Iron and neurodegeneration in the multiple sclerosis brain

Simon Hametner, Isabella Wimmer, Lukas Haider, Sabine Pfeifenbring, Wolfgang Brück, Hans Lassmann, Simon Hametner, Isabella Wimmer, Lukas Haider, Sabine Pfeifenbring, Wolfgang Brück, Hans Lassmann

Abstract

Objective: Iron may contribute to the pathogenesis and progression of multiple sclerosis (MS) due to its accumulation in the human brain with age. Our study focused on nonheme iron distribution and the expression of the iron-related proteins ferritin, hephaestin, and ceruloplasmin in relation to oxidative damage in the brain tissue of 33 MS and 30 control cases.

Methods: We performed (1) whole-genome microarrays including 4 MS and 3 control cases to analyze the expression of iron-related genes, (2) nonheme iron histochemistry, (3) immunohistochemistry for proteins of iron metabolism, and (4) quantitative analysis by digital densitometry and cell counting in regions representing different stages of lesion maturation.

Results: We found an age-related increase of iron in the white matter of controls as well as in patients with short disease duration. In chronic MS, however, there was a significant decrease of iron in the normal-appearing white matter (NAWM) corresponding with disease duration, when corrected for age. This decrease of iron in oligodendrocytes and myelin was associated with an upregulation of iron-exporting ferroxidases. In active MS lesions, iron was apparently released from dying oligodendrocytes, resulting in extracellular accumulation of iron and uptake into microglia and macrophages. Iron-containing microglia showed signs of cell degeneration. At lesion edges and within centers of lesions, iron accumulated in astrocytes and axons.

Interpretation: Iron decreases in the NAWM of MS patients with increasing disease duration. Cellular degeneration in MS lesions leads to waves of iron liberation, which may propagate neurodegeneration together with inflammatory oxidative burst.

© 2013 The Authors. Annals of Neurology published by Wiley Periodicals, Inc. on behalf of the American Neurological Association.

Figures

Figure 1
Figure 1
Correlation of whole tissue iron with age (A) and disease duration (B). (A) Whole tissue iron, as quantified by digital optical densitometry, correlated with age (in years) at the nonlesioned subcortical white matter (WM). Lines represent separate linear regressions of controls and multiple sclerosis (MS) cases. (B) Whole tissue iron is subtracted from the iron levels predicted by the age-dependent linear regression equation derived from controls (continuous line in A). There is a significant correlation between the reduction of iron load and disease duration. Furthermore, a significant negative partial correlation between whole tissue iron and disease duration (in months) was found when pooling all data points and setting disease duration of controls to zero (rpartial = −0.497, p < 0.001). Analysis of whole tissue iron in the deep WM led to comparable results (not shown). AMS = acute MS; RRMS = relapsing–remitting MS.
Figure 2
Figure 2
Iron and the expression of iron-related proteins in the white matter (WM) of controls and multiple sclerosis (MS) patients. (A) Luxol fast blue–periodic acid Schiff myelin staining (blue) and (B) total nonheme iron (brown) of consecutive double-hemispheric slides of MS Subject 25. High iron load is visible in the basal ganglia and the leukocortical boundary. Multiple demyelinated WM lesions, which harbor less iron than the normal-appearing WM (NAWM), are indicated by arrows. (C–Q) Presence of total nonheme iron and iron-related proteins in the normal WM (NWM) of controls (left panel of images; C, F, I, L, O), in the NAWM of MS patients (middle panel; D, G, J, M, P), and in the periplaque WM (PPWM) of MS patients close to the edge of an active lesion (right panel; E, H, K, N, Q). (C–E) In the NWM (C), iron is mainly present in myelin and cells with oligodendrocytic morphology (inset in C) and is reduced in the NAWM of MS patients (D). Inset in D shows small round cells indicative of oligodendrocytes and a larger cell indicative of activated microglia morphology. Close to active plaques, iron is largely lost from oligodendrocytes and myelin, but is present in activated microglia cells (E). (F–H) Immunohistochemistry for ferritin (brown) shows a similar cellular distribution as iron; however, more cells are detected with ferritin immunohistochemistry than with total nonheme iron staining. (I–N) Hephaestin (blue) is expressed in the NWM in oligodendrocytes (I; oligodendrocyte marker TPPP/p25 = red in I–K) and in astrocytes (L; astrocyte marker glial fibrillary acidic protein = red in L–N). In the MS NAWM, both hephaestin+ oligodendrocytes (J) and astrocytes (M) are reduced. In the PPWM close to active lesions, hephaestin is upregulated in relation to the NAWM in oligodendrocytes (K), but not in astrocytes (N). (O–Q) Ceruloplasmin (brown) is mainly observed in astrocytes. Its expression is consistently low in the NWM (O), elevated in the NAWM of some MS patients (P), and consistently strong at the edge of active lesions (Q). AG = astroglia; OG = oligodendroglia; scale bars = 100μm; inset scale bars = 10μm (C, I–N) or 25μm (D–H).
Figure 3
Figure 3
Iron and iron-related molecules in different types of MS lesions. (A–F) There are 3 different types of MS lesions regarding iron content. (A, D) In slowly expanding lesions with demyelinating activity at the lesion edge, and less frequently and less pronounced in inactive lesions, a rim of iron within microglia and macrophages is seen at the lesion edge. The iron content within the lesion is reduced, but perivascular accumulation of iron is occasionally seen within lesions. (A) Proteolipid protein (PLP) immunohistochemistry (IHC). (D) Total nonheme iron staining. (B, E) Some classic late active MS lesions contain more iron compared to the normal-appearing white matter (NAWM). The iron is found within macrophages, astrocytes, and axons in these lesions. (B) Luxol fast blue–periodic acid Schiff myelin staining (blue). (E) Total nonheme iron staining. (C, F) In the majority of MS lesion, iron is reduced compared to the NAWM, and total nonheme iron staining closely matches the staining for myelin. (C) PLP IHC. (F) Total nonheme iron staining. (G–U) Iron and iron-related proteins are shown in early active lesions (left panel; G, J, M, P, S), late active lesions (middle panel; H, K, N, Q, T), and inactive lesions (right panel; I, L, O, R, U). The figure documents an extreme example (MS Subject 5, who developed acute MS at the age of 78 years; the lesions formed in a brain with a high age-related iron load). Lesional activity was defined according to Brück et al. In early active lesions, total nonheme iron (G, brown) is mainly seen in the cytoplasm of macrophages and microglia and to a lesser extent as fine extracellular granules. In contrast, ferrous nonheme iron (J, brown) is largely detected as extracellular granules or in lysosomes or endosomes of macrophages and microglia. Ferritin expression (M, brown) additionally reflects the massive macrophage and microglia activation. Few oligodendrocytes (P; TPPP/p25 = red in P–R) and many astrocytes (S; glial fibrillary acidic protein [GFAP] = red in S to U) express hephaestin (blue in P–U) in this early active lesion. In late active lesions, there is a shift of iron-containing macrophages toward the perivascular space (H). Extracellular total (H, brown) and ferrous nonheme iron (K, brown) are sparse in comparison to early active lesions. Ferritin (N; brown) is predominantly expressed in macrophages. Oligodendrocytes are largely lost (Q), and there is sparse hephaestin expression in astrocytes (T). In inactive lesions, total nonheme iron is seen in few scattered astrocytes (I), whereas ferrous nonheme iron (L) is not detectable. Ferritin expression (O; brown) is only seen in some cells, predominantly astrocytes. Oligodendrocytes are lost from the lesions (R), and there is profound fibrillary gliosis (U). Hephaestin expression is minor or absent (R, U). (V) Confocal laser microscopy shows the expression of hephaestin (green) in oligodendrocytes (carbonic anhydrase II = red) and astrocytes (GFAP = blue). In this area of MS periplaque white matter, hephaestin is detected predominantly in oligodendrocytes. In active lesions (W–Y), ceruloplasmin (green in W–Y) is detected mainly in astrocytes (W; GFAP = red in W) and axons (X). However, in many cells and axons, ceruloplasmin is colocalized with fibrin (red in X and Y), suggesting nonspecific uptake of soluble serum ceruloplasmin from the extracellular space under conditions of severe blood–brain barrier damage. Nonetheless, some cells with astrocyte morphology are stained for ceruloplasmin in the absence of fibrin reactivity, suggesting autochthonous expression of ceruloplasmin in at least some astrocytes. AG = astroglia; OG = oligodendroglia; scale bars = 4mm (A–F), 100μm (G–U); inset scale bars = 10μm.
Figure 4
Figure 4
Dystrophic microglia, axonal iron, and oxidized phospholipids in multiple sclerosis (MS) lesions. Iron-loaded microglia and macrophages in active MS lesions show signs of degeneration (dystrophy) with process beading, retraction, and fragmentation (A), which is also visible in microglia stained for ferritin light polypeptide (FTL; B, brown). At active lesion edges, total (C) and rarely also ferrous nonheme iron (D, brown) accumulates in axons. (E–H) Oxidized phospholipids (E06 reactivity; E and G; brown) are detected in lesions with high iron content (total nonheme iron staining in F and H). Scale bars = 20μm (A, B); 100μm (C, D); 200μm (E, F); 75μm (G, H).

References

    1. Frischer JM, Bramow S, Dal-Bianco A, et al. The relation between inflammation and neurodegeneration in multiple sclerosis brains. Brain. 2009;132(pt 5):1175–1189.
    1. Comi G, Jeffery D, Kappos L, et al. Placebo-controlled trial of oral laquinimod for multiple sclerosis. N Engl J Med. 2012;366:1000–1009.
    1. Lassmann H, van Horssen J, Mahad D. Progressive multiple sclerosis: pathology and pathogenesis. Nat Rev Neurol. 2012;8:647–656.
    1. Khalil M, Langkammer C, Ropele S, et al. Determinants of brain iron in multiple sclerosis: a quantitative 3T MRI study. Neurology. 2011;77:1691–1697.
    1. Singh AV, Zamboni P. Anomalous venous blood flow and iron deposition in multiple sclerosis. J Cereb Blood Flow Metab. 2009;29:1867–1878.
    1. Hallgren B, Sourander P. The effect of age on the non-haemin iron in the human brain. J Neurochem. 1958;3:41–51.
    1. Confavreux C, Vukusic S, Moreau T, Adeleine P. Relapses and progression of disability in multiple sclerosis. N Engl J Med. 2000;343:1430–1438.
    1. Connor JR, Menzies SL. Cellular management of iron in the brain. J Neurol Sci. 1995;134(suppl):33–44.
    1. Todorich B, Pasquini JM, Garcia CI, et al. Oligodendrocytes and myelination: the role of iron. Glia. 2009;57:467–478.
    1. Kell DB. Iron behaving badly: inappropriate iron chelation as a major contributor to the aetiology of vascular and other progressive inflammatory and degenerative diseases. BMC Med Genomics. 2009;2:2.
    1. Roede JR, Jones DP. Reactive species and mitochondrial dysfunction: mechanistic significance of 4-hydroxynonenal. Environ Mol Mutagen. 2010;51:380–390.
    1. Murphy MP. How mitochondria produce reactive oxygen species. Biochem J. 2009;417:1–13.
    1. Fischer MT, Sharma R, Lim JL, et al. NADPH oxidase expression in active multiple sclerosis lesions in relation to oxidative tissue damage and mitochondrial injury. Brain. 2012;135(pt 3):886–899.
    1. Campbell GR, Ziabreva I, Reeve AK, et al. Mitochondrial DNA deletions and neurodegeneration in multiple sclerosis. Ann Neurol. 2011;69:481–492.
    1. Dutta R, McDonough J, Yin X, et al. Mitochondrial dysfunction as a cause of axonal degeneration in multiple sclerosis patients. Ann Neurol. 2006;59:478–489.
    1. Ziabreva I, Campbell G, Rist J, et al. Injury and differentiation following inhibition of mitochondrial respiratory chain complex IV in rat oligodendrocytes. Glia. 2010;58:1827–1837.
    1. Craelius W, Migdal MW, Luessenhop CP, et al. Iron deposits surrounding multiple sclerosis plaques. Arch Pathol Lab Med. 1982;106:397–399.
    1. Adams CW. Perivascular iron deposition and other vascular damage in multiple sclerosis. J Neurol Neurosurg Psychiatry. 1988;51:260–265.
    1. LeVine SM. Iron deposits in multiple sclerosis and Alzheimer’s disease brains. Brain Res. 1997;760:298–303.
    1. Breitschopf H, Suchanek G, Gould RM, et al. In situ hybridization with digoxigenin-labeled probes: sensitive and reliable detection method applied to myelinating rat brain. Acta Neuropathol. 1992;84:581–587.
    1. Barnett MH, Prineas JW. Relapsing and remitting multiple sclerosis: pathology of the newly forming lesion. Ann Neurol. 2004;55:458–468.
    1. Marik C, Felts PA, Bauer J, et al. Lesion genesis in a subset of patients with multiple sclerosis: a role for innate immunity? Brain. 2007;130(pt 11):2800–2815.
    1. Fischer MT, Wimmer I, Hoftberger R, et al. Disease-specific molecular events in cortical multiple sclerosis lesions. Brain. 2013;136(pt 6):1799–1815.
    1. Brück W, Porada P, Poser S, et al. Monocyte/macrophage differentiation in early multiple sclerosis lesions. Ann Neurol. 1995;38:788–796.
    1. Meguro R, Asano Y, Odagiri S, et al. Nonheme-iron histochemistry for light and electron microscopy: a historical, theoretical and technical review. Arch Histol Cytol. 2007;70:1–19.
    1. Bagnato F, Hametner S, Yao B, et al. Tracking iron in multiple sclerosis: a combined imaging and histopathological study at 7 tesla. Brain. 2011;134(pt 12):3602–3615.
    1. Bauer J, Elger CE, Hans VH, et al. Astrocytes are a specific immunological target in Rasmussen’s encephalitis. Ann Neurol. 2007;62:67–80.
    1. Haider L, Fischer MT, Frischer JM, et al. Oxidative damage in multiple sclerosis lesions. Brain. 2011;134(pt 7):1914–1924.
    1. Lassmann H. The architecture of inflammatory demyelinating lesions: implications for studies on pathogenesis [review] Neuropathol Applied Neurobiol. 2011;37:698–710.
    1. Schulz K, Vulpe CD, Harris LZ, David S. Iron efflux from oligodendrocytes is differentially regulated in gray and white matter. J Neurosci. 2011;31:13301–13311.
    1. Mahad D, Ziabreva I, Lassmann H, Turnbull D. Mitochondrial defects in acute multiple sclerosis lesions. Brain. 2008;131(pt 7):1722–1735.
    1. Mews I, Bergmann M, Bunkowski S, et al. Oligodendrocyte and axon pathology in clinically silent multiple sclerosis lesions. Mult Scler. 1998;4:55–62.
    1. Streit WJ, Braak H, Xue QS, Bechmann I. Dystrophic (senescent) rather than activated microglial cells are associated with tau pathology and likely precede neurodegeneration in Alzheimer’s disease. Acta Neuropathol. 2009;118:475–485.
    1. Lopes KO, Sparks DL, Streit WJ. Microglial dystrophy in the aged and Alzheimer’s disease brain is associated with ferritin immunoreactivity. Glia. 2008;56:1048–1060.
    1. Simmons DA, Casale M, Alcon B, et al. Ferritin accumulation in dystrophic microglia is an early event in the development of Huntington’s disease. Glia. 2007;55:1074–1084.
    1. Paling D, Tozer D, Wheeler-Kingshott C, et al. Reduced R2′ in multiple sclerosis normal appearing white matter and lesions may reflect decreased myelin and iron content. J Neurol Neurosurg Psychiatry. 2012;83:785–792.
    1. Rathore KI, Redensek A, David S. Iron homeostasis in astrocytes and microglia is differentially regulated by TNF-alpha and TGF-beta1. Glia. 2012;60:738–750.
    1. Zhang X, Haaf M, Todorich B, et al. Cytokine toxicity to oligodendrocyte precursors is mediated by iron. Glia. 2005;52:199–208.
    1. Zhang X, Surguladze N, Slagle-Webb B, et al. Cellular iron status influences the functional relationship between microglia and oligodendrocytes. Glia. 2006;54:795–804.
    1. van Horssen J, Drexhage JA, Flor T, et al. Nrf2 and DJ1 are consistently upregulated in inflammatory multiple sclerosis lesions. Free Radic Biol Med. 2010;49:1283–1289.
    1. Gold R, Kappos L, Arnold DL, et al. Placebo-controlled phase 3 study of oral BG-12 for relapsing multiple sclerosis. N Engl J Med. 2012;367:1098–1107.
    1. Gray E, Thomas TL, Betmouni S, et al. Elevated myeloperoxidase activity in white matter in multiple sclerosis. Neurosci Lett. 2008;444:195–198.
    1. Liu Y, Hao W, Letiembre M, et al. Suppression of microglial inflammatory activity by myelin phagocytosis: role of p47-PHOX-mediated generation of reactive oxygen species. J Neurosci. 2006;26:12904–12913.
    1. Jomova K, Valko M. Advances in metal-induced oxidative stress and human disease. Toxicology. 2011;283:65–87.
    1. Nunez MT, Urrutia P, Mena N, et al. Iron toxicity in neurodegeneration. Biometals. 2012;25:761–776.
    1. Williams R, Buchheit CL, Berman NE, LeVine SM. Pathogenic implications of iron accumulation in multiple sclerosis. J Neurochem. 2012;120:7–25.
    1. Pitt D, Boster A, Pei W, et al. Imaging cortical lesions in multiple sclerosis with ultra-high-field magnetic resonance imaging. Arch Neurol. 2010;67:812–818.
    1. Rathnasamy G, Ling EA, Kaur C. Iron and iron regulatory proteins in amoeboid microglial cells are linked to oligodendrocyte death in hypoxic neonatal rat periventricular white matter through production of proinflammatory cytokines and reactive oxygen/nitrogen species. J Neurosci. 2011;31:17982–17995.
    1. Aboul-Enein F, Rauschka H, Kornek B, et al. Preferential loss of myelin-associated glycoprotein reflects hypoxia-like white matter damage in stroke and inflammatory brain diseases. J Neuropathol Exp Neurol. 2003;62:25–33.
    1. Trapp BD, Stys PK. Virtual hypoxia and chronic necrosis of demyelinated axons in multiple sclerosis. Lancet Neurol. 2009;8:280–291.
    1. van Zwam M, Huizinga R, Melief MJ, et al. Brain antigens in functionally distinct antigen-presenting cell populations in cervical lymph nodes in MS and EAE. J Mol Med (Berl) 2009;87:273–286.
    1. Oude Engberink RD, Blezer EL, Dijkstra CD, et al. Dynamics and fate of USPIO in the central nervous system in experimental autoimmune encephalomyelitis. NMR Biomed. 2010;23:1087–1096.
    1. Schonberg DL, Goldstein EZ, Sahinkaya FR, et al. Ferritin stimulates oligodendrocyte genesis in the adult spinal cord and can be transferred from macrophages to NG2 cells in vivo. J Neurosci. 2012;32:5374–5384.
    1. Palinski W, Ylä-Herttuala S, Rosenfeld ME, Butler SW, Socher SA, Parthasarathy S, Curtiss LK, Witztum JL. Antisera and monoclonal antibodies specific for epitopes generated during oxidative modification of low density lipoprotein. Arteriosclerosis. 1990;10:325–335.

Source: PubMed

3
Předplatit