Depression is an early disease manifestation in lupus-prone MRL/lpr mice

Hua-Xin Gao, Sean R Campbell, Min-Hui Cui, Pu Zong, Jong Hee-Hwang, Maria Gulinello, Chaim Putterman, Hua-Xin Gao, Sean R Campbell, Min-Hui Cui, Pu Zong, Jong Hee-Hwang, Maria Gulinello, Chaim Putterman

Abstract

Many lupus patients develop neuropsychiatric manifestations, including cognitive dysfunction, depression, and anxiety. However, it is not clear if neuropsychiatric lupus is a primary disease manifestation, or is secondary to non-CNS disease. We found that MRL/lpr lupus-prone mice exhibited significant depression-like behavior already at 8 weeks of age, despite normal visual working memory, locomotor coordination and social preference. Moreover, depression was significantly correlated with titers of autoantibodies against DNA, NMDA receptors and cardiolipin. Our results indicate that lupus mice develop depression and CNS dysfunction very early in the course of disease, in the absence of substantial pathology involving other target organs.

Figures

Fig. 1
Fig. 1
Proteinuria and autoantibody levels in MRL/lpr and MRL/+ mice. A–D, Mouse serum autoantibody titers examined by ELISA, from age 6w to 18w. A. IgG anti-dsDNA titers. B. IgG anti-chromatin titers. C. IgG anti-NMDAR titers. D. IgG anti-cardiolipin titers. E. Urine protein concentrations in MRL/+ and MRL/lpr mice at 6–18 weeks. N=10 in each of the mice groups in this Figure. *, p

Fig. 1

Proteinuria and autoantibody levels in…

Fig. 1

Proteinuria and autoantibody levels in MRL/lpr and MRL/+ mice. A–D, Mouse serum autoantibody…

Fig. 1
Proteinuria and autoantibody levels in MRL/lpr and MRL/+ mice. A–D, Mouse serum autoantibody titers examined by ELISA, from age 6w to 18w. A. IgG anti-dsDNA titers. B. IgG anti-chromatin titers. C. IgG anti-NMDAR titers. D. IgG anti-cardiolipin titers. E. Urine protein concentrations in MRL/+ and MRL/lpr mice at 6–18 weeks. N=10 in each of the mice groups in this Figure. *, p

Fig. 2

Lupus-prone MRL/lpr mice behave normally…

Fig. 2

Lupus-prone MRL/lpr mice behave normally in the open field test. The total track…

Fig. 2
Lupus-prone MRL/lpr mice behave normally in the open field test. The total track lengths and center track lengths in the open field of 8w (A) and 18w (B) MRL/+ and MRL/lpr mice (n=10) were measured. Total time in the field: 15 minutes.

Fig. 3

Lupus-prone MRL/lpr mice display normal…

Fig. 3

Lupus-prone MRL/lpr mice display normal behavior in the balance beam walking test. 8w…

Fig. 3
Lupus-prone MRL/lpr mice display normal behavior in the balance beam walking test. 8w old MRL/+ and MRL/lpr mice were tested in beam walking for locomotor coordination. A. Number of slips recorded during beam crossing. B. Total time each group spent crossing the beam.

Fig. 4

Cognition is preserved in 8w…

Fig. 4

Cognition is preserved in 8w and 18w MRL/lpr mice. Object recognition (visual memory)…

Fig. 4
Cognition is preserved in 8w and 18w MRL/lpr mice. Object recognition (visual memory) tests on 8w (A) and 18w (B) mice were performed. A 45 minute delay time was used between the first and second object exposures. Mice having normal visual memory are expected to have >50% preference for a novel object (above the dotted line).

Fig. 5

Elevated plus maze test detects…

Fig. 5

Elevated plus maze test detects less anxiety in lupus mice than control mice.…

Fig. 5
Elevated plus maze test detects less anxiety in lupus mice than control mice. Anxiety levels were tested in an EPM test. A. Time spent in the open arms was recorded and converted to a percentage of the total time in the EPM (5 minutes). The percent time spent on exploration is shown in the graph. B. Open arm entries were recorded. C. stretched attend postures counted during EPM tests.

Fig. 6

MRL/lpr mice had similar social…

Fig. 6

MRL/lpr mice had similar social preference as MRL/+ controls. 8w (A) and 18w…

Fig. 6
MRL/lpr mice had similar social preference as MRL/+ controls. 8w (A) and 18w (B) MRL/lpr mice were tested for social activity in a Y maze. Preference was calculated based on the social target contacting time over total exploring time. Mice having normal social activities are expected to have >50% preference to a social target (above the dotted line).

Fig. 7

MRL/lpr mice display depression-like behavior…

Fig. 7

MRL/lpr mice display depression-like behavior at 8 weeks of age. The forced swim…

Fig. 7
MRL/lpr mice display depression-like behavior at 8 weeks of age. The forced swim test was used to assess depression-like behavior in mice. % floating time: total floating time converted to percentage of total test time (A, test on 8 weeks of age. C, test on 18 weeks of age). B and D, floating latency is the delay time until mice give up struggling in the water and start to show the ‘still’ floating behavior. Time was recorded in seconds. B, testing 8 week old mice. D, testing 18 week old mice. E, F. Two way ANOVA analysis of the effects of age and genotype on total floating time (E) and floating latency (F) in the tested mice.

Fig. 7

MRL/lpr mice display depression-like behavior…

Fig. 7

MRL/lpr mice display depression-like behavior at 8 weeks of age. The forced swim…

Fig. 7
MRL/lpr mice display depression-like behavior at 8 weeks of age. The forced swim test was used to assess depression-like behavior in mice. % floating time: total floating time converted to percentage of total test time (A, test on 8 weeks of age. C, test on 18 weeks of age). B and D, floating latency is the delay time until mice give up struggling in the water and start to show the ‘still’ floating behavior. Time was recorded in seconds. B, testing 8 week old mice. D, testing 18 week old mice. E, F. Two way ANOVA analysis of the effects of age and genotype on total floating time (E) and floating latency (F) in the tested mice.

Fig. 8

Lupus autoantibody levels correlate with…

Fig. 8

Lupus autoantibody levels correlate with depression behavior. Linear regression correlation of % floating…

Fig. 8
Lupus autoantibody levels correlate with depression behavior. Linear regression correlation of % floating time in forced swim test with serum anti-dsDNA titers (A, 8w; D, 18w), anti-NMDAR antibody titers (B, 8w; E, 18w), or anti-cardiolipin antibody titers (C, 8w; F, 18w) were shown. MRL/+: ■, MRL/lpr: ▼. P values and Pearson’s R values for each correlation are also shown in the graphs.

Fig. 9

MRI/MRSI analyses show differences in…

Fig. 9

MRI/MRSI analyses show differences in regional brain metabolism between control and lupus mice.…

Fig. 9
MRI/MRSI analyses show differences in regional brain metabolism between control and lupus mice. The figure illustrates the type of spectra obtained from the hippocampus (top) and thalamus (bottom), with a corresponding anatomical image of a lupus mouse brain. Data from the comparative analysis of lupus and control mouse brains is provided in Table 2.
All figures (11)
Similar articles
Cited by
References
    1. Bessa JM, Oliveira M, Cerqueira JJ, Almeida OF, Sousa N. Age-related qualitative shift in emotional behaviour: paradoxical findings after re-exposure of rats in the elevated-plus maze. Behav Brain Res. 2005;162:135–142. - PubMed
    1. Bluestein HG. The central nervous system in systemic lupus erythematosus. In: Lahita RG, editor. Systemic lupus erythematosus. Churchill Livingstone; New York: 1992. pp. 639–655.
    1. Bonfa E, Marshak-Rothstein A, Weissbach H, Brot N, Elkon K. Frequency and epitope recognition of anti-ribosome P antibodies from humans with systemic lupus erythematosus and MRL/lpr mice are similar. J Immunol. 1988;140:3434–3437. - PubMed
    1. Brey RL, Abbott RD, Curb JD, Sharp DS, Ross GW, Stallworth CL, Kittner SJ. beta(2)-Glycoprotein 1-dependent anticardiolipin antibodies and risk of ischemic stroke and myocardial infarction: the honolulu heart program. Stroke. 2001;32:1701–1706. - PubMed
    1. Brooks WM, Sabet A, Sibbitt WL, Jr, Barker PB, van Zijl PC, Duyn JH, Moonen CT. Neurochemistry of brain lesions determined by spectroscopic imaging in systemic lupus erythematosus. J Rheumatol. 1997;24:2323–2329. - PubMed
Show all 66 references
MeSH terms
[x]
Cite
Copy Download .nbib .nbib
Format: AMA APA MLA NLM
Fig. 1
Fig. 1
Proteinuria and autoantibody levels in MRL/lpr and MRL/+ mice. A–D, Mouse serum autoantibody titers examined by ELISA, from age 6w to 18w. A. IgG anti-dsDNA titers. B. IgG anti-chromatin titers. C. IgG anti-NMDAR titers. D. IgG anti-cardiolipin titers. E. Urine protein concentrations in MRL/+ and MRL/lpr mice at 6–18 weeks. N=10 in each of the mice groups in this Figure. *, p

Fig. 2

Lupus-prone MRL/lpr mice behave normally…

Fig. 2

Lupus-prone MRL/lpr mice behave normally in the open field test. The total track…

Fig. 2
Lupus-prone MRL/lpr mice behave normally in the open field test. The total track lengths and center track lengths in the open field of 8w (A) and 18w (B) MRL/+ and MRL/lpr mice (n=10) were measured. Total time in the field: 15 minutes.

Fig. 3

Lupus-prone MRL/lpr mice display normal…

Fig. 3

Lupus-prone MRL/lpr mice display normal behavior in the balance beam walking test. 8w…

Fig. 3
Lupus-prone MRL/lpr mice display normal behavior in the balance beam walking test. 8w old MRL/+ and MRL/lpr mice were tested in beam walking for locomotor coordination. A. Number of slips recorded during beam crossing. B. Total time each group spent crossing the beam.

Fig. 4

Cognition is preserved in 8w…

Fig. 4

Cognition is preserved in 8w and 18w MRL/lpr mice. Object recognition (visual memory)…

Fig. 4
Cognition is preserved in 8w and 18w MRL/lpr mice. Object recognition (visual memory) tests on 8w (A) and 18w (B) mice were performed. A 45 minute delay time was used between the first and second object exposures. Mice having normal visual memory are expected to have >50% preference for a novel object (above the dotted line).

Fig. 5

Elevated plus maze test detects…

Fig. 5

Elevated plus maze test detects less anxiety in lupus mice than control mice.…

Fig. 5
Elevated plus maze test detects less anxiety in lupus mice than control mice. Anxiety levels were tested in an EPM test. A. Time spent in the open arms was recorded and converted to a percentage of the total time in the EPM (5 minutes). The percent time spent on exploration is shown in the graph. B. Open arm entries were recorded. C. stretched attend postures counted during EPM tests.

Fig. 6

MRL/lpr mice had similar social…

Fig. 6

MRL/lpr mice had similar social preference as MRL/+ controls. 8w (A) and 18w…

Fig. 6
MRL/lpr mice had similar social preference as MRL/+ controls. 8w (A) and 18w (B) MRL/lpr mice were tested for social activity in a Y maze. Preference was calculated based on the social target contacting time over total exploring time. Mice having normal social activities are expected to have >50% preference to a social target (above the dotted line).

Fig. 7

MRL/lpr mice display depression-like behavior…

Fig. 7

MRL/lpr mice display depression-like behavior at 8 weeks of age. The forced swim…

Fig. 7
MRL/lpr mice display depression-like behavior at 8 weeks of age. The forced swim test was used to assess depression-like behavior in mice. % floating time: total floating time converted to percentage of total test time (A, test on 8 weeks of age. C, test on 18 weeks of age). B and D, floating latency is the delay time until mice give up struggling in the water and start to show the ‘still’ floating behavior. Time was recorded in seconds. B, testing 8 week old mice. D, testing 18 week old mice. E, F. Two way ANOVA analysis of the effects of age and genotype on total floating time (E) and floating latency (F) in the tested mice.

Fig. 7

MRL/lpr mice display depression-like behavior…

Fig. 7

MRL/lpr mice display depression-like behavior at 8 weeks of age. The forced swim…

Fig. 7
MRL/lpr mice display depression-like behavior at 8 weeks of age. The forced swim test was used to assess depression-like behavior in mice. % floating time: total floating time converted to percentage of total test time (A, test on 8 weeks of age. C, test on 18 weeks of age). B and D, floating latency is the delay time until mice give up struggling in the water and start to show the ‘still’ floating behavior. Time was recorded in seconds. B, testing 8 week old mice. D, testing 18 week old mice. E, F. Two way ANOVA analysis of the effects of age and genotype on total floating time (E) and floating latency (F) in the tested mice.

Fig. 8

Lupus autoantibody levels correlate with…

Fig. 8

Lupus autoantibody levels correlate with depression behavior. Linear regression correlation of % floating…

Fig. 8
Lupus autoantibody levels correlate with depression behavior. Linear regression correlation of % floating time in forced swim test with serum anti-dsDNA titers (A, 8w; D, 18w), anti-NMDAR antibody titers (B, 8w; E, 18w), or anti-cardiolipin antibody titers (C, 8w; F, 18w) were shown. MRL/+: ■, MRL/lpr: ▼. P values and Pearson’s R values for each correlation are also shown in the graphs.

Fig. 9

MRI/MRSI analyses show differences in…

Fig. 9

MRI/MRSI analyses show differences in regional brain metabolism between control and lupus mice.…

Fig. 9
MRI/MRSI analyses show differences in regional brain metabolism between control and lupus mice. The figure illustrates the type of spectra obtained from the hippocampus (top) and thalamus (bottom), with a corresponding anatomical image of a lupus mouse brain. Data from the comparative analysis of lupus and control mouse brains is provided in Table 2.
All figures (11)
Fig. 2
Fig. 2
Lupus-prone MRL/lpr mice behave normally in the open field test. The total track lengths and center track lengths in the open field of 8w (A) and 18w (B) MRL/+ and MRL/lpr mice (n=10) were measured. Total time in the field: 15 minutes.
Fig. 3
Fig. 3
Lupus-prone MRL/lpr mice display normal behavior in the balance beam walking test. 8w old MRL/+ and MRL/lpr mice were tested in beam walking for locomotor coordination. A. Number of slips recorded during beam crossing. B. Total time each group spent crossing the beam.
Fig. 4
Fig. 4
Cognition is preserved in 8w and 18w MRL/lpr mice. Object recognition (visual memory) tests on 8w (A) and 18w (B) mice were performed. A 45 minute delay time was used between the first and second object exposures. Mice having normal visual memory are expected to have >50% preference for a novel object (above the dotted line).
Fig. 5
Fig. 5
Elevated plus maze test detects less anxiety in lupus mice than control mice. Anxiety levels were tested in an EPM test. A. Time spent in the open arms was recorded and converted to a percentage of the total time in the EPM (5 minutes). The percent time spent on exploration is shown in the graph. B. Open arm entries were recorded. C. stretched attend postures counted during EPM tests.
Fig. 6
Fig. 6
MRL/lpr mice had similar social preference as MRL/+ controls. 8w (A) and 18w (B) MRL/lpr mice were tested for social activity in a Y maze. Preference was calculated based on the social target contacting time over total exploring time. Mice having normal social activities are expected to have >50% preference to a social target (above the dotted line).
Fig. 7
Fig. 7
MRL/lpr mice display depression-like behavior at 8 weeks of age. The forced swim test was used to assess depression-like behavior in mice. % floating time: total floating time converted to percentage of total test time (A, test on 8 weeks of age. C, test on 18 weeks of age). B and D, floating latency is the delay time until mice give up struggling in the water and start to show the ‘still’ floating behavior. Time was recorded in seconds. B, testing 8 week old mice. D, testing 18 week old mice. E, F. Two way ANOVA analysis of the effects of age and genotype on total floating time (E) and floating latency (F) in the tested mice.
Fig. 7
Fig. 7
MRL/lpr mice display depression-like behavior at 8 weeks of age. The forced swim test was used to assess depression-like behavior in mice. % floating time: total floating time converted to percentage of total test time (A, test on 8 weeks of age. C, test on 18 weeks of age). B and D, floating latency is the delay time until mice give up struggling in the water and start to show the ‘still’ floating behavior. Time was recorded in seconds. B, testing 8 week old mice. D, testing 18 week old mice. E, F. Two way ANOVA analysis of the effects of age and genotype on total floating time (E) and floating latency (F) in the tested mice.
Fig. 8
Fig. 8
Lupus autoantibody levels correlate with depression behavior. Linear regression correlation of % floating time in forced swim test with serum anti-dsDNA titers (A, 8w; D, 18w), anti-NMDAR antibody titers (B, 8w; E, 18w), or anti-cardiolipin antibody titers (C, 8w; F, 18w) were shown. MRL/+: ■, MRL/lpr: ▼. P values and Pearson’s R values for each correlation are also shown in the graphs.
Fig. 9
Fig. 9
MRI/MRSI analyses show differences in regional brain metabolism between control and lupus mice. The figure illustrates the type of spectra obtained from the hippocampus (top) and thalamus (bottom), with a corresponding anatomical image of a lupus mouse brain. Data from the comparative analysis of lupus and control mouse brains is provided in Table 2.

References

    1. Bessa JM, Oliveira M, Cerqueira JJ, Almeida OF, Sousa N. Age-related qualitative shift in emotional behaviour: paradoxical findings after re-exposure of rats in the elevated-plus maze. Behav Brain Res. 2005;162:135–142.
    1. Bluestein HG. The central nervous system in systemic lupus erythematosus. In: Lahita RG, editor. Systemic lupus erythematosus. Churchill Livingstone; New York: 1992. pp. 639–655.
    1. Bonfa E, Marshak-Rothstein A, Weissbach H, Brot N, Elkon K. Frequency and epitope recognition of anti-ribosome P antibodies from humans with systemic lupus erythematosus and MRL/lpr mice are similar. J Immunol. 1988;140:3434–3437.
    1. Brey RL, Abbott RD, Curb JD, Sharp DS, Ross GW, Stallworth CL, Kittner SJ. beta(2)-Glycoprotein 1-dependent anticardiolipin antibodies and risk of ischemic stroke and myocardial infarction: the honolulu heart program. Stroke. 2001;32:1701–1706.
    1. Brooks WM, Sabet A, Sibbitt WL, Jr, Barker PB, van Zijl PC, Duyn JH, Moonen CT. Neurochemistry of brain lesions determined by spectroscopic imaging in systemic lupus erythematosus. J Rheumatol. 1997;24:2323–2329.
    1. Chu JL, Drappa J, Parnassa A, Elkon KB. The defect in Fas mRNA expression in MRL/lpr mice is associated with insertion of the retrotransposon, ETn. J Exp Med. 1993;178:723–730.
    1. Cohen-Solal JF, Jeganathan V, Grimaldi CM, Peeva E, Diamond B. Sex hormones and SLE: influencing the fate of autoreactive B cells. Curr Top Microbiol Immunol. 2006;305:67–88.
    1. Dantzer R, Bluthe RM, Koob GF, Le Moal M. Modulation of social memory in male rats by neurohypophyseal peptides. Psychopharmacology. 1987;91:363–368.
    1. DeGiorgio LA, Konstantinov KN, Lee SC, Hardin JA, Volpe BT, Diamond B. A subset of lupus anti-DNA antibodies cross-reacts with the NR2 glutamate receptor in systemic lupus erythematosus. Nat Med. 2001;7:1189–1193.
    1. Deocharan B, Qing X, Beger E, Putterman C. Antigenic triggers and molecular targets for anti-double-stranded DNA antibodies. Lupus. 2002;11:865–871.
    1. Deocharan B, Zhou Z, Antar K, Siconolfi-Baez L, Angeletti RH, Hardin J, Putterman C. Alpha-actinin immunization elicits anti-chromatin autoimmunity in nonautoimmune mice. J Immunol. 2007;179:1313–1321.
    1. Eber T, Chapman J, Shoenfeld Y. Anti-ribosomal P-protein and its role in psychiatric manifestations of systemic lupus erythematosus: myth or reality? Lupus. 2005;14:571–575.
    1. Elkon KB, Bonfa E, Llovet R, Eisenberg RA. Association between anti-Sm and anti-ribosomal P protein autoantibodies in human systemic lupus erythematosus and MRL/lpr mice. J Immunol. 1989;143:1549–1554.
    1. Emmer BJ, van der Grond J, Steup-Beekman GM, Huizinga TW, van Buchem MA. Selective involvement of the amygdala in systemic lupus erythematosus. PLoS Med. 2006;3:e499.
    1. Ennaceur A, Neave N, Aggleton JP. Spontaneous object recognition and object location memory in rats: the effects of lesions in the cingulate cortices, the medial prefrontal cortex, the cingulum bundle and the fornix. Exp Brain Res. 1997;113:509–519.
    1. Fragoso-Loyo H, Richaud-Patin Y, Orozco-Narvaez A, Davila-Maldonado L, Atisha-Fregoso Y, Llorente L, Sanchez-Guerrero J. Interleukin-6 and chemokines in the neuropsychiatric manifestations of systemic lupus erythematosus. Arthritis Rheum. 2007;56:1242–1250.
    1. Garwood M, DelaBarre L. The return of the frequency sweep: designing adiabatic pulses for contemporary NMR. J Magn Reson. 2001;153:155–177.
    1. Gorelick PB. Stroke prevention therapy beyond antithrombotics: unifying mechanisms in ischemic stroke pathogenesis and implications for therapy: an invited review. Stroke. 2002;33:862–875.
    1. Grimaldi CM. Sex and systemic lupus erythematosus: the role of the sex hormones estrogen and prolactin on the regulation of autoreactive B cells. Curr Opin Rheumatol. 2006;18:456–461.
    1. Hermosillo-Romo D, Brey RL. Diagnosis and management of patients with neuropsychiatric systemic lupus erythematosus (NPSLE) Best Pract Res Clin Rheumatol. 2002;16:229–244.
    1. Hess DC, Taormina M, Thompson J, Sethi KD, Diamond B, Rao R, Chamberlain CR, Feldman DS. Cognitive and neurologic deficits in the MRL/lpr mouse: a clinicopathologic study. J Rheumatol. 1993;20:610–617.
    1. Huerta PT, Kowal C, DeGiorgio LA, Volpe BT, Diamond B. Immunity and behavior: antibodies alter emotion. Proc Natl Acad Sci U S A. 2006;103:678–683.
    1. Katzav A, Solodeev I, Brodsky O, Chapman J, Pick CG, Blank M, Zhang W, Reichlin M, Shoenfeld Y. Induction of autoimmune depression in mice by anti-ribosomal P antibodies via the limbic system. Arthritis Rheum. 2007;56:938–948.
    1. Kowal C, Degiorgio LA, Lee JY, Edgar MA, Huerta PT, Volpe BT, Diamond B. Human lupus autoantibodies against NMDA receptors mediate cognitive impairment. Proc Natl Acad Sci U S A. 2006;103:19854–19859.
    1. Kowal C, DeGiorgio LA, Nakaoka T, Hetherington H, Huerta PT, Diamond B, Volpe BT. Cognition and immunity; antibody impairs memory. Immunity. 2004;21:179–188.
    1. Lahita RG. The role of sex hormones in systemic lupus erythematosus. Curr Opin Rheumatol. 1999;11:352–356.
    1. Lapteva L, Nowak M, Yarboro CH, Takada K, Roebuck-Spencer T, Weickert T, Bleiberg J, Rosenstein D, Pao M, Patronas N, Steele S, Manzano M, van der Veen JW, Lipsky PE, Marenco S, Wesley R, Volpe B, Diamond B, Illei GG. Anti-N-methyl-D-aspartate receptor antibodies, cognitive dysfunction, and depression in systemic lupus erythematosus. Arthritis Rheum. 2006;54:2505–2514.
    1. Lawrence DA, Bolivar VJ, Hudson CA, Mondal TK, Pabello NG. Antibody induction of lupus-like neuropsychiatric manifestations. J Neuroimmunol. 2007;182:185–194.
    1. Leritz E, Brandt J, Minor M, Reis-Jensen F, Petri M. “Subcortical” cognitive impairment in patients with systemic lupus erythematosus. J Int Neuropsychol Soc. 2000;6:821–825.
    1. Miyasaka N, Takahashi K, Hetherington HP. 1H NMR spectroscopic imaging of the mouse brain at 9.4 T. J Magn Reson Imaging. 2006;24:908–913.
    1. Montes GS, Luque EH. Effects of ovarian steroids on vaginal smears in the rat. Acta Anat. 1988;133:192–199.
    1. Muscal E, Myones BL. The role of autoantibodies in pediatric neuropsychiatric systemic lupus erythematosus. Autoimmun Rev. 2007;6:215–217.
    1. Nery FG, Borba EF, Viana VS, Hatch JP, Soares JC, Bonfa E, Neto FL. Prevalence of depressive and anxiety disorders in systemic lupus erythematosus and their association with anti-ribosomal P antibodies. Prog Neuropsychopharmacol Biol Psychiatry. 2008;32:695–700.
    1. Pan JW, Kim JH, Cohen-Gadol A, Pan C, Spencer DD, Hetherington HP. Regional energetic dysfunction in hippocampal epilepsy. Acta Neurol Scand. 2005;111:218–224.
    1. Pause BM, Miranda A, Goder R, Aldenhoff JB, Ferstl R. Reduced olfactory performance in patients with major depression. J Psychiatr Res. 2001;35:271–277.
    1. Pellow S, Chopin P, File SE, Briley M. Validation of open:closed arm entries in an elevated plus-maze as a measure of anxiety in the rat. J Neurosci Methods. 1985;14:149–167.
    1. Petri M. Epidemiology of the antiphospholipid antibody syndrome. J Autoimmun. 2000;15:145–151.
    1. Porsolt RD, Bertin A, Jalfre M. Behavioral despair in mice: a primary screening test for antidepressants. Arch Int Pharmacodyn Ther. 1977a;229:327–336.
    1. Porsolt RD, Le Pichon M, Jalfre M. Depression: a new animal model sensitive to antidepressant treatments. Nature. 1977b;266:730–732.
    1. Putterman C. New approaches to the renal pathogenicity of anti-DNA antibodies in systemic lupus erythematosus. Autoimmun Rev. 1994;3:7–11.
    1. Putterman C, Diamond B. Immunization with a peptide surrogate for double-stranded DNA (dsDNA) induces autoantibody production and renal immunoglobulin deposition. J Exp Med. 1998;188:29–38.
    1. Rudofsky UH, Evans BD, Balaban SL, Mottironi VD, Gabrielsen AE. Differences in expression of lupus nephritis in New Zealand mixed H-2z homozygous inbred strains of mice derived from New Zealand black and New Zealand white mice. Origins and initial characterization. Lab Invest. 1993;68:419–426.
    1. Rudofsky UH, Lawrence DA. New Zealand mixed mice: a genetic systemic lupus erythematosus model for assessing environmental effects. Environ Health Perspect. 1999;107(Suppl 5):713–721.
    1. Sakic B, Szechtman H, Braciak T, Richards C, Gauldie J, Denburg JA. Reduced preference for sucrose in autoimmune mice: a possible role of interleukin-6. Brain Res Bull. 1997a;44:155–165.
    1. Sakic B, Szechtman H, Denburg JA. Neurobehavioral alterations in autoimmune mice. Neurosci Biobehav Rev. 1997b;21:327–340.
    1. Sakic B, Szechtman H, Denburg S, Carbotte R, Denburg JA. Spatial learning during the course of autoimmune disease in MRL mice. Behav Brain Res. 1993;54:57–66.
    1. Sakic B, Szechtman H, Keffer M, Talangbayan H, Stead R, Denburg JA. A behavioral profile of autoimmune lupus-prone MRL mice. Brain Behav Immun. 1992;6:265–285.
    1. Sakic B, Szechtman H, Stead RH, Denburg JA. Joint pathology and behavioral performance in autoimmune MRL-lpr Mice. Physiol Behav. 1996;60:901–905.
    1. Sakic B, Szechtman H, Talangbayan H, Denburg SD, Carbotte RM, Denburg JA. Disturbed emotionality in autoimmune MRL-lpr mice. Physiol Behav. 1994;56:609–617.
    1. Schneebaum AB, Singleton JD, West SG, Blodgett JK, Allen LG, Cheronis JC, Kotzin BL. Association of psychiatric manifestations with antibodies to ribosomal P proteins in systemic lupus erythematosus. Am J Med. 1991;90:54–62.
    1. Schrott LM, Crnic LS. Anxiety behavior, exploratory behavior, and activity in NZB x NZW F1 hybrid mice: role of genotype and autoimmune disease progression. Brain Behav Immun. 1996;10:260–274.
    1. Shoenfeld Y. To smell autoimmunity: anti-P-ribosomal autoantibodies, depression, and the olfactory system. J Autoimmun. 2007;28:165–9.
    1. Shoenfeld Y, Nahum A, Korczyn AD, Dano M, Rabinowitz R, Beilin O, Pick CG, Leider-Trejo L, Kalashnikova L, Blank M, Chapman J. Neuronal-binding antibodies from patients with antiphospholipid syndrome induce cognitive deficits following intrathecal passive transfer. Lupus. 2003;12:436–442.
    1. Sibbitt WL, Jr, Haseler LJ, Griffey RH, Hart BL, Sibbitt RR, Matwiyoff NA. Analysis of cerebral structural changes in systemic lupus erythematosus by proton MR spectroscopy. AJNR Am J Neuroradiol. 1994;15:923–928.
    1. Sibbitt WL, Jr, Haseler LJ, Griffey RR, Friedman SD, Brooks WM. Neurometabolism of active neuropsychiatric lupus determined with proton MR spectroscopy. AJNR Am J Neuroradiol. 1997;18:1271–1277.
    1. Sibbitt WL, Jr, Sibbitt RR. Magnetic resonance spectroscopy and positron emission tomography scanning in neuropsychiatric systemic lupus erythematosus. Rheum Dis Clin North Am. 1993;19:851–868.
    1. Sidor MM, Sakic B, Malinowski PM, Ballok DA, Oleschuk CJ, Macri J. Elevated immunoglobulin levels in the cerebrospinal fluid from lupus-prone mice. J Neuroimmunol. 2005;165:104–113.
    1. Stojanovich L, Zandman-Goddard G, Pavlovich S, Sikanich N. Psychiatric manifestations in systemic lupus erythematosus. Autoimmun Rev. 2007;6:421–426.
    1. Theofilopoulos AN. Murine models of lupus. In: Lahita RG, editor. Systemic lupus erythematosus. Churchill Livingstone; New York: 1992. pp. 121–194.
    1. Toubi E, Shoenfeld Y. Clinical and biological aspects of anti-P-ribosomal protein autoantibodies. Autoimmun Rev. 2007;6:119–125.
    1. Twieg DB, Meyerhoff DJ, Hubesch B, Roth K, Sappey-Marinier D, Boska MD, Gober JR, Schaefer S, Weiner MW. Phosphorus-31 magnetic resonance spectroscopy in humans by spectroscopic imaging: localized spectroscopy and metabolite imaging. Magn Reson Med. 1989;12:291–305.
    1. Vogelweid CM, Johnson GC, Besch-Williford CL, Basler J, Walker SE. Inflammatory central nervous system disease in lupus-prone MRL/lpr mice: comparative histologic and immunohistochemical findings. J Neuroimmunol. 1991;35:89–99.
    1. Wekking EM. Psychiatric symptoms in systemic lupus erythematosus: an update. Psychosom Med. 1993;55:219–228.
    1. Zandman-Goddard G, Chapman J, Shoenfeld Y. Autoantibodies involved in neuropsychiatric SLE and antiphospholipid syndrome. Semin Arthritis Rheum. 2007;36:297–315.
    1. Ziporen L, Shoenfeld Y, Levy Y, Korczyn AD. Neurological dysfunction and hyperactive behavior associated with antiphospholipid antibodies. A mouse model. J Clin Invest. 1997;100:613–619.
    1. Zueger M, Urani A, Chourbaji S, Zacher C, Roche M, Harkin A, Gass P. Olfactory bulbectomy in mice induces alterations in exploratory behavior. Neurosci Lett. 2005;374:142–146.

Source: PubMed

3
Předplatit