronegative MG plasmas (Neg), but not

from control plasmas (HC), precipitated MuSK from detergent extracts of

COS7 cells expressing MuSK, and from C2C12 (C2) myotubes. The same

band was immunoprecipitated by IgG from a rabbit immunized against

MuSK (Rb). MuSK was not precipitated by AChR-Ab–seropositive MG plasmas

(Pos) or after absorption of AChR-Ab–seronegative plasma (Abs) with soluble

MuSK Ig1-4. MuSK appears as a 110 kD band from COS-cells and as several

bands representing different MuSK splice variants in the C2C12 cells

11

.

HC



Neg

Pos


Thy

ON

-1



0

1

2



3

OD (A


492

)

0.25



0.75

0.00


0.50

1.00


Dilution of plasma

 (1:6 serial dilutions)

0

1

2



3

4

5



6

Fig. 2

MuSK antibodies can be detected by ELISA. a, Antibodies to MuSK

were found in 17 of 24 AChR-Ab–seronegative MG patients (Neg) com-

pared with 13 healthy controls (HC). Negative or borderline values were

found in anti-AChR–seropositive MG patients (Pos), including those with

thymomas (Thy), and in patients with other neurological disorders (ON). b,

Titration of one AChR-Ab–seronegative MG plasma against different do-

mains of MuSK. The antibodies bound strongly to MuSK constructs express-

ing the N-terminal domains, Ig1-4 (

˾) and Ig1-2 (̆; see Fig. 1a), but

binding to the membrane-proximal Ig3-4 (

b) domains was similar to that

of a healthy control (

ć).


a

b

OD (A


492

)

©



2001 Nature Pub

lishing Gr

oup  http://medicine

.nature

.com

© 2001 Nature Publishing Group  http://medicine.nature.com


NATURE MEDICINE • VOLUME 7 • NUMBER 3 • MARCH 2001

367


A RT I C L E S  

(data not shown) did not affect agrin-induced clustering.

We show that AChR-Ab–seronegative IgG preparations con-

taining MuSK antibodies interfere with the agrin/MuSK/AChR

clustering pathway in myotubes and have the potential to alter

MuSK function at the adult neuromuscular junction. As agrin

does not appear to bind directly to MuSK, but via a hypothetical

agrin-binding component called MASC (refs. 10,12), we specu-

late that the antibodies in AChR-Ab–seronegative patients bind

to MuSK in such a manner as to prevent its interaction with

MASC. This interaction is known to depend on the N-terminal

half of the extracellular domains of MuSK (ref. 17) that we find

to be the main target for the IgG antibodies in AChR-

Ab–seronegative patients (Fig. 2b). How these antibodies cause

seronegative MG is not yet clear. AChR-Ab–seronegative MG is

mediated by humoral IgG antibodies that lead to a defect in neu-

romuscular transmission

4

. A few limited human biopsy stud-



ies

18,19


indicate that there is a reduction in AChR function at the

neuromuscular junction, although total AChR numbers were

variable. We propose that interference with MuSK function at

the mature neuromuscular junction is one pathogenic mecha-

nism in MuSK-Ab–seropositive MG patients, leading secondarily

to reduced numbers and/or altered distribution of AChRs and

other postsynaptic proteins. By analogy with AChR-Ab–positive

MG (ref. 2), MuSK antibodies may not only inhibit MuSK func-

tion directly but also increase the turnover of MuSK, further re-

ducing its activity. In addition, there could be

complement-mediated damage to the AChR-containing postsy-

naptic membrane following IgG binding to MuSK and comple-

ment activation. Some evidence for complement deposition at

the endplates of muscle from AChR-Ab–negative MG patients

has been reported

20

. Elucidating the contribution of different



mechanisms to the defect in neuromuscular transmission will re-

quire further in vitro and in vivo studies.

We have defined a novel specific target antigen, the receptor

tyrosine kinase MuSK, for the auto-antibodies in AChR-

Ab–seronegative MG. The antibodies have functional effects on

agrin-induced AChR distribution in cultured myotubes, indicat-

ing that they could interfere with AChR numbers or distribution

in mature muscle. The IgG antibodies were found in 70% of

AChR-Ab–seronegative MG patients and not in AChR-

Ab–seropositive MG patients, clearly distinguishing the two

forms of the disease. Moreover, the antibodies can be detected

by a simple ELISA that could be used for routine diagnosis and

clinical management. Our results add a receptor tyrosine kinase

(RTK) to the AChR and voltage-gated calcium and potassium

channels that are already defined targets for antibody-mediated

disorders at the neuromuscular junction

21

. Auto-antibodies to



other RTKs have occasionally been implicated in some forms of

endocrine disease

22

. Since RTKs are cell-surface molecules, regu-



lating intracellular functions and activated by dimerization, they

are attractive candidate antigens and should be considered in

other antibody-mediated disorders. Moreover, as members of the

RTK family are mutated in inherited diseases

23

, MuSK may be in-



volved not only in this acquired autoimmune disorder but also

in congenital muscle diseases.



Methods

Patients. Samples were obtained from 24 patients (18 female, 6 male) with

moderate or severe generalized MG in whom the standard radio-immuno-

precipitation assay for anti-AChR antibodies

24

was negative on several occa-



sions. All had typical fatigable muscle weakness. The diagnosis was

confirmed by electromyographic evidence of a defect in neuromuscular

transmission (a decrement of more than 10% in the amplitude of the com-

pound muscle action potential on repetitive nerve stimulation at 3 Hz and/or

an increase in jitter on single fiber studies), or by a positive response to anti-

cholinesterase medication (edrophonium or pyridostigmine). The age at

onset was 2–68 years (median 24) and the duration of symptoms at sam-

pling was between 1 month and 13 years (median 1.0 year). In 18 cases,

plasma was obtained during therapeutic plasmapheresis, which improved

muscle strength. The remaining 6 samples were sera taken on first examina-

tion. Six of the patients had received corticosteroids for up to two months

before sampling. Sera or plasmas were also obtained from healthy volun-

teers, from patients with AChR-Ab–positive MG, and from patients with

other immune-mediated neurological disorders. IgG preparations were

made using a ImmunoPure (G) IgG purification kit (Pierce, Rockford, IL)

MuSK and agrin expression constructs. Constructs encoding full-length

MuSK (ref. 13) and the soluble fragment s-agrin (4/19)

25

have been de-



scribed. MuSK deletion fragments comprising the entire extracellular do-

main (Ig1-4; aa 1–490)

9

, or the first half containing two Ig-domains (Ig1-2;



aa 1–231), were generated by insertion of artificial stop signals at these po-

sitions by a PCR approach. N-terminal fragments of MuSK comprising the

membrane-proximal extracellular domains, including Ig-domains 3 and 4

(Ig3-4; aa 203–490), or the transmembrane region and intracellular domain

(MuSK–Ig1-4, aa 491–868) were generated. The corresponding cDNA-frag-

ments, including a newly introduced SphI site, were linked to a vector con-

taining an artificial signal sequence followed by 6 histidines and a 10-aa

epitope-tag

25

. All constructs were transiently transfected into COS7 cells



11

.

For the production of soluble agrin and MuSK constructs, cells were



switched to serum-free medium the second day after transfection.

- Ag


+ Ag

1 1 2 2 3 3 4 4 5 5 6 6

HC

HC

Abs



0

5

10



15

20

25



30

35

40



- Agrin

Number of clusters/ field

- Ag

+ Ag


1 1 2 2 3 3 4 4 5 5 6 6

HC HC Abs

Pos

Pos


0

5

10



15

20

25



30

35

40



+ Agrin

Number of clusters/ field



a

b

c

Fig. 3

MuSK IgG antibodies induce AChR clusters but inhibit agrin-in-

duced clustering. a, In the absence of agrin, a moderate number of AChR

clusters, visualised by rhodamine-

α

-bungarotoxin, were induced in the



presence of MuSK-Ab–positive MG plasma compared with that in healthy

control plasma. Conversely, in the presence of agrin, large numbers of clus-

ters were induced in healthy control plasma-treated myotubes, but not in

myotubes treated with MuSK-Ab–positive MG plasma. and c, The AChR

clusters without (b) or with (c) added agrin (Ag) in cultures treated with

plasma (


í) or IgG ( ). Only the MuSK-Ab–positive plasmas and IgG prepa-

rations (1–5) affected AChR clusters; there was no effect of a MuSK-Ab–neg-

ative plasma (6), of 2 healthy control plasmas (HC), or of 1

MuSK-Ab–positive plasma after absorption with MuSK (Abs). Two AChR-

Ab–seropositive samples (Pos) did not affect agrin-induced clustering.

Clusters in control cultures without plasma or IgG (

२). 

©

2001 Nature Pub

lishing Gr

oup  http://medicine

.nature

.com

© 2001 Nature Publishing Group  http://medicine.nature.com



368

NATURE MEDICINE • VOLUME 7 • NUMBER 3 • MARCH 2001

A RT I C L E S

Conditioned media, containing MuSK or agrin fragments, were removed

24 hours later and analyzed by western blotting to confirm expression.

Immunostaining of MuSK-transfected COS7 cells. COS7 cells were plated

onto chamber slides the day after transfection. Two days later, cells were

fixed with 2% paraformaldehyde and stained as described

13

. Plasmas of MG



patients and controls were analyzed at various dilutions (between 1:20 and

1:5000). Bound antibodies were visualized with secondary antibodies conju-

gated to Cy3 (anti-human IgG, Dianova, Hamburg, Germany). In all experi-

ments, expression of transfected MuSK constructs was confirmed by staining

parallel slides with rabbit antibodies against MuSK (ref. 13).

Immunoprecipitation of MuSK. Detergent extracts were prepared from

MuSK-transfected COS7 cells or from C2C12 myotubes that had been

fused for 5 days

11

. The immunoprecipitation was performed as de-



scribed

11,13


. AChR-Ab–seronegative MG, AChR-Ab–seropositive MG plasmas

and control plasmas were incubated with the extracts at 1:20. Rabbit anti-

MuSK serum was used at 1:100. MuSK in the immunoprecipitates was ana-

lyzed by western blotting using affinity-purified serum antibodies directed

against the MuSK cytoplasmic sequence

13

. One AChR-Ab–seronegative MG



plasma was absorbed with soluble MuSK, by addition of an equal volume of

conditioned medium of COS7 cells transfected with the soluble MuSK frag-

ment Ig1-4, for 2 hours. The mixture was then diluted further before use.

ELISA detection of antibodies to MuSK. Conditioned medium from

MuSK-transfected COS7 cells or from control cells mock-transfected with

salmon sperm DNA, was diluted 1:1 with 100 mM NaHCO3 buffer (pH 9.5)

and applied overnight to ELISA plates. Plasmas were first tested at 1:5 in

triplicates and subsequently at 1:10 in duplicates. Bound antibodies were

detected by horseradish peroxidase-protein A (Amersham, Braunschweig,

Germany) followed by o-phenylenediamine and measuring A

492


. For each

sample, nonspecific immunoreactivity, determined by incubation of plates

coated with conditioned medium from mock-transfected COS7 cells, was

subtracted. The efficient immobilization of deletion fragments of MuSK was

confirmed by ELISA with an antibody directed against the HA-epitope.

AChR aggregation assay. The mouse muscle cell line, C2C12, was used to

determine functional effects of antibodies. Cells were plated onto chamber

slides, fused and treated with or without agrin and/or plasmas or IgGs for 5

hours


13

. After fixation, AChRs were visualised with rhodamine-

α

-bungaro-



toxin and the number of aggregates from more than 20 microscopic fields,

and at least 2 independent cultures, were measured as described

25

. Control



experiments showed that the MuSK fragment, used for preincubation of

some plasmas, did not inhibit AChR aggregation in the final concentration

present in the assay. Results are expressed as mean 

±

s.e.m.



Acknowledgments

We thank U. Schwarz for his support; C. Hopf for participating in cloning some

of the constructs; A. Evoli and S. Robb for two of the plasmas and clinical infor-

mation; and C.-M. Becker, C. Schuster and D. Roberts for critical reading of the

manuscript. JMcC is supported by a Wellcome Trust Research Training

Fellowship. J.N.-D. and A.V. thank the Muscular Dystrophy

Campaign/Myasthenia Gravis Association for support.

RECEIVED 2 AUGUST 2000; ACCEPTED 22 JANUARY 2001

1. Lindstrom, J., Seybold, M.E., Lennon, V.A., Whittingham, S. & Duane, D.D.

Antibody to acetylcholine receptor in myasthenia gravis: prevalence, clinical cor-

relates and diagnostic values. Neurology 26, 1054–1059 (1976).

2. Drachman, D.B. Myasthenia gravis. New Engl. J. Med. 330, 1797–1810 (1994).

3. Sanders, D.B., Andrews, I., Howard, J.F. & Massey, J.M. Seronegative myasthenia

gravis. Neurology 48, S40–S45 (1997).

4. Mossman, S., Vincent, A. & Newsom-Davis, J. Myasthenia gravis without acetyl-

choline-receptor antibody: a distinct disease entity. Lancet 1, 116–119 (1986).

5. Miers, A.K. & Havard, C.W.H. Diaphragmatic myasthenia in mother and child.

Postgrad. Med. J61, 725–727 (1985).

6. Yamamoto, T. et al. Seronegative myasthenia gravis: a plasma factor inhibiting

agonist-induced acetylcholine receptor function copurifies with IgM. Ann. Neurol.

30, 550–557 (1991).

7. Brooks, E.B., Pachner, A.R., Drachman, D.B. & Kantor, F.S. A sensitive rosetting

assay for detection of acetylcholine receptor antibodies using BC3H-1 cells: posi-

tive results in ‘antibody-negative’ myasthenia gravis. J. Neuroimmunol. 28, 83–93

(1990).

8. Blaes, F., Beeson, D., Plested, P., Lang, B. & Vincent, A. IgG from “seronegative”



myasthenia gravis patients binds to a muscle cell line, TE671, but not to human

acetylcholine receptor. Ann. Neurol. 47, 504–510 (2000).

9. Valenzuela, D.M. et al. Receptor tyrosine kinase specific for the skeletal muscle lin-

eage: expression in embryonic muscle, at the neuromuscular junction, and after

injury. Neuron 15, 573–584 (1995).

10. Glass, D.J. et al. Agrin acts via a MuSK receptor complex. Cell 85, 513–523

(1996).

11. Hopf, C. & Hoch, W. Tyrosine phosphorylation of the muscle-specific kinase is ex-



clusively induced by acetylcholine receptor-aggregating agrin fragments. Eur. J.

Biochem. 253, 382–389 (1998).

12. Sanes, J.R. & Lichtman, J.W. Development of the vertebrate neuromuscular junc-

tion. Ann. Rev. Neurosci. 22, 389–442 (1999).

13. Hopf, C. & Hoch, W. Dimerization of the muscle-specific kinase induces tyrosine

phosphorylation of acetylcholine receptors and their aggregation on the surface

of myotubes. J. Biol. Chem. 273, 6467–6473 (1998).

14. Robertson, N.P., Deans, J. & Compston, D.A. Myasthenia Gravis: a population

based epidemiological study in Cambridgeshire, England. J. Neurol. Neurosurg.



Psychiatry 65, 492–496 (1998).

15. Aarli, J.A., Skeie, G.O., Mygland, A. & Gilhus, N.E. Muscle striation antibodies in

myasthenia gravis. Diagnostic and functional significance. Ann. NY Acad. Sci. 841,

505–515 (1998).

16. Willcox, N., Schluep, M., Ritter, M.A. & Newsom-Davis, J. The thymus in seroneg-

ative myasthenia gravis patients. J. Neurol. 238, 256–261 (1991).

17. Zhou, H., Glass, D.J., Yancopoulos, G.D. & Sanes, J.R. Distinct domains of MuSK

mediate its ability to induce and to associate with postsynaptic specializations. J.



Cell Biol. 146, 1133–1146 (1999).

18. Drachman, D.B., De Silva, S., Ramsay, D. & Pestronk, A. Humoral pathogenesis of

myathenia gravis. Ann. NY Acad. Sci. 505, 90–105 (1987).

19. Vincent, A. et al. Seronegative myasthenia gravis. Ann. NY Acad. Sci. 681,

529–538 (1993).

20. Nakano, S. & Engel, A.G. Myasthenia gravis: quantitative immunocytochemical

analysis of inflammatory cells and detection of complement membrane attack

complex at the end-plate in 30 patients. Neurology 43, 1167–1172 (1993).

21. Newsom-Davis, J. Autoantibody-mediated channelopathies at the neuromuscular

junction. The Neuroscientist 3, 337–346 (1997).

22. Taylor, S.I., Barbetti, F., Accili, D., Roth, J. & Gorden, P. Syndromes of autoimmu-

nity and hypoglycaemia. Autoantibodies directed against insulin and its receptor.



Endocrinol. Metab. Clin. North Am. 18, 123–143 (1989).

23. Robertson, S.C., Tynan, J.A. & Donoghue, D.J. RTK mutations and human syn-

dromes: when good receptors turn bad. Trends Genet. 16, 265–271 (2000).

24. Vincent, A. & Newsom-Davis, J. Acetylcholine receptor antibodies as a diagnostic

test for myasthenia gravis: results in 153 validated cases and 2967 diagnostic as-

says. J. Neurol. Neurosurg. Psychiatry 48, 1246–1252 (1985).

25. Hopf, C. & Hoch, W. Heparin inhibits acetylcholine receptor aggregation at two

distinct steps in the agrin-induced pathway. Eur. J. Neurosci. 9, 1170–1177



(1997).

©

2001 Nature Pub

lishing Gr

oup  http://medicine

.nature

.com

© 2001 Nature Publishing Group  http://medicine.nature.com

Yüklə 85,66 Kb.

Dostları ilə paylaş:




Verilənlər bazası müəlliflik hüququ ilə müdafiə olunur ©genderi.org 2024
rəhbərliyinə müraciət

    Ana səhifə