SlideShare a Scribd company logo
1 of 13
Mechanisms by which pro-inflammatory cytokines and anti-heterogeneous nuclear
ribonuclear protein A1-M9 (anti-hnRNP A1-M9) antibodies cause neurodegeneration
in multiple sclerosis
Research proposal:
NIH Medical Student Research Fellowship Program
Peter Ketch
1177 Union Ave, #302
Memphis, TN 38104
(214) 901-4525
pketch@uthsc.edu
University of Tennessee Health Science Center
College of Medicine, Class of 2019
Mentor: Dr. Michael C. Levin, MD
Professor, Department of Neurology & Anatomy Neurobiology
University of Tennessee Health Science Center
Chief, Neurology Service, VAMC-Memphis
Director, Multiple Sclerosis Center
Laboratory for Viral and Demyelinating Diseases
855 Madison Avenue, Room 415
Memphis, TN 38163
(901) 448-2243
mlevin@uthsc.edu
May 31, 2016 – August 16, 2016
Memphis VA Medical Center
Abstract
Multiple sclerosis (MS) is a chronic inflammatory disease of the central nervous system (CNS) with
hallmark demyelination and neurodegeneration. Data suggest that Th1 and Th17 CD4+ lymphocytes and their
cytokines are thought to play a role in pathogenesis of neurodegeneration seen in MS. Neuronal exposure to
a mixture of these pro-inflammatory cytokines causes mislocalization of heterogeneous nuclear ribonuclear
protein A1 (hnRNP A1), which accumulates in the cytoplasm. Importantly, MS patients develop an antibody
against hnRNP A1. Similar experiments demonstrate that anti-hnRNP A1-M9 antibodies induce similar
molecular events, in addition to stress granule (SG) formation and changes in RNA metabolism of clinically
relevant RNAs. We hypothesize that an individual or combinations of pro-inflammatory Th17 cytokines can
influence molecular events important to the pathogenesis of MS, including mislocalization of hnRNP A1 and
the formation of SGs. In vitro experiments with SK-N-SH cells demonstrate that select individual and
combinations of Th17 cytokines (IL-17A, IL-23, IL-1β, TGF-β, and IL-6) can induce the formation of SGs,
mislocalization of hnRNP A1, and colocalization of hnRNP A1 with a SG marker, Poly(A)-binding protein
(PABP). Taken together, these data suggest a potential role of pro-inflammatory Th17 cytokines in the
pathogenesis of neurodegeneration through an hnRNP A1 mechanism seen in MS.
I. Introduction
Multiple sclerosis (MS) is an immune-mediated inflammatory disease that results in demyelination of
the central nervous system (CNS). The neurological disability seen in patients with MS is highly variable,
but can progress irreversibly in association with neuronal and axonal damage, or neurodegeneration.
Neurodegeneration in the CNS has been shown to contribute to the pathogenesis of MS, but was thought to
be secondary to the disease’s hallmark inflammatory demyelination. However, neurodegeneration can occur
independently of demyelinating plaque formation and is now viewed a critical component of the long-term
neurological disability seen in MS, especially progressive forms. In fact, a number of studies demonstrate a
significant reduction in nerve fiber density in corticospinal tracts and dorsal columns, independent of plaque
formation2,8-22. Although demyelination and its associated inflammatory processes are well described, the
pathophysiological mechanisms of neurodegeneration in MS are not well understood. Evidence of neuronal
and axonal degeneration has been shown in the brains and spinal cords of patients with MS, although
specific mechanisms are not clearly indicated and appear to be multifactorial9-11,23-25.
RNA binding proteins (RBPs) belong to a diverse class of proteins with varying functions including
RNA transcription, transport and translation. An important function of some of these RBPs, including
heterogeneous nuclear ribonuclearprotein A1 (hnRNP A1), is transport of mRNA from the nucleus to the
cytoplasm. Extensive studies in Dr. Levin’s lab have demonstrated that MS patients develop anti-hnRNP A1
antibodies and that these antibodies cause neurodegeneration3,4,21,33,35,37. Normally functioning hnRNP A1
binds mRNA and transportin in the nucleus, transports mRNA to the cytoplasm via a nuclear pore, then
quickly translocates back into nucleus4. The M9 region of hnRNP A1 contains a sequence that binds
transportin. When antibodies bind to this region, hnRNP A1 cannot return to the nucleus. The presence of
autoantibodies to M9 in patients with MS causes hnRNP A1 to accumulate in the cytoplasm, instead of the
nucleus where it is usually observed in highest quantity4,32. Furthermore, this accumulation corresponds to
reduced levels of ATP and increased levels of apoptosis, important markers of neurodegeneration4. These
findings demonstrate an important association between anti-A1-M9 antibodies and neurodegeneration in MS.
Anti-hnRNPA1-M9 antibodies can induce stress granule (SG) formation in neurons and can cause
changes in RNA metabolism of the hnRNP A1 protein’s RNA cargo44. During times of cellular stress, SGs
contain dysfunctional RBPs and their translationally repressed mRNA cargo45. Studies in Dr. Levin’s lab
demonstrated that adding anti-A1-M9 antibodies to neuronal cell lines in culture resulted in SG formation
and that the antibodies colocalize with the SGs44. In previous studies, Dr. Levin’s lab showed that the levels
of specific genes, including SPG4 (spastin) and SPG7 (paraplegin), were altered in response to transfection
of neuronal cells with anti-
hnRNP A1-M9 antibodies.
RNA immunoprecipitation
studies revealed that SPG4
and SPG7 (to a lesser
extent) bound hnRNPA1
(Fig SA2-4). Furthermore,
Western blots showed that anti-hnRNP A1-M9 antibodies caused a profound reduction in SPG4 and SPG7
protein levels3. This experiment demonstrated that anti-hnRNPA1-M9 antibodies specifically altered the
translation of clinically relevant RNAs that were shown to bind to A1.
In addition to antibody-mediated
immunoreactivity, there are other factors, like
pro-inflammatory cytokines, that may act in
conjunction to produce the neurodegeneration
seen in MS. Different subtypes of MS display
distinct inflammatory activity. Early forms of
MS (acute and relapsing-remitting) are
characterized by Th1 and Th17 CD4+
lymphocyte responses and demyelination and
neuronal injury correlated with T and B cells11,14,26,29-31. More diffuse CNS inflammation and IgG-positive
Fig SA2-4. RNAImmunoprecipitation andWesternblot
analyses. SK-N-SHcells lysateswere incubated with
agarose beads labeled anti-A1-M9 or control abs. A.
Western blots showed thathnRNP A1 waspresentin the
lysate (L) and the anti-A1-M9 IP (A1) butnotin control IgG
IP (IgG). B. RNA wasisolated from protein eluents and
amplified by RT-PCR. Values were normalizedto GAPDH.
Results revealed thathnRNP A1 and SPG4 are RNA
binding partners ofhnRNP A1 and SPG-7 showed a
positive trend towardsRNA binding. p≤0.05 analyzed by t-
test. C. Anti-hnRNP A1 antibodiesalterproteinlevels as
measuredby WesternBlot. SK-N-SHcells were treated
with anti-A1-M9 abs or control IgG and proteins run on
Western blots. Results revealed hnRNP A1, SPG4,and
SPG7 had markedly reducedprotein levelsin anti-hnRNP
A1-M9 antibody treated cells compared to controls. There
was no change in Beta actin or SPG20.
Fig SA2-5A: In primary
neurons in culture, A1
mis-localizes to the
cytoplasm when
exposed to Th1and
Th17 cytokines. Under
normal conditions (0),
hnRNP A1 (green) co-
localizes in the nucleus
(DAPI-blue). When
exposed to Th1 or Th17
cytokines (see Table
below – column labeled
‘C’), hnRNP A1
mislocalizes to the
cytoplasm (arrows).
Arrowheads identify cell
bodies of neurons.
plasma cells predominate in progressive forms of MS14. One specific study showed that neurodegeneration
of specific neural pathways was directly associated with T-cell recruitment5. Other studies have
demonstrated that both CD4+ Th1 and Th17 lymphocytes induced disease6,7,27,28. Preliminary studies in Dr.
Levin’s lab, using a mixture of both Th cytokine inducers (priming cytokines) and effectors (cytokine
products), demonstrated that both Th1 and Th17 cytokines caused mislocalization of A1 from the nucleus to
the cytoplasm (Fig SA2-5A).
II. Hypothesis
We hypothesize that an individual or combinations of pro-inflammatory Th17 cytokines can
influence molecular events important to the pathogenesis of MS.
III. Specific Aims
The objective is to determine the mechanistic role that pro-inflammatory Th17 cytokines play in the
mislocalization of hnRNP A1 and stress granule formation in SK-N-SH cells.
1. Use immunocytochemistry to determine whether individual or combinations of cytokines
cause mislocalization of hnRNP A1
2. Use immunocytochemistry to determine whether individual or combinations of cytokines
induce stress granule formation
IV. Materials and Methods
Cells
The SK-N-SH cell line (ATCC, HTB-11), an immortalized human neuroblastoma cell line was
maintained in Dulbecco’s modified Eagle’s medium/F12 supplemented with 10% fetal bovine serum and 1%
penicillin-streptomycin antibiotics. 105 SK-N-SH cells were seeded per well into 500 uL of complete
DMEM/F12 media into 8 well chamber slides (Corning #354632) for immunocytochemistry experiments.
Exposure of SK-N-SH cells to cytokines in vitro
Anti-hnRNP A1 antibodies were obtained from Millipore (04-1469). Poly(A)-binding protein (PABP)
antibodies were obtained from abcam (ab-21060). The anti-hnRNP A1 antibodies are specific for hnRNP
A1-M9 and have been shown to overlap the M9 immunodominant epitope recognized by IgG isolated from
MS patients3. Cytokines were added to the
culture media of SK-N-SH cells in the
following concentrations, individually and
in combination, represented in Figure 1:
IL-17A (20ng/ml), IL-23 (20ng/ml), IL-1β
(20 ng/ml), TGF-β (2 ng/ml), IL-6 (25
ng/ml) .
Immunocytochemistry of hnRNP A1-M9 mislocalization and stress granule formation
105 SK-N-SH cells were seeded per well into 500 uL of complete DMEM/F12 media into 8 well
chamber slides (Corning #354632). 48 hours following cytokine addition, cells were fixed with 4%
paraformaldehyde for 15 minutes at room temperature (RT). Fixed cells were then blocked and
permeabilized with 6% Milk-PBS + 0.4% TritonX-100. Cells were then labeled for stress granules using
Poly(A)-binding protein (PABP) and hnRNP A1 at the manufacturer’s suggested concentration overnight at
4°C. Following primary antibody incubation, cells were washed 3 X 5 minutes with PBS-T. Cells were
treated with secondary anti-mouse conjugated FITC and anti-rabbit conjugated Texas Red at manufacturer’s
suggested concentration for 2 hours at RT. Cells were washed 3 X 5 minutes with PBS-T, 2 X 5 minutes
with PBS and mounted with DAPI mounting medium, and imaged by dual confocal microscopy.
VII. Results
Untreated SK-N-SH cells show nuclear localization of hnRNP-A1 (Figures 1-7, top) and no stress
granule formation. After treatment with Th17 effector cytokines (IL-17A, IL-23, IL-1β), hnRNP-A1 is
mislocalized to the cytoplasm where it colocalizes with stress granules (Figure 2, bottom, arrows). However
not all stress granules colocalized with hnRNP-A1 (Figure 2, bottom, circles). Following exposure to IL-1β,
SK-N-SH cells show hnRNP-A1 mislocalization and hnRNP-A1 colocalization with stress granules (Figure
3, bottom, arrows). SK-N-SH cells treated with IL-23 showed mislocalization of hnRNP-A1 to the
cytoplasm and the formation of stress granules; however hnRNP-A1 does not colocalize with these granules
(Figure 4, bottom). After treatment with IL-17A, hnRNP-A1 is mislocalized to the cytoplasm. PABP
staining revealed several small, punctuated areas of staining suggestive of stress granules, but further studies
are warranted to clarify whether or not IL-17A treatment induces stress granules in SK-N-SH cells (Figure 5,
bottom). Furthermore, hnRNP-A1 did not colocalize with PABP staining in IL-17A treated SK-N-SH cells.
Th17 inducer cytokine (TGF-β, IL-6) treated SK-N-SH cells show decreased localization to the nucleus
compared to untreated cells and possible mislocalization, although high background in this
immunofluorescence image warrants further experimentation to confirm this finding (Figure 6, bottom).
hnRNP-A1 did not colocalize with the stress granules formed in these Th17 inducer cytokine treated cells
(Figure 6, bottom). Following exposure to TGF-β, SK-N-SH cells show hnRNP-A1 mislocalization, the
formation of stress granules, and hnRNP-A1 colocalization with these stress granules (Figure 7, bottom,
arrows). After treatment with IL-6, there was mislocalization of hnRNP-A1 to the cytoplasm and the
formation of stress granules; however hnRNP-A1 does not colocalize with these granules (Figure 8, bottom).
A summary table detailing hnRNP-A1 mislocalization, stress granule formation and colocalization of
hnRNP-A1 with stress granules for each cytokine treatment group is provided (Figure 9).
VIII. Conclusion
In vitro experiments with SK-N-SH cells demonstrate that select individual and combinations of
Th17 cytokines (IL-17A, IL-23, IL-1β, TGF-β, and IL-6) can influence molecular events important to the
pathogenesis of MS including, mislocalization of hnRNP A1 (accumulation in the cytoplasm) and formation
of SGs. Furthermore, select individual and combinations of pro-inflammatory cytokines caused
colocalization of hnRNP A1 and PABP, a stress granule marker.
Taken together, these data suggest a potential role of pro-inflammatory Th17 cytokines in the
pathogenesis of neurodegeneration in MS via an hnRNP A1 mechanism. Previous studies have shown that
anti-hnRNP A1-M9 antibodies in patients with MS cause hnRNP A1 to accumulate in the cytoplasm, which
may cause cellular stress and resulting SG formation44. This study showed that similar molecular changes
occur in response to select individual and combinations of Th17 pro-inflammatory cytokines. These findings
suggest that both antibody-mediated immunoreactivity and pro-inflammatory cytokines may share a potential
mechanism that might contribute to neurodegeneration seen in MS, involving mislocalization and
accumulation of hnRNP A1 and SG formation. The exact mechanism by which this occurs requires further
study. Studies should be conducted involving simultaneous exposure to anti-hnRNP A1 antibodies and pro-
inflammatory cytokines to more closely mimic in vivo conditions and to further explore the role of Th17 pro-
inflammatory cytokines in the pathogenesis of neurodegeneration in MS.
IX. Potential Conflicts
Dr. Levin is the owner of a patent: “A biomarker for neurodegeneration in neurological disease.”
X. References
1. Lee, S. and M. Levin, Novel somatic single nucleotide variants within the RNA binding protein
hnRNP A1 in multiple sclerosis patients [v2; ref status: indexed]. Vol. 3. 2014.
2. Friese, M.A., B. Schattling, and L. Fugger, Mechanisms of neurodegeneration and axonal
dysfunction in multiple sclerosis. Nat Rev Neurol, 2014. 10(4): p. 225-38.
3. Lee, S., L. Xu, Y. Shin, L. Gardner, A. Hartzes, F.C. Dohan, C. Raine, R. Homayouni, and M.C.
Levin, A potential link between autoimmunity and neurodegeneration in immune-mediated
neurological disease. J Neuroimmunol, 2011. 235(1-2): p. 56-69.
4. Douglas, J., L. Gardner, and M.C. Levin, Antibodies to an intracellular antigen penetrate neuronal
cells and cause deleterious effects. J Clinical & Cellular Immunology 2013. 4(1): p. 134.
5. Brown, D.A. and P.E. Sawchenko, Time course & distribution of inflammatory & neurodegenerative
events suggest structural bases for the pathogenesis of experimental autoimmune encephalomyelitis. J
Comp Neurol, 2007. 502:236-60.
6. Arima, Y., M. Harada, D. Kamimura, J.H. Park, F. Kawano, F.E. Yull, T. Kawamoto, Y. Iwakura,
U.A. Betz, G. Marquez, T.S. Blackwell, Y. Ohira, T. Hirano, and M. Murakami, Regional neural
activation defines a gateway for autoreactive T cells to cross the blood-brain barrier. Cell, 2012.
148(3): p. 447-57.
7. Kamimura, D., M. Yamada, M. Harada, L. Sabharwal, J. Meng, H. Bando, H. Ogura, T. Atsumi, Y.
Arima, and M. Murakami, The gateway theory: bridging neural and immune interactions in the CNS.
Front Neurosci, 2013. 7: p. 204.
8. Trapp, B.D. and K.A. Nave, MS: an immune or neurodegenerative disorder? Annu Rev Neurosci,
2008. 31: p. 247-69.
9. Lassmann, H., MS: is there neurodegeneration independent from inflammation? J Neurol Sci, 2007.
259(1-2): p. 3-6.
10. Lassmann, H. and C.F. Lucchinetti, Cortical demyelination in CNS inflammatory demyelinating
diseases. Neurology, 2008. 70(5): p. 332-3.
11. Lassmann, H. and J. van Horssen, The molecular basis of neurodegeneration in multiple sclerosis.
FEBS Lett, 2011. 585(23): p. 3715-23.
12. Lucchinetti, C.F., B.F. Popescu, R.F. Bunyan, N.M. Moll, S.F. Roemer, H. Lassmann, W. Bruck, J.E.
Parisi, B.W. Scheithauer, C. Giannini, S.D. Weigand, J. Mandrekar, and R.M. Ransohoff,
Inflammatory cortical demyelination in early multiple sclerosis. N Engl J Med, 2011. 365(23): p.
2188-97.
13. Peterson, J.W. and B.D. Trapp, Neuropathobiology of multiple sclerosis. Neurol Clin, 2005. 23(1): p.
107-29, vi-vii.
14. Frischer, J.M., S. Bramow, A. Dal-Bianco, C.F. Lucchinetti, H. Rauschka, M. Schmidbauer, H.
Laursen, P.S. Sorensen, and H. Lassmann, The relation between inflammation and neurodegeneration
in multiple sclerosis brains. Brain, 2009. 132(Pt 5): p. 1175-89.
15. Fisniku, L.K., D.T. Chard, J.S. Jackson, V.M. Anderson, D.R. Altmann, K.A. Miszkiel, A.J.
Thompson, and D.H. Miller, Gray matter atrophy is related to long-term disability in multiple
sclerosis. Ann Neurol, 2008. 64(3): p. 247-54.
16. Geurts, J.J., Is progressive multiple sclerosis a gray matter disease? Ann Neurol, 2008. 64(3): p. 230-
2.
17. Ganter, P., C. Prince, and M.M. Esiri, Spinal cord axonal loss in multiple sclerosis: a post-mortem
study. Neuropathol Appl Neurobiol, 1999. 25(6): p. 459-67.
18. Lovas, G., N. Szilagyi, K. Majtenyi, M. Palkovits, and S. Komoly, Axonal changes in chronic
demyelinated cervical spinal cord plaques. Brain, 2000. 123 ( Pt 2): p. 308-17.
19. DeLuca, G.C., K. Williams, N. Evangelou, G.C. Ebers, and M.M. Esiri, The contribution of
demyelination to axonal loss in multiple sclerosis. Brain, 2006. 129(Pt 6): p. 1507-16.
20. Evangelou, N., G.C. DeLuca, T. Owens, and M.M. Esiri, Pathological study of spinal cord atrophy in
multiple sclerosis suggests limited role of local lesions. Brain, 2005. 128(Pt 1): p. 29-34.
21. Levin, M.C., S. Lee, L. Gardner, Y. Shin, J.N. Douglas, and C. Groover, Pathogenic mechanisms of
neurodegeneration based on the phenotypic expression of progressive forms of immune-mediated
neurologic disease. Degenerative Neurological and Neuromuscular Disease, 2012. 2: p. 175-187.
22. Levin, M.C., J. Douglas, L. Myers, S. Lee, Y. Shin, and L. Gardner, Neurodegeneration in multiple
sclerosis involves multiple pathogenic mechanisms. Degenerative Neurological and Neuromuscular
Disease, 2014. 4: p. 49-63.
23. Dutta, R. and B.D. Trapp, Pathogenesis of axonal and neuronal damage in multiple sclerosis.
Neurology, 2007. 68(22 Suppl 3): p. S22-31; discussion S43-54.
24. Benarroch, E.E., Acquired axonal degeneration and regeneration: Recent insights and clinical
correlations. Neurology, 2015. 84(20): p. 2076-85.
25. Coleman, M., Axon degeneration mechanisms: commonality amid diversity. Nat Rev Neurosci, 2005.
6(11): p. 889-98.
26. Dutta, R. and B. Trapp, Mechanisms of neuronal dysfunction and degeneration in multiple sclerosis.
Progress in Neurobiology, 2011. 93: p. 1-12.
27. Siffrin, V., H. Radbruch, R. Glumm, R. Niesner, M. Paterka, J. Herz, T. Leuenberger, S.M. Lehmann,
S. Luenstedt, J.L. Rinnenthal, G. Laube, H. Luche, S. Lehnardt, H.J. Fehling, O. Griesbeck, and F.
Zipp, In vivo imaging of partially reversible th17 cell-induced neuronal dysfunction in the course of
encephalomyelitis. Immunity, 2010. 33(3): p. 424-36.
28. Tanabe, S. and T. Yamashita, Repulsive guidance molecule-a is involved in Th17-cell-induced
neurodegeneration in autoimmune encephalomyelitis. Cell Rep, 2014. 9(4): p. 1459-70.
29. Steinman, L., Mixed results with modulation of TH-17 cells in human autoimmune diseases. Nat
Imm, 2010. 11(1): 41-4.
30. Antel, J., S. Antel, Z. Caramanos, D.L. Arnold, and T. Kuhlmann, Primary progressive multiple
sclerosis: part of the MS disease spectrum or separate disease entity? Acta Neuropathol, 2012.
123(5): p. 627-38.
31. Beecham, et al. Analysis of immune-related loci identifies 48 new susceptibility variants for multiple
sclerosis. Nat Genet, 2013. 45(11): p. 1353-60.
32. Levin, M.C., S. Lee, L.A. Gardner, Y. Shin, J.N. Douglas, and C. Cooper, Autoantibodies to Non-
myelin Antigens as Contributors to the Pathogenesis of Multiple Sclerosis. J Clin Cell Immunol,
2013. 4.
33. Levin, M., T. Lehky, N. Flerlage, D. Katz, D. Kingma, E. Jaffe, J. Heiss, N. Patronas, H. McFarland,
and S. Jacobson, Immunopathogenesis of HTLV-1 associated neurologic disease based on a spinal
cord biopsy from a patient with HTLV-1 associated myelopathy/tropical spastic paraparesis
(HAM/TSP). New Eng J Med, 1997. 336: p. 839-845.
34. Levin, M., M. Krichavsky, J. Berk, S. Foley, M. Rosenfeld, J. Dalmau, G. Chen, J. Posner, and S.
Jacobson, Neuronal molecular mimicry in immune mediated neurologic disease. Ann Neurol, 1998.
44(1): p. 87-98.
35. Levin, M.C., S.M. Lee, F. Kalume, Y. Morcos, F.C. Dohan, Jr., K.A. Hasty, J.C. Callaway, J. Zunt,
D. Desiderio, and J.M. Stuart, Autoimmunity due to molecular mimicry as a cause of neurological
disease. Nat Med, 2002. 8(5): p. 509-13.
36. Lee, S.M., Y. Morocos, H. Jang, J.M. Stuart, M.C. Levin, HTLV-1 induced molecular mimicry in
neurologic disease,In: Molecular Mimicry: Infection Inducing Autoimmune Disease, M. Oldstone,
Editor. 2005, Springer: New York.
37. Lee, S., M.C. Levin, Molecular mimicry in neurological disease: what is the evidence? Cell Mol Life
Sci, 2008. 65:.1161-75.
38. Lee, S.M., F.D. Dunnavant, H. Jang, J. Zunt, and M.C. Levin, Autoantibodies that recognize
functional domains of hnRNPA1 implicate molecular mimicry in the pathogenesis of neurological
disease. Neurosci Lett, 2006. 401(1-2): p. 188-93.
39. Kalume, F., S.M. Lee, Y. Morcos, J.C. Callaway, and M.C. Levin, Molecular mimicry: cross-reactive
antibodies from patients with immune-mediated neurologic disease inhibit neuronal firing. J Neurosci
Res, 2004. 77(1): p. 82-9.
40. Douglas, J.N., L.A. Gardner, S. Lee, Y. Shin, C.J. Groover, and M.C. Levin, Antibody transfection
into neurons as a tool to study disease pathogenesis. J Vis Exp, 2012(67).
41. Gascon, E., L. Vutskits, and J.Z. Kiss, Polysialic acid-neural cell adhesion molecule in brain
plasticity: from synapses to integration of new neurons. Brain Res Rev, 2007. 56(1): p. 101-18.
42. Levin, M.C., et al., Organization of galanin-immunoreactive inputs to the paraventricular nucleus
with special reference to their relationship to catecholaminergic afferents. J Comp Neurol, 1987.
261(4): p. 562-82.
43. Meyers, L., C.J. Groover, J. Douglas, S. Lee, D. Brand, M.C. Levin, and L.A. Gardner, A role for
Apolipoprotein A-I in the pathogenesis of multiple sclerosis. J Neuroimmunol, 2014. 277(1-2): p.
176-85.
44. Levin, M., Lee, S., Gardner, L., Douglas, J., Shin, Y., Sawchenko, P., Lalaor, S., Segal, B,
Contribution of a dysfunctional RNA binding protein, hnRNP A1, to neurodegeneration in MS.
Neurology, 2014. 82(10).
45. Kiebler, M.A., Bassell, G.J, Neuronal RNA granules: movers and makers. Neuron, 2006. 51(6):
p685-90.

More Related Content

What's hot

acs.jmedchem.5b01760
acs.jmedchem.5b01760acs.jmedchem.5b01760
acs.jmedchem.5b01760Marta Wylot
 
luisetto m. brain response in some systemic immune condition-toxicological as...
luisetto m. brain response in some systemic immune condition-toxicological as...luisetto m. brain response in some systemic immune condition-toxicological as...
luisetto m. brain response in some systemic immune condition-toxicological as...M. Luisetto Pharm.D.Spec. Pharmacology
 
Interesting medical news presentation
Interesting medical news presentation Interesting medical news presentation
Interesting medical news presentation Christian Granda
 
Poster_Emerging Scholars Program 2013
Poster_Emerging Scholars Program 2013Poster_Emerging Scholars Program 2013
Poster_Emerging Scholars Program 2013Unalissa Freso
 
CIRCULATIONAHA2014013675
CIRCULATIONAHA2014013675CIRCULATIONAHA2014013675
CIRCULATIONAHA2014013675Zhen Chen
 
Research Day Poster Spring 15
Research Day Poster Spring 15Research Day Poster Spring 15
Research Day Poster Spring 15Victor Suarez
 
Oligonucleotide Therapeutics
Oligonucleotide TherapeuticsOligonucleotide Therapeutics
Oligonucleotide Therapeuticsasarode
 
i1552-5783-57-2-372
i1552-5783-57-2-372i1552-5783-57-2-372
i1552-5783-57-2-372Ryo Iwata
 
Final Dissertation
Final DissertationFinal Dissertation
Final DissertationRachel Li
 
Inestrosa idn 2011
Inestrosa idn 2011Inestrosa idn 2011
Inestrosa idn 2011Jorge Parodi
 
Luisetto m, behzad n, ghulam r. brain response in some systemic immune condit...
Luisetto m, behzad n, ghulam r. brain response in some systemic immune condit...Luisetto m, behzad n, ghulam r. brain response in some systemic immune condit...
Luisetto m, behzad n, ghulam r. brain response in some systemic immune condit...M. Luisetto Pharm.D.Spec. Pharmacology
 
J Neurochem 2003
J Neurochem 2003J Neurochem 2003
J Neurochem 2003Raul Pardo
 
Luisetto m et al amyotropyc lateral sclerosis and endogenous -esogenous toxic...
Luisetto m et al amyotropyc lateral sclerosis and endogenous -esogenous toxic...Luisetto m et al amyotropyc lateral sclerosis and endogenous -esogenous toxic...
Luisetto m et al amyotropyc lateral sclerosis and endogenous -esogenous toxic...M. Luisetto Pharm.D.Spec. Pharmacology
 
Blain_AlzResTherapy_2016_Characterization of FRM-36143 as a new γ-secretase m...
Blain_AlzResTherapy_2016_Characterization of FRM-36143 as a new γ-secretase m...Blain_AlzResTherapy_2016_Characterization of FRM-36143 as a new γ-secretase m...
Blain_AlzResTherapy_2016_Characterization of FRM-36143 as a new γ-secretase m...Gerhard Koenig
 
Role of-mek1-in-tlr4-mediated-signaling
Role of-mek1-in-tlr4-mediated-signalingRole of-mek1-in-tlr4-mediated-signaling
Role of-mek1-in-tlr4-mediated-signalingscience journals
 
Nickerson M Publications
Nickerson M PublicationsNickerson M Publications
Nickerson M PublicationsMolly Nickerson
 
10. jULIETA gONZALEZ. Xerostomia no solo un problema de agua
10. jULIETA gONZALEZ. Xerostomia no solo un problema de agua10. jULIETA gONZALEZ. Xerostomia no solo un problema de agua
10. jULIETA gONZALEZ. Xerostomia no solo un problema de aguacrea-autoinmunidad
 

What's hot (20)

BIOL3200 PROPOSAL
BIOL3200 PROPOSALBIOL3200 PROPOSAL
BIOL3200 PROPOSAL
 
acs.jmedchem.5b01760
acs.jmedchem.5b01760acs.jmedchem.5b01760
acs.jmedchem.5b01760
 
luisetto m. brain response in some systemic immune condition-toxicological as...
luisetto m. brain response in some systemic immune condition-toxicological as...luisetto m. brain response in some systemic immune condition-toxicological as...
luisetto m. brain response in some systemic immune condition-toxicological as...
 
Interesting medical news presentation
Interesting medical news presentation Interesting medical news presentation
Interesting medical news presentation
 
Poster_Emerging Scholars Program 2013
Poster_Emerging Scholars Program 2013Poster_Emerging Scholars Program 2013
Poster_Emerging Scholars Program 2013
 
CIRCULATIONAHA2014013675
CIRCULATIONAHA2014013675CIRCULATIONAHA2014013675
CIRCULATIONAHA2014013675
 
Research Day Poster Spring 15
Research Day Poster Spring 15Research Day Poster Spring 15
Research Day Poster Spring 15
 
Oligonucleotide Therapeutics
Oligonucleotide TherapeuticsOligonucleotide Therapeutics
Oligonucleotide Therapeutics
 
i1552-5783-57-2-372
i1552-5783-57-2-372i1552-5783-57-2-372
i1552-5783-57-2-372
 
Final Dissertation
Final DissertationFinal Dissertation
Final Dissertation
 
Inestrosa idn 2011
Inestrosa idn 2011Inestrosa idn 2011
Inestrosa idn 2011
 
Bordet-JPET-2007
Bordet-JPET-2007Bordet-JPET-2007
Bordet-JPET-2007
 
Neuroimmunology update
Neuroimmunology updateNeuroimmunology update
Neuroimmunology update
 
Luisetto m, behzad n, ghulam r. brain response in some systemic immune condit...
Luisetto m, behzad n, ghulam r. brain response in some systemic immune condit...Luisetto m, behzad n, ghulam r. brain response in some systemic immune condit...
Luisetto m, behzad n, ghulam r. brain response in some systemic immune condit...
 
J Neurochem 2003
J Neurochem 2003J Neurochem 2003
J Neurochem 2003
 
Luisetto m et al amyotropyc lateral sclerosis and endogenous -esogenous toxic...
Luisetto m et al amyotropyc lateral sclerosis and endogenous -esogenous toxic...Luisetto m et al amyotropyc lateral sclerosis and endogenous -esogenous toxic...
Luisetto m et al amyotropyc lateral sclerosis and endogenous -esogenous toxic...
 
Blain_AlzResTherapy_2016_Characterization of FRM-36143 as a new γ-secretase m...
Blain_AlzResTherapy_2016_Characterization of FRM-36143 as a new γ-secretase m...Blain_AlzResTherapy_2016_Characterization of FRM-36143 as a new γ-secretase m...
Blain_AlzResTherapy_2016_Characterization of FRM-36143 as a new γ-secretase m...
 
Role of-mek1-in-tlr4-mediated-signaling
Role of-mek1-in-tlr4-mediated-signalingRole of-mek1-in-tlr4-mediated-signaling
Role of-mek1-in-tlr4-mediated-signaling
 
Nickerson M Publications
Nickerson M PublicationsNickerson M Publications
Nickerson M Publications
 
10. jULIETA gONZALEZ. Xerostomia no solo un problema de agua
10. jULIETA gONZALEZ. Xerostomia no solo un problema de agua10. jULIETA gONZALEZ. Xerostomia no solo un problema de agua
10. jULIETA gONZALEZ. Xerostomia no solo un problema de agua
 

Similar to MSRF.finalpaper.pketch.abstractedit

A Proposed Mechanism for Autoimmune Mediated Motor Dysfunctions, and a Hypoth...
A Proposed Mechanism for Autoimmune Mediated Motor Dysfunctions, and a Hypoth...A Proposed Mechanism for Autoimmune Mediated Motor Dysfunctions, and a Hypoth...
A Proposed Mechanism for Autoimmune Mediated Motor Dysfunctions, and a Hypoth...ramdab
 
Pathogenesis of Paraneoplastic Syndromes.pptx
Pathogenesis of Paraneoplastic Syndromes.pptxPathogenesis of Paraneoplastic Syndromes.pptx
Pathogenesis of Paraneoplastic Syndromes.pptxMohamed AbdElhady
 
MS pathogenesis and risk factors.pptx
MS pathogenesis and risk factors.pptxMS pathogenesis and risk factors.pptx
MS pathogenesis and risk factors.pptxLobna A.Mohamed
 
Autoimmune Encephalitis a Mini Review_Crimson Publishers
Autoimmune Encephalitis a Mini Review_Crimson PublishersAutoimmune Encephalitis a Mini Review_Crimson Publishers
Autoimmune Encephalitis a Mini Review_Crimson PublishersCrimsonPublishersAICS
 
Poster_Template_Horizontal_Red
Poster_Template_Horizontal_RedPoster_Template_Horizontal_Red
Poster_Template_Horizontal_RedAlexander Seutin
 
Plegable Molecular biology.
Plegable Molecular biology.Plegable Molecular biology.
Plegable Molecular biology.jcardonavelez
 
Plegable jonathan cardona vélez
Plegable jonathan cardona vélezPlegable jonathan cardona vélez
Plegable jonathan cardona vélezjcardonavelez
 
Isabela Palacio Jaramillo 3 semestre
Isabela Palacio Jaramillo 3 semestre Isabela Palacio Jaramillo 3 semestre
Isabela Palacio Jaramillo 3 semestre isabelapalacio5
 
SSC4a Report (final)
SSC4a Report (final)SSC4a Report (final)
SSC4a Report (final)Gregory Myles
 
hinckley poster5 (2)
hinckley poster5 (2)hinckley poster5 (2)
hinckley poster5 (2)Sj Afshari
 
Taxanes induced Neuropathy
Taxanes induced Neuropathy Taxanes induced Neuropathy
Taxanes induced Neuropathy Badheeb
 
"Bioluminescent Imaging of Histidyl-Transfer RNA Synthetase-Induced Myositis ...
"Bioluminescent Imaging of Histidyl-Transfer RNA Synthetase-Induced Myositis ..."Bioluminescent Imaging of Histidyl-Transfer RNA Synthetase-Induced Myositis ...
"Bioluminescent Imaging of Histidyl-Transfer RNA Synthetase-Induced Myositis ...Nicholas Young
 
Approcci bioenergetici per la neuroprotezione nella malattia parkinsoniana
Approcci bioenergetici per la neuroprotezione nella malattia parkinsonianaApprocci bioenergetici per la neuroprotezione nella malattia parkinsoniana
Approcci bioenergetici per la neuroprotezione nella malattia parkinsonianaMerqurioEditore_redazione
 
Topic of the month...Paraneoplastic syndromes
Topic of the month...Paraneoplastic syndromesTopic of the month...Paraneoplastic syndromes
Topic of the month...Paraneoplastic syndromesProfessor Yasser Metwally
 

Similar to MSRF.finalpaper.pketch.abstractedit (20)

Mecanismo les
Mecanismo lesMecanismo les
Mecanismo les
 
2014 BDSRA Micsenyi LINCL and JNCL
2014 BDSRA Micsenyi LINCL and JNCL2014 BDSRA Micsenyi LINCL and JNCL
2014 BDSRA Micsenyi LINCL and JNCL
 
A Proposed Mechanism for Autoimmune Mediated Motor Dysfunctions, and a Hypoth...
A Proposed Mechanism for Autoimmune Mediated Motor Dysfunctions, and a Hypoth...A Proposed Mechanism for Autoimmune Mediated Motor Dysfunctions, and a Hypoth...
A Proposed Mechanism for Autoimmune Mediated Motor Dysfunctions, and a Hypoth...
 
Pathogenesis of Paraneoplastic Syndromes.pptx
Pathogenesis of Paraneoplastic Syndromes.pptxPathogenesis of Paraneoplastic Syndromes.pptx
Pathogenesis of Paraneoplastic Syndromes.pptx
 
2014 BDSRA Storch CLN7
2014 BDSRA Storch CLN72014 BDSRA Storch CLN7
2014 BDSRA Storch CLN7
 
Abstract(English)
Abstract(English)Abstract(English)
Abstract(English)
 
MS pathogenesis and risk factors.pptx
MS pathogenesis and risk factors.pptxMS pathogenesis and risk factors.pptx
MS pathogenesis and risk factors.pptx
 
Autoimmune Encephalitis a Mini Review_Crimson Publishers
Autoimmune Encephalitis a Mini Review_Crimson PublishersAutoimmune Encephalitis a Mini Review_Crimson Publishers
Autoimmune Encephalitis a Mini Review_Crimson Publishers
 
Poster_Template_Horizontal_Red
Poster_Template_Horizontal_RedPoster_Template_Horizontal_Red
Poster_Template_Horizontal_Red
 
Plegable Molecular biology.
Plegable Molecular biology.Plegable Molecular biology.
Plegable Molecular biology.
 
Plegable jonathan cardona vélez
Plegable jonathan cardona vélezPlegable jonathan cardona vélez
Plegable jonathan cardona vélez
 
Isabela Palacio Jaramillo 3 semestre
Isabela Palacio Jaramillo 3 semestre Isabela Palacio Jaramillo 3 semestre
Isabela Palacio Jaramillo 3 semestre
 
cronier 2012
cronier 2012cronier 2012
cronier 2012
 
SSC4a Report (final)
SSC4a Report (final)SSC4a Report (final)
SSC4a Report (final)
 
hinckley poster5 (2)
hinckley poster5 (2)hinckley poster5 (2)
hinckley poster5 (2)
 
Taxanes induced Neuropathy
Taxanes induced Neuropathy Taxanes induced Neuropathy
Taxanes induced Neuropathy
 
Sepsis primer
Sepsis primerSepsis primer
Sepsis primer
 
"Bioluminescent Imaging of Histidyl-Transfer RNA Synthetase-Induced Myositis ...
"Bioluminescent Imaging of Histidyl-Transfer RNA Synthetase-Induced Myositis ..."Bioluminescent Imaging of Histidyl-Transfer RNA Synthetase-Induced Myositis ...
"Bioluminescent Imaging of Histidyl-Transfer RNA Synthetase-Induced Myositis ...
 
Approcci bioenergetici per la neuroprotezione nella malattia parkinsoniana
Approcci bioenergetici per la neuroprotezione nella malattia parkinsonianaApprocci bioenergetici per la neuroprotezione nella malattia parkinsoniana
Approcci bioenergetici per la neuroprotezione nella malattia parkinsoniana
 
Topic of the month...Paraneoplastic syndromes
Topic of the month...Paraneoplastic syndromesTopic of the month...Paraneoplastic syndromes
Topic of the month...Paraneoplastic syndromes
 

MSRF.finalpaper.pketch.abstractedit

  • 1. Mechanisms by which pro-inflammatory cytokines and anti-heterogeneous nuclear ribonuclear protein A1-M9 (anti-hnRNP A1-M9) antibodies cause neurodegeneration in multiple sclerosis Research proposal: NIH Medical Student Research Fellowship Program Peter Ketch 1177 Union Ave, #302 Memphis, TN 38104 (214) 901-4525 pketch@uthsc.edu University of Tennessee Health Science Center College of Medicine, Class of 2019 Mentor: Dr. Michael C. Levin, MD Professor, Department of Neurology & Anatomy Neurobiology University of Tennessee Health Science Center Chief, Neurology Service, VAMC-Memphis Director, Multiple Sclerosis Center Laboratory for Viral and Demyelinating Diseases 855 Madison Avenue, Room 415 Memphis, TN 38163 (901) 448-2243 mlevin@uthsc.edu May 31, 2016 – August 16, 2016 Memphis VA Medical Center
  • 2. Abstract Multiple sclerosis (MS) is a chronic inflammatory disease of the central nervous system (CNS) with hallmark demyelination and neurodegeneration. Data suggest that Th1 and Th17 CD4+ lymphocytes and their cytokines are thought to play a role in pathogenesis of neurodegeneration seen in MS. Neuronal exposure to a mixture of these pro-inflammatory cytokines causes mislocalization of heterogeneous nuclear ribonuclear protein A1 (hnRNP A1), which accumulates in the cytoplasm. Importantly, MS patients develop an antibody against hnRNP A1. Similar experiments demonstrate that anti-hnRNP A1-M9 antibodies induce similar molecular events, in addition to stress granule (SG) formation and changes in RNA metabolism of clinically relevant RNAs. We hypothesize that an individual or combinations of pro-inflammatory Th17 cytokines can influence molecular events important to the pathogenesis of MS, including mislocalization of hnRNP A1 and the formation of SGs. In vitro experiments with SK-N-SH cells demonstrate that select individual and combinations of Th17 cytokines (IL-17A, IL-23, IL-1β, TGF-β, and IL-6) can induce the formation of SGs, mislocalization of hnRNP A1, and colocalization of hnRNP A1 with a SG marker, Poly(A)-binding protein (PABP). Taken together, these data suggest a potential role of pro-inflammatory Th17 cytokines in the pathogenesis of neurodegeneration through an hnRNP A1 mechanism seen in MS.
  • 3. I. Introduction Multiple sclerosis (MS) is an immune-mediated inflammatory disease that results in demyelination of the central nervous system (CNS). The neurological disability seen in patients with MS is highly variable, but can progress irreversibly in association with neuronal and axonal damage, or neurodegeneration. Neurodegeneration in the CNS has been shown to contribute to the pathogenesis of MS, but was thought to be secondary to the disease’s hallmark inflammatory demyelination. However, neurodegeneration can occur independently of demyelinating plaque formation and is now viewed a critical component of the long-term neurological disability seen in MS, especially progressive forms. In fact, a number of studies demonstrate a significant reduction in nerve fiber density in corticospinal tracts and dorsal columns, independent of plaque formation2,8-22. Although demyelination and its associated inflammatory processes are well described, the pathophysiological mechanisms of neurodegeneration in MS are not well understood. Evidence of neuronal and axonal degeneration has been shown in the brains and spinal cords of patients with MS, although specific mechanisms are not clearly indicated and appear to be multifactorial9-11,23-25. RNA binding proteins (RBPs) belong to a diverse class of proteins with varying functions including RNA transcription, transport and translation. An important function of some of these RBPs, including heterogeneous nuclear ribonuclearprotein A1 (hnRNP A1), is transport of mRNA from the nucleus to the cytoplasm. Extensive studies in Dr. Levin’s lab have demonstrated that MS patients develop anti-hnRNP A1 antibodies and that these antibodies cause neurodegeneration3,4,21,33,35,37. Normally functioning hnRNP A1 binds mRNA and transportin in the nucleus, transports mRNA to the cytoplasm via a nuclear pore, then quickly translocates back into nucleus4. The M9 region of hnRNP A1 contains a sequence that binds transportin. When antibodies bind to this region, hnRNP A1 cannot return to the nucleus. The presence of autoantibodies to M9 in patients with MS causes hnRNP A1 to accumulate in the cytoplasm, instead of the nucleus where it is usually observed in highest quantity4,32. Furthermore, this accumulation corresponds to reduced levels of ATP and increased levels of apoptosis, important markers of neurodegeneration4. These findings demonstrate an important association between anti-A1-M9 antibodies and neurodegeneration in MS.
  • 4. Anti-hnRNPA1-M9 antibodies can induce stress granule (SG) formation in neurons and can cause changes in RNA metabolism of the hnRNP A1 protein’s RNA cargo44. During times of cellular stress, SGs contain dysfunctional RBPs and their translationally repressed mRNA cargo45. Studies in Dr. Levin’s lab demonstrated that adding anti-A1-M9 antibodies to neuronal cell lines in culture resulted in SG formation and that the antibodies colocalize with the SGs44. In previous studies, Dr. Levin’s lab showed that the levels of specific genes, including SPG4 (spastin) and SPG7 (paraplegin), were altered in response to transfection of neuronal cells with anti- hnRNP A1-M9 antibodies. RNA immunoprecipitation studies revealed that SPG4 and SPG7 (to a lesser extent) bound hnRNPA1 (Fig SA2-4). Furthermore, Western blots showed that anti-hnRNP A1-M9 antibodies caused a profound reduction in SPG4 and SPG7 protein levels3. This experiment demonstrated that anti-hnRNPA1-M9 antibodies specifically altered the translation of clinically relevant RNAs that were shown to bind to A1. In addition to antibody-mediated immunoreactivity, there are other factors, like pro-inflammatory cytokines, that may act in conjunction to produce the neurodegeneration seen in MS. Different subtypes of MS display distinct inflammatory activity. Early forms of MS (acute and relapsing-remitting) are characterized by Th1 and Th17 CD4+ lymphocyte responses and demyelination and neuronal injury correlated with T and B cells11,14,26,29-31. More diffuse CNS inflammation and IgG-positive Fig SA2-4. RNAImmunoprecipitation andWesternblot analyses. SK-N-SHcells lysateswere incubated with agarose beads labeled anti-A1-M9 or control abs. A. Western blots showed thathnRNP A1 waspresentin the lysate (L) and the anti-A1-M9 IP (A1) butnotin control IgG IP (IgG). B. RNA wasisolated from protein eluents and amplified by RT-PCR. Values were normalizedto GAPDH. Results revealed thathnRNP A1 and SPG4 are RNA binding partners ofhnRNP A1 and SPG-7 showed a positive trend towardsRNA binding. p≤0.05 analyzed by t- test. C. Anti-hnRNP A1 antibodiesalterproteinlevels as measuredby WesternBlot. SK-N-SHcells were treated with anti-A1-M9 abs or control IgG and proteins run on Western blots. Results revealed hnRNP A1, SPG4,and SPG7 had markedly reducedprotein levelsin anti-hnRNP A1-M9 antibody treated cells compared to controls. There was no change in Beta actin or SPG20. Fig SA2-5A: In primary neurons in culture, A1 mis-localizes to the cytoplasm when exposed to Th1and Th17 cytokines. Under normal conditions (0), hnRNP A1 (green) co- localizes in the nucleus (DAPI-blue). When exposed to Th1 or Th17 cytokines (see Table below – column labeled ‘C’), hnRNP A1 mislocalizes to the cytoplasm (arrows). Arrowheads identify cell bodies of neurons.
  • 5. plasma cells predominate in progressive forms of MS14. One specific study showed that neurodegeneration of specific neural pathways was directly associated with T-cell recruitment5. Other studies have demonstrated that both CD4+ Th1 and Th17 lymphocytes induced disease6,7,27,28. Preliminary studies in Dr. Levin’s lab, using a mixture of both Th cytokine inducers (priming cytokines) and effectors (cytokine products), demonstrated that both Th1 and Th17 cytokines caused mislocalization of A1 from the nucleus to the cytoplasm (Fig SA2-5A). II. Hypothesis We hypothesize that an individual or combinations of pro-inflammatory Th17 cytokines can influence molecular events important to the pathogenesis of MS. III. Specific Aims The objective is to determine the mechanistic role that pro-inflammatory Th17 cytokines play in the mislocalization of hnRNP A1 and stress granule formation in SK-N-SH cells. 1. Use immunocytochemistry to determine whether individual or combinations of cytokines cause mislocalization of hnRNP A1 2. Use immunocytochemistry to determine whether individual or combinations of cytokines induce stress granule formation IV. Materials and Methods Cells The SK-N-SH cell line (ATCC, HTB-11), an immortalized human neuroblastoma cell line was maintained in Dulbecco’s modified Eagle’s medium/F12 supplemented with 10% fetal bovine serum and 1% penicillin-streptomycin antibiotics. 105 SK-N-SH cells were seeded per well into 500 uL of complete DMEM/F12 media into 8 well chamber slides (Corning #354632) for immunocytochemistry experiments. Exposure of SK-N-SH cells to cytokines in vitro Anti-hnRNP A1 antibodies were obtained from Millipore (04-1469). Poly(A)-binding protein (PABP) antibodies were obtained from abcam (ab-21060). The anti-hnRNP A1 antibodies are specific for hnRNP
  • 6. A1-M9 and have been shown to overlap the M9 immunodominant epitope recognized by IgG isolated from MS patients3. Cytokines were added to the culture media of SK-N-SH cells in the following concentrations, individually and in combination, represented in Figure 1: IL-17A (20ng/ml), IL-23 (20ng/ml), IL-1β (20 ng/ml), TGF-β (2 ng/ml), IL-6 (25 ng/ml) . Immunocytochemistry of hnRNP A1-M9 mislocalization and stress granule formation 105 SK-N-SH cells were seeded per well into 500 uL of complete DMEM/F12 media into 8 well chamber slides (Corning #354632). 48 hours following cytokine addition, cells were fixed with 4% paraformaldehyde for 15 minutes at room temperature (RT). Fixed cells were then blocked and permeabilized with 6% Milk-PBS + 0.4% TritonX-100. Cells were then labeled for stress granules using Poly(A)-binding protein (PABP) and hnRNP A1 at the manufacturer’s suggested concentration overnight at 4°C. Following primary antibody incubation, cells were washed 3 X 5 minutes with PBS-T. Cells were treated with secondary anti-mouse conjugated FITC and anti-rabbit conjugated Texas Red at manufacturer’s suggested concentration for 2 hours at RT. Cells were washed 3 X 5 minutes with PBS-T, 2 X 5 minutes with PBS and mounted with DAPI mounting medium, and imaged by dual confocal microscopy. VII. Results Untreated SK-N-SH cells show nuclear localization of hnRNP-A1 (Figures 1-7, top) and no stress granule formation. After treatment with Th17 effector cytokines (IL-17A, IL-23, IL-1β), hnRNP-A1 is mislocalized to the cytoplasm where it colocalizes with stress granules (Figure 2, bottom, arrows). However not all stress granules colocalized with hnRNP-A1 (Figure 2, bottom, circles). Following exposure to IL-1β, SK-N-SH cells show hnRNP-A1 mislocalization and hnRNP-A1 colocalization with stress granules (Figure 3, bottom, arrows). SK-N-SH cells treated with IL-23 showed mislocalization of hnRNP-A1 to the
  • 7. cytoplasm and the formation of stress granules; however hnRNP-A1 does not colocalize with these granules (Figure 4, bottom). After treatment with IL-17A, hnRNP-A1 is mislocalized to the cytoplasm. PABP staining revealed several small, punctuated areas of staining suggestive of stress granules, but further studies are warranted to clarify whether or not IL-17A treatment induces stress granules in SK-N-SH cells (Figure 5, bottom). Furthermore, hnRNP-A1 did not colocalize with PABP staining in IL-17A treated SK-N-SH cells. Th17 inducer cytokine (TGF-β, IL-6) treated SK-N-SH cells show decreased localization to the nucleus compared to untreated cells and possible mislocalization, although high background in this immunofluorescence image warrants further experimentation to confirm this finding (Figure 6, bottom). hnRNP-A1 did not colocalize with the stress granules formed in these Th17 inducer cytokine treated cells (Figure 6, bottom). Following exposure to TGF-β, SK-N-SH cells show hnRNP-A1 mislocalization, the formation of stress granules, and hnRNP-A1 colocalization with these stress granules (Figure 7, bottom, arrows). After treatment with IL-6, there was mislocalization of hnRNP-A1 to the cytoplasm and the formation of stress granules; however hnRNP-A1 does not colocalize with these granules (Figure 8, bottom). A summary table detailing hnRNP-A1 mislocalization, stress granule formation and colocalization of hnRNP-A1 with stress granules for each cytokine treatment group is provided (Figure 9).
  • 8.
  • 9.
  • 10. VIII. Conclusion In vitro experiments with SK-N-SH cells demonstrate that select individual and combinations of Th17 cytokines (IL-17A, IL-23, IL-1β, TGF-β, and IL-6) can influence molecular events important to the pathogenesis of MS including, mislocalization of hnRNP A1 (accumulation in the cytoplasm) and formation of SGs. Furthermore, select individual and combinations of pro-inflammatory cytokines caused colocalization of hnRNP A1 and PABP, a stress granule marker. Taken together, these data suggest a potential role of pro-inflammatory Th17 cytokines in the pathogenesis of neurodegeneration in MS via an hnRNP A1 mechanism. Previous studies have shown that anti-hnRNP A1-M9 antibodies in patients with MS cause hnRNP A1 to accumulate in the cytoplasm, which may cause cellular stress and resulting SG formation44. This study showed that similar molecular changes occur in response to select individual and combinations of Th17 pro-inflammatory cytokines. These findings suggest that both antibody-mediated immunoreactivity and pro-inflammatory cytokines may share a potential mechanism that might contribute to neurodegeneration seen in MS, involving mislocalization and accumulation of hnRNP A1 and SG formation. The exact mechanism by which this occurs requires further
  • 11. study. Studies should be conducted involving simultaneous exposure to anti-hnRNP A1 antibodies and pro- inflammatory cytokines to more closely mimic in vivo conditions and to further explore the role of Th17 pro- inflammatory cytokines in the pathogenesis of neurodegeneration in MS. IX. Potential Conflicts Dr. Levin is the owner of a patent: “A biomarker for neurodegeneration in neurological disease.” X. References 1. Lee, S. and M. Levin, Novel somatic single nucleotide variants within the RNA binding protein hnRNP A1 in multiple sclerosis patients [v2; ref status: indexed]. Vol. 3. 2014. 2. Friese, M.A., B. Schattling, and L. Fugger, Mechanisms of neurodegeneration and axonal dysfunction in multiple sclerosis. Nat Rev Neurol, 2014. 10(4): p. 225-38. 3. Lee, S., L. Xu, Y. Shin, L. Gardner, A. Hartzes, F.C. Dohan, C. Raine, R. Homayouni, and M.C. Levin, A potential link between autoimmunity and neurodegeneration in immune-mediated neurological disease. J Neuroimmunol, 2011. 235(1-2): p. 56-69. 4. Douglas, J., L. Gardner, and M.C. Levin, Antibodies to an intracellular antigen penetrate neuronal cells and cause deleterious effects. J Clinical & Cellular Immunology 2013. 4(1): p. 134. 5. Brown, D.A. and P.E. Sawchenko, Time course & distribution of inflammatory & neurodegenerative events suggest structural bases for the pathogenesis of experimental autoimmune encephalomyelitis. J Comp Neurol, 2007. 502:236-60. 6. Arima, Y., M. Harada, D. Kamimura, J.H. Park, F. Kawano, F.E. Yull, T. Kawamoto, Y. Iwakura, U.A. Betz, G. Marquez, T.S. Blackwell, Y. Ohira, T. Hirano, and M. Murakami, Regional neural activation defines a gateway for autoreactive T cells to cross the blood-brain barrier. Cell, 2012. 148(3): p. 447-57. 7. Kamimura, D., M. Yamada, M. Harada, L. Sabharwal, J. Meng, H. Bando, H. Ogura, T. Atsumi, Y. Arima, and M. Murakami, The gateway theory: bridging neural and immune interactions in the CNS. Front Neurosci, 2013. 7: p. 204. 8. Trapp, B.D. and K.A. Nave, MS: an immune or neurodegenerative disorder? Annu Rev Neurosci, 2008. 31: p. 247-69. 9. Lassmann, H., MS: is there neurodegeneration independent from inflammation? J Neurol Sci, 2007. 259(1-2): p. 3-6. 10. Lassmann, H. and C.F. Lucchinetti, Cortical demyelination in CNS inflammatory demyelinating diseases. Neurology, 2008. 70(5): p. 332-3. 11. Lassmann, H. and J. van Horssen, The molecular basis of neurodegeneration in multiple sclerosis. FEBS Lett, 2011. 585(23): p. 3715-23. 12. Lucchinetti, C.F., B.F. Popescu, R.F. Bunyan, N.M. Moll, S.F. Roemer, H. Lassmann, W. Bruck, J.E. Parisi, B.W. Scheithauer, C. Giannini, S.D. Weigand, J. Mandrekar, and R.M. Ransohoff, Inflammatory cortical demyelination in early multiple sclerosis. N Engl J Med, 2011. 365(23): p. 2188-97. 13. Peterson, J.W. and B.D. Trapp, Neuropathobiology of multiple sclerosis. Neurol Clin, 2005. 23(1): p. 107-29, vi-vii. 14. Frischer, J.M., S. Bramow, A. Dal-Bianco, C.F. Lucchinetti, H. Rauschka, M. Schmidbauer, H. Laursen, P.S. Sorensen, and H. Lassmann, The relation between inflammation and neurodegeneration in multiple sclerosis brains. Brain, 2009. 132(Pt 5): p. 1175-89. 15. Fisniku, L.K., D.T. Chard, J.S. Jackson, V.M. Anderson, D.R. Altmann, K.A. Miszkiel, A.J. Thompson, and D.H. Miller, Gray matter atrophy is related to long-term disability in multiple sclerosis. Ann Neurol, 2008. 64(3): p. 247-54.
  • 12. 16. Geurts, J.J., Is progressive multiple sclerosis a gray matter disease? Ann Neurol, 2008. 64(3): p. 230- 2. 17. Ganter, P., C. Prince, and M.M. Esiri, Spinal cord axonal loss in multiple sclerosis: a post-mortem study. Neuropathol Appl Neurobiol, 1999. 25(6): p. 459-67. 18. Lovas, G., N. Szilagyi, K. Majtenyi, M. Palkovits, and S. Komoly, Axonal changes in chronic demyelinated cervical spinal cord plaques. Brain, 2000. 123 ( Pt 2): p. 308-17. 19. DeLuca, G.C., K. Williams, N. Evangelou, G.C. Ebers, and M.M. Esiri, The contribution of demyelination to axonal loss in multiple sclerosis. Brain, 2006. 129(Pt 6): p. 1507-16. 20. Evangelou, N., G.C. DeLuca, T. Owens, and M.M. Esiri, Pathological study of spinal cord atrophy in multiple sclerosis suggests limited role of local lesions. Brain, 2005. 128(Pt 1): p. 29-34. 21. Levin, M.C., S. Lee, L. Gardner, Y. Shin, J.N. Douglas, and C. Groover, Pathogenic mechanisms of neurodegeneration based on the phenotypic expression of progressive forms of immune-mediated neurologic disease. Degenerative Neurological and Neuromuscular Disease, 2012. 2: p. 175-187. 22. Levin, M.C., J. Douglas, L. Myers, S. Lee, Y. Shin, and L. Gardner, Neurodegeneration in multiple sclerosis involves multiple pathogenic mechanisms. Degenerative Neurological and Neuromuscular Disease, 2014. 4: p. 49-63. 23. Dutta, R. and B.D. Trapp, Pathogenesis of axonal and neuronal damage in multiple sclerosis. Neurology, 2007. 68(22 Suppl 3): p. S22-31; discussion S43-54. 24. Benarroch, E.E., Acquired axonal degeneration and regeneration: Recent insights and clinical correlations. Neurology, 2015. 84(20): p. 2076-85. 25. Coleman, M., Axon degeneration mechanisms: commonality amid diversity. Nat Rev Neurosci, 2005. 6(11): p. 889-98. 26. Dutta, R. and B. Trapp, Mechanisms of neuronal dysfunction and degeneration in multiple sclerosis. Progress in Neurobiology, 2011. 93: p. 1-12. 27. Siffrin, V., H. Radbruch, R. Glumm, R. Niesner, M. Paterka, J. Herz, T. Leuenberger, S.M. Lehmann, S. Luenstedt, J.L. Rinnenthal, G. Laube, H. Luche, S. Lehnardt, H.J. Fehling, O. Griesbeck, and F. Zipp, In vivo imaging of partially reversible th17 cell-induced neuronal dysfunction in the course of encephalomyelitis. Immunity, 2010. 33(3): p. 424-36. 28. Tanabe, S. and T. Yamashita, Repulsive guidance molecule-a is involved in Th17-cell-induced neurodegeneration in autoimmune encephalomyelitis. Cell Rep, 2014. 9(4): p. 1459-70. 29. Steinman, L., Mixed results with modulation of TH-17 cells in human autoimmune diseases. Nat Imm, 2010. 11(1): 41-4. 30. Antel, J., S. Antel, Z. Caramanos, D.L. Arnold, and T. Kuhlmann, Primary progressive multiple sclerosis: part of the MS disease spectrum or separate disease entity? Acta Neuropathol, 2012. 123(5): p. 627-38. 31. Beecham, et al. Analysis of immune-related loci identifies 48 new susceptibility variants for multiple sclerosis. Nat Genet, 2013. 45(11): p. 1353-60. 32. Levin, M.C., S. Lee, L.A. Gardner, Y. Shin, J.N. Douglas, and C. Cooper, Autoantibodies to Non- myelin Antigens as Contributors to the Pathogenesis of Multiple Sclerosis. J Clin Cell Immunol, 2013. 4. 33. Levin, M., T. Lehky, N. Flerlage, D. Katz, D. Kingma, E. Jaffe, J. Heiss, N. Patronas, H. McFarland, and S. Jacobson, Immunopathogenesis of HTLV-1 associated neurologic disease based on a spinal cord biopsy from a patient with HTLV-1 associated myelopathy/tropical spastic paraparesis (HAM/TSP). New Eng J Med, 1997. 336: p. 839-845. 34. Levin, M., M. Krichavsky, J. Berk, S. Foley, M. Rosenfeld, J. Dalmau, G. Chen, J. Posner, and S. Jacobson, Neuronal molecular mimicry in immune mediated neurologic disease. Ann Neurol, 1998. 44(1): p. 87-98. 35. Levin, M.C., S.M. Lee, F. Kalume, Y. Morcos, F.C. Dohan, Jr., K.A. Hasty, J.C. Callaway, J. Zunt, D. Desiderio, and J.M. Stuart, Autoimmunity due to molecular mimicry as a cause of neurological disease. Nat Med, 2002. 8(5): p. 509-13.
  • 13. 36. Lee, S.M., Y. Morocos, H. Jang, J.M. Stuart, M.C. Levin, HTLV-1 induced molecular mimicry in neurologic disease,In: Molecular Mimicry: Infection Inducing Autoimmune Disease, M. Oldstone, Editor. 2005, Springer: New York. 37. Lee, S., M.C. Levin, Molecular mimicry in neurological disease: what is the evidence? Cell Mol Life Sci, 2008. 65:.1161-75. 38. Lee, S.M., F.D. Dunnavant, H. Jang, J. Zunt, and M.C. Levin, Autoantibodies that recognize functional domains of hnRNPA1 implicate molecular mimicry in the pathogenesis of neurological disease. Neurosci Lett, 2006. 401(1-2): p. 188-93. 39. Kalume, F., S.M. Lee, Y. Morcos, J.C. Callaway, and M.C. Levin, Molecular mimicry: cross-reactive antibodies from patients with immune-mediated neurologic disease inhibit neuronal firing. J Neurosci Res, 2004. 77(1): p. 82-9. 40. Douglas, J.N., L.A. Gardner, S. Lee, Y. Shin, C.J. Groover, and M.C. Levin, Antibody transfection into neurons as a tool to study disease pathogenesis. J Vis Exp, 2012(67). 41. Gascon, E., L. Vutskits, and J.Z. Kiss, Polysialic acid-neural cell adhesion molecule in brain plasticity: from synapses to integration of new neurons. Brain Res Rev, 2007. 56(1): p. 101-18. 42. Levin, M.C., et al., Organization of galanin-immunoreactive inputs to the paraventricular nucleus with special reference to their relationship to catecholaminergic afferents. J Comp Neurol, 1987. 261(4): p. 562-82. 43. Meyers, L., C.J. Groover, J. Douglas, S. Lee, D. Brand, M.C. Levin, and L.A. Gardner, A role for Apolipoprotein A-I in the pathogenesis of multiple sclerosis. J Neuroimmunol, 2014. 277(1-2): p. 176-85. 44. Levin, M., Lee, S., Gardner, L., Douglas, J., Shin, Y., Sawchenko, P., Lalaor, S., Segal, B, Contribution of a dysfunctional RNA binding protein, hnRNP A1, to neurodegeneration in MS. Neurology, 2014. 82(10). 45. Kiebler, M.A., Bassell, G.J, Neuronal RNA granules: movers and makers. Neuron, 2006. 51(6): p685-90.