Angiogenesis in multiple sclerosis and experimental autoimmune encephalomyelitis

Francesco Girolamo, Cristiana Coppola, Domenico Ribatti, Maria Trojano, Francesco Girolamo, Cristiana Coppola, Domenico Ribatti, Maria Trojano

Abstract

Angiogenesis, the formation of new vessels, is found in Multiple Sclerosis (MS) demyelinating lesions following Vascular Endothelial Growth Factor (VEGF) release and the production of several other angiogenic molecules. The increased energy demand of inflammatory cuffs and damaged neural cells explains the strong angiogenic response in plaques and surrounding white matter. An angiogenic response has also been documented in an experimental model of MS, experimental allergic encephalomyelitis (EAE), where blood-brain barrier disruption and vascular remodelling appeared in a pre-symptomatic disease phase. In both MS and EAE, VEGF acts as a pro-inflammatory factor in the early phase but its reduced responsivity in the late phase can disrupt neuroregenerative attempts, since VEGF naturally enhances neuron resistance to injury and regulates neural progenitor proliferation, migration, differentiation and oligodendrocyte precursor cell (OPC) survival and migration to demyelinated lesions. Angiogenesis, neurogenesis and oligodendroglia maturation are closely intertwined in the neurovascular niches of the subventricular zone, one of the preferential locations of inflammatory lesions in MS, and in all the other temporary vascular niches where the mutual fostering of angiogenesis and OPC maturation occurs. Angiogenesis, induced either by CNS inflammation or by hypoxic stimuli related to neurovascular uncoupling, appears to be ineffective in chronic MS due to a counterbalancing effect of vasoconstrictive mechanisms determined by the reduced axonal activity, astrocyte dysfunction, microglia secretion of free radical species and mitochondrial abnormalities. Thus, angiogenesis, that supplies several trophic factors, should be promoted in therapeutic neuroregeneration efforts to combat the progressive, degenerative phase of MS.

Figures

Figure 1
Figure 1
Vessel density is increased in EAE mice. A. Vessel density, calculated as the cumulative vessel profile length per standard area (ImageJ software, NIH, Bethesda, USA; observer blinded to section ranking: CC), is significantly increased in EAE brains at 19 days post-immunization (dpi) (ctrl: 12 week-old control mice, n = 5; EAE: 12 week-old EAE mice, n = 5; t-test, p = 0.0026). B, C. Representative images of the vasculature of the same cerebral cortex area (area frontalis) in healthy mice and EAE (mouse at 19 dpi after 200 μg of MOG35–55 immunization; clinical onset: 10 dpi, clinical score (cs) at 19 dpi = 3.0) are immunolabelled with CD13, a pericyte marker, and collagen IV of the vessel basal lamina. Some glomeruloid microvessels associated with a high number of pericytes are visible in the EAE brains.
Figure 2
Figure 2
Hypothetical model of Neurovascular Unit function (NVU) (A) and dysfunction in an acute MS lesion (B) and a chronic end-stage lesion (C).(A) Composition of blood–brain barrier (BBB)-provided microvessels, formed by endothelial cells which are connected by tight junctions (TJ), pericytes, astrocyte endfeet, and a continuous layer of basement membrane (BM). Neurovascular coupling is brought about by astrocyte processes which remove excess K+ ions at active synaptic spaces and release these ions into perivascular spaces; at the same time Glutamate (Glu) bound to astrocyte receptors can increase astrocytic Ca++ levels and produce vasodilatatory substances. Microglia and oligodendrocyte precursor cells (OPCs) contribute to NVU function. (B) An acute MS lesion, dominated by high levels of VEGF and other angiogenic molecules, shows BBB leakiness, vasogenic swelling of BM and disrupted NVU interactions: claudin-5 and occludin, two TJ proteins, are mislocalized and downregulated; the BM is degraded by MMPs, mainly released by leukocytes infiltrating vessel BM; microglia are activated and release large amounts of pro-inflammatory mediators; astrocyte endfeet are detached from pericytes. Activated B lymphocytes release self-targeted antibodies damaging myelinated axons. (C) Chronic end-stage MS lesion dominated by hypoperfusion, and persistence of an inflammatory milieu with abundant reactive oxygen species (ROS), peroxynitrite (RNS) and stress-associated proteins, all together inhibiting the net pro-angiogenic activity. The drawing shows pro-inflammatory microglia and also the influence of demyelination on reduced axonal activities, decreased vasodilatatory stimuli and consequent vasoconstriction. Hypoperfusion is also due to vessel wall hyalinization, collagen deposition and astrocyte endfeet hypertrophy. Persistent inflammation is also responsible for endothelial-derived protective molecules and growth factors downregulation which, in turn, maintains neural stem cells (NSC) in a resting state and impedes neuroblast and OPC maturation in the neurovascular niches in the subependymal layer of the lateral ventricle (LV) and in other neuroregenerative sites around blood microvessels.

References

    1. Brück W, Bitsch A, Kolenda H, Brück Y, Stiefel M, Lassmann H. Inflammatory central nervous system demyelination: correlation of magnetic resonance imaging findings with lesion pathology. Ann Neurol. 1997;42:783–793. doi: 10.1002/ana.410420515.
    1. Kermode AG, Thompson AJ, Tofts P, MacManus DG, Kendall BE, Kingsley DP, Moseley IF, Rudge P, McDonald WI. Breakdown of the blood–brain barrier precedes symptoms and other MRI signs of new lesions in multiple sclerosis. Pathogenetic and clinical implications. Brain. 1990;113:1477–1489. doi: 10.1093/brain/113.5.1477.
    1. Alvarez JI, Cayrol R, Prat A. Disruption of central nervous system barriers in multiple sclerosis. Biochim Biophys Acta. 2011;812:252–264. doi: 10.1016/j.bbadis.2010.06.017.
    1. Claudio L, Raine CS, Brosnan CF. Evidence of persistent blood–brain barrier abnormalities in chronic-progressive multiple sclerosis. Acta Neuropathol. 1995;90:228–238. doi: 10.1007/BF00296505.
    1. Kirk J, Plumb J, Mirakhur M, McQuaid S. Tight junctional abnormality in multiple sclerosis white matter affects all calibers of vessel and is associated with blood–brain barrier leakage and active demyelination. J Pathol. 2003;201:319–327. doi: 10.1002/path.1434.
    1. McQuaid S, Kirk JT. The blood–brain barrier in multiple sclerosis. Int Congress Series. 2005;1277:235–243. doi: 10.1016/j.ics.2005.02.026.
    1. Minagar A, Alexander JS. Blood–brain barrier disruption in multiple sclerosis. Mult Scler. 2003;9:540–549. doi: 10.1191/1352458503ms965oa.
    1. Plumb J, McQuaid S, Mirakhur M, Kirk J. Abnormal endothelial tight junctions in active lesions and normal-appearing white matter in multiple sclerosis. Brain Pathol. 2002;12:154–169. doi: 10.1111/j.1750-3639.2002.tb00430.x.
    1. van Horssen J, Bö L, Vos CM, Virtanen I, de Vries HE. Basement membrane proteins in multiple sclerosis-associated inflammatory cuffs: Potential role in influx and transport of leukocytes. J Neuropathol Exp Neurol. 2005;64:722–729. doi: 10.1097/01.jnen.0000173894.09553.13.
    1. Wosik K, Cayrol R, Dodelet-Devillers A, Berthelet F, Bernard M, Moumdjian R, Bouthillier A, Reudelhuber TL, Prat A. Angiotensin II controls occludin function and is required for blood brain barrier maintenance: relevance to multiple sclerosis. J Neurosci. 2007;27:9032–9042. doi: 10.1523/JNEUROSCI.2088-07.2007.
    1. Gay D, Esiri M. Blood–brain barrier damage in acute multiple sclerosis plaques. An immunocytological study. Brain. 1991;114:557–572. doi: 10.1093/brain/114.1.557.
    1. Su JJ, Osoegawa M, Matsuoka T, Minohara M, Tanaka M, Ishizu T, Mihara F, Taniwaki T, Kira J. Upregulation of vascular growth factors in multiple sclerosis: correlation with MRI findings. J Neurol Sci. 2006;243:21–30. doi: 10.1016/j.jns.2005.11.006.
    1. Dousset V, Brochet B, Deloire MS, Lagoarde L, Barroso B, Caille JM, Petry KG. MR imaging of relapsing multiple sclerosis patients using ultra-small-particle iron oxide and compared with gadolinium. Am J Neuroradiol. 2006;27:1000–1005.
    1. Vellinga MM, Engberink RD, Seewann A, Pouwels P, Wattjes M, van der Pol S, Pering C, Polman CH, de Vries HE, Geurts JJ, Barkhof F. Pluriformity of inflammation in multiple sclerosis shown by ultra-small iron oxide particle enhancement. Brain. 2008;131:800–807. doi: 10.1093/brain/awn009.
    1. Ladewig G, Jestaedt L, Misselwitz B, Solymosi L, Toyka K, Bendszus M, Stoll G. Spatial diversity of blood–brain barrier alteration and macrophage invasion in experimental autoimmune encephalomyelitis: a comparative MRI study. Exp Neurol. 2009;220:207–211. doi: 10.1016/j.expneurol.2009.08.027.
    1. Mayhan WG. VEGF increases permeability of the blood–brain barrier via a nitric oxide synthase/cGMP-dependent pathway. Am J Physiol. 1999;276:C1148–1153.
    1. Barleon B, Sozzani S, Zhou D, Weich HA, Mantovani A, Marmé D. Migration of human monocytes in response to vascular endothelial growth factor (VEGF) is mediated via the VEGF receptor flt-1. Blood. 1996;87:3336–3343.
    1. Clauss M, Gerlach M, Gerlach H, Brett J, Wang F, Familletti PC, Pan YC, Olander JV, Connolly DT, Stern D. Vascular permeability factor: a tumor-derived polypeptide that induces endothelial cell and monocyte procoagulant activity, and promotes monocyte migration. J Exp Med. 1990;172:1535–1545. doi: 10.1084/jem.172.6.1535.
    1. Proescholdt MA, Heiss JD, Walbridge S, Mühlhauser J, Capogrossi MC, Oldfield EH, Merrill MJ. Vascular endothelial growth factor (VEGF) modulates vascular permeability and inflammation in rat brain. J Neuropathol Exp Neurol. 1999;58:613–627. doi: 10.1097/00005072-199906000-00006.
    1. Virgintino D, Girolamo F, Errede M, Capobianco C, Robertson D, Stallcup WB, Perris R, Roncali L. An intimate interplay between precocious, migrating pericytes and endothelial cells governs human fetal brain angiogenesis. Angiogenesis. 2007;10:35–45. doi: 10.1007/s10456-006-9061-x.
    1. Sung HK, Michael IP, Nagy A. Multifaceted role of vascular endothelial growth factor signaling in adult tissue physiology: an emerging concept with clinical implications. Curr Opin Hematol. 2010;17:206–212.
    1. Holley JE, Newcombe J, Whatmore JL, Gutowski NJ. Increased blood vessel density and endothelial cell proliferation in multiple sclerosis cerebral white matter. Neurosci Lett. 2010;470:65–70. doi: 10.1016/j.neulet.2009.12.059.
    1. Ludwin S. Vascular proliferation and angiogenesis in MS: clinical and pathogenic implications. J Neuropath Exp Neurol. 2001;60:505.
    1. Vagnucci AH, Jr, Li WW. Alzheimer's disease and angiogenesis. Lancet. 2003;361:605–608. doi: 10.1016/S0140-6736(03)12521-4.
    1. Desai Bradaric B, Patel A, Schneider JA, Carvey PM, Hendey B. Evidence for angiogenesis in Parkinson's disease, incidental Lewy body disease, and progressive supranuclear palsy. J Neural Transm. 2012;119:59–71. doi: 10.1007/s00702-011-0684-8.
    1. Plate KH, Scholz A, Dumont DJ. Tumor angiogenesis and anti-angiogenic therapy in malignant gliomas revisited. Acta Neuropathol. 2012;124:763–775. doi: 10.1007/s00401-012-1066-5.
    1. Xiong M, Elson G, Legarda D, Leibovich SJ. Production of vascular endothelial growth factor by murine macrophages - regulation by hypoxia, lactate, and the inducible nitric oxide synthase pathway. Am J Pathol. 1998;153:587–598. doi: 10.1016/S0002-9440(10)65601-5.
    1. Papadaki EZ, Simos PG, Mastorodemos VC, Panou T, Maris TG, Karantanas AH, Plaitakis A. Regional MRI perfusion measures predict motor/executive function in patients with Clinically Isolated Syndrome. Behav Neurol. 2014;ᅟ:ᅟ.
    1. Fog T. The topography of plaques in multiple sclerosis. With special reference to cerebral plaques. Acta Neurol Scand. 1965;41(Suppl 15):1–161.
    1. Macchi G. The pathology of the blood vessels in multiple sclerosis. J Neuropath Exp Neurol. 1954;13:378–84. doi: 10.1097/00005072-195404000-00008.
    1. Putnam TJ. The pathogenesis of multiple sclerosis: a possible vascular factor. N Engl J Med. 1933;209:786–790. doi: 10.1056/NEJM193310192091604.
    1. Rindfleisch E. Histologische Details zu der grauen degeneration von Gehirn und Rückenmark. Virchow’s Archiv für Pathologische Anatomie und Physiologie und für klinische Medizin. 1863;26:474–83.
    1. Scheinker M. Histogenesis of the early lesions of multiple sclerosis. 1. Significance of the vascular changes. Arch Neurol Psychiat. 1943;49:178. doi: 10.1001/archneurpsyc.1943.02290140038002.
    1. Broman T. Supravital analysis of disorders in the cerebral vascular permeability II. Two cases of multiple sclerosis. Acta Psychiatr Neurol Scand (Suppl) 1947;46:58–71. doi: 10.1111/j.1600-0447.1947.tb08101.x.
    1. Proescholdt MA, Jacobson S, Tresser N, Oldfield EH, Merrill MJ. Vascular endothelial growth factor is expressed in multiple sclerosis plaques and can induce inflammatory lesions in experimental allergic encephalomyelitis rats. J Neuropathol Exp Neurol. 2002;61:914–925. doi: 10.1093/jnen/61.10.914.
    1. Karlik SJ, Roscoe WA, Patinote C, Contino-Pepin C. Targeting vascular changes in lesions in multiple sclerosis and experimental autoimmune encephalomyelitis. Cent Nerv Syst Agents Med Chem. 2012;12:7–14. doi: 10.2174/187152412800229125.
    1. Kirk S, Frank JA, Karlik S. Angiogenesis in multiple sclerosis: is it good, bad or an epiphenomenon? J Neurol Sci. 2004;217:125–130. doi: 10.1016/j.jns.2003.10.016.
    1. Trapp BD, Stys PK. Virtual hypoxia and chronic necrosis of demyelinated axons in multiple sclerosis. Lancet Neurol. 2009;8:280–291. doi: 10.1016/S1474-4422(09)70043-2.
    1. Argaw AT, Asp L, Zhang J, Navrazhina K, Pham T, Mariani JN, Mahase S, Dutta DJ, Seto J, Kramer EG, Ferrara N, Sofroniew MV, John GR. Astrocyte-derived VEGF-A drives blood–brain barrier disruption in CNS inflammatory disease. J Clin Invest. 2012;122:2454–2468. doi: 10.1172/JCI60842.
    1. Boroujerdi A, Welser-Alves JV, Milner R. Extensive vascular remodeling in the spinal cord of pre-symptomatic experimental autoimmune encephalomyelitis mice; increased vessel expression of fibronectin and the α5β1 integrin. Exp Neurol. 2013;250:43–51. doi: 10.1016/j.expneurol.2013.09.009.
    1. Daniel PM, Lam DK, Pratt OE. Changes in the effectiveness of the blood–brain and blood-spinal cord barriers in experimental allergic encephalomyelitis. J Neurol Sci. 1981;52:211–219. doi: 10.1016/0022-510X(81)90006-X.
    1. Errede M, Girolamo F, Ferrara G, Strippoli M, Morando S, Boldrin V, Rizzi M, Uccelli A, Perris R, Bendotti C, Salmona M, Roncali L, Virgintino D. Blood–brain barrier alterations in the cerebral cortex in experimental autoimmune encephalomyelitis. J Neuropathol Exp Neurol. 2012;71:840–854. doi: 10.1097/NEN.0b013e31826ac110.
    1. Juhler M, Barry DI, Offner H, Konat G, Klinken L, Paulson OB. Blood–brain and blood-spinal cord barrier permeability during the course of experimental allergic encephalomyelitis in the rat. Brain Res. 1984;302:347–355. doi: 10.1016/0006-8993(84)90249-X.
    1. Zlokovic BV, Skundric DS, Segal MB, Colover J, Jankov RM, Pejnovic N, Lackovic V, Mackic J, Lipovac MN, Dawson H, Kasp E, Dumonde D, Rakic L. Blood–brain barrier permeability changes during acute allergic encephalomyelitis induced in the guinea pig. Metab Brain Dis. 1989;4:33–40. doi: 10.1007/BF00999491.
    1. MacMillan CJ, Starkey RJ, Easton AS. Angiogenesis is regulated by angiopoietins during experimental autoimmune encephalomyelitis and is indirectly related to vascular permeability. J Neuropathol Exp Neurol. 2011;70:1107–23. doi: 10.1097/NEN.0b013e31823a8b6a.
    1. Roscoe WA, Welsh ME, Carter DE, Karlik SJ. VEGF and angiogenesis in acute and chronic MOG((35–55)) peptide induced EAE. J Neuroimmunol. 2009;209:6–15. doi: 10.1016/j.jneuroim.2009.01.009.
    1. Kirk SL, Karlik SJ. VEGF and vascular changes in chronic neuroinflammation. J Autoimmun. 2003;21:353–363. doi: 10.1016/S0896-8411(03)00139-2.
    1. Piraino PS, Yednock TA, Messersmith EK, Pleiss MA, Freedman SB, Hammond RR, Karlik SJ. Spontaneous remyelination following prolonged inhibition of alpha4 integrin in chronic EAE. J Neuroimmunol. 2005;167:53–63. doi: 10.1016/j.jneuroim.2005.06.018.
    1. Sobel RA, Blanchette BW, Bhan AK, Colvin RB. The immunopathology of experimental allergic encephalomyelitis. II. Endothelial cell Ia increases prior to inflammatory cell infiltration. J Immunol. 1984;132:2402–2407.
    1. Seabrook TJ, Littlewood-Evans A, Brinkmann V, Pöllinger B, Schnell C, Hiestand PC. Angiogenesis is present in experimental autoimmune encephalomyelitis and pro-angiogenic factors are increased in multiple sclerosis lesions. J Neuroinflammation. 2010;7:95. doi: 10.1186/1742-2094-7-95.
    1. Mor F, Quintana FJ, Cohen IR. Angiogenesis-inflammation cross-talk: vascular endothelial growth factor is secreted by activated T cells and induces Th1 polarization. J Immunol. 2004;172:4618–4623. doi: 10.4049/jimmunol.172.7.4618.
    1. Sasaki M, Lankford KL, Brown RJ, Ruddle NH, Kocsis JD. Focal experimental autoimmune encephalomyelitis in the Lewis rat induced by immunization with myelin oligodendrocyte glycoprotein and intraspinal injection of vascular endothelial growth factor. Glia. 2010;58:1523–1531. doi: 10.1002/glia.21026.
    1. Tham E, Gielen AW, Khademi M, Martin C, Piehl F. Decreased expression of VEGF-A in rat experimental autoimmune encephalomyelitis and in cerebrospinal fluid mononuclear cells from patients with multiple sclerosis. Scand J Immunol. 2006;64:609–622. doi: 10.1111/j.1365-3083.2006.01851.x.
    1. McCloskey DP, Tana MH, Scharfman HE. Modulation of vascular endothelial growth factor (VEGF) expression in motor neurons and its electrophysiological effects. Brain Res Bull. 2008;76:36–44. doi: 10.1016/j.brainresbull.2007.11.018.
    1. Knizetova P, Ehrmann J, Hlobilkova A, Vancova I, Kalita O, Kolar Z, Bartek J. Autocrine regulation of glioblastoma cell cycle progression, viability and radioresistance through the VEGF-VEGFR2 (KDR) interplay. Cell Cycle. 2008;7:2553–2561. doi: 10.4161/cc.7.16.6442.
    1. Storkebaum E, Lambrechts D, Carmeliet P. VEGF: once regarded as a specific angiogenic factor, now implicated in neuroprotection. Bioessays. 2004;26:943–954. doi: 10.1002/bies.20092.
    1. Zhang ZG, Zhang L, Tsang W, Soltanian-Zadeh H, Morris D, Zhang R, Goussev A, Powers C, Yeich T, Chopp M. Correlation of VEGF and angiopoietin expression with disruption of blood–brain barrier and angiogenesis after focal cerebral ischemia. J Cereb Blood Flow Metab. 2002;22:379–392. doi: 10.1097/00004647-200204000-00002.
    1. Graumann U, Reynolds R, Steck AJ, Schaeren-Wiemers N. Molecular changes in normal appearing white matter in multiple sclerosis are characteristic of neuroprotective mechanisms against hypoxic insult. Brain Pathol. 2003;13:554–573. doi: 10.1111/j.1750-3639.2003.tb00485.x.
    1. Theoharides TC, Konstantinidou AD. Corticotropin-releasing hormone and the blood–brain-barrier. Front Biosci. 2007;12:1615–1628. doi: 10.2741/2174.
    1. van Bruggen N, Thibodeaux H, Palmer JT, Lee WP, Fu L, Cairns B, Tumas D, Gerlai R, Williams SP, van Lookeren CM, Ferrara N. VEGF antagonism reduces edema formation and tissue damage after ischemia/reperfusion injury in the mouse brain. J Clin Invest. 1999;104:1613–1620. doi: 10.1172/JCI8218.
    1. Benton RL, Whittemore SR. VEGF165 therapy exacerbates secondary damage following spinal cord injury. Neurochem Res. 2003;28:1693–1703. doi: 10.1023/A:1026013106016.
    1. Widenfalk J, Lipson A, Jubran M, Hofstetter C, Ebendal T, Cao Y, Olson L. Vascular endothelial growth factor improves functional outcome and decreases secondary degeneration in experimental spinal cord contusion injury. Neuroscience. 2003;120:951–960. doi: 10.1016/S0306-4522(03)00399-3.
    1. Iacobaeus E, Amoudruz P, Ström M, Khademi M, Brundin L, Hillert J, Kockum I, Malmström V, Olsson T, Tham E, Piehl F. The expression of VEGF-A is down regulated in peripheral blood mononuclear cells of patients with secondary progressive multiple sclerosis. PLoS One. 2011;6:e19138. doi: 10.1371/journal.pone.0019138.
    1. MacMillan CJ, Furlong SJ, Doucette CD, Chen PL, Hoskin DW, Easton AS. Bevacizumab diminishes experimental autoimmune encephalomyelitis by inhibiting spinal cord angiogenesis and reducing peripheral T-cell responses. J Neuropathol Exp Neurol. 2012;71:983–999. doi: 10.1097/NEN.0b013e3182724831.
    1. Lambrechts D, Storkebaum E, Morimoto M, Del-Favero J, Desmet F, Marklund SL, Wyns S, Thijs V, Andersson J, van Marion I, Al-Chalabi A, Bornes S, Musson R, Hansen V, Beckman L, Adolfsson R, Pall HS, Prats H, Vermeire S, Rutgeerts P, Katayama S, Awata T, Leigh N, Lang-Lazdunski L, Dewerchin M, Shaw C, Moons L, Vlietinck R, Morrison KE, Robberecht W, Van Broeckhoven C, et al. VEGF is a modifier of amyotrophic lateral sclerosis in mice and humans and protects motoneurons against ischemic death. Nat Genet. 2003;34:383–394. doi: 10.1038/ng1211.
    1. Oosthuyse B, Moons L, Storkebaum E, Beck H, Nuyens D, Brusselmans K, Van Dorpe J, Hellings P, Gorselink M, Heymans S, Theilmeier G, Dewerchin M, Laudenbach V, Vermylen P, Raat H, Acker T, Vleminckx V, Van Den Bosch L, Cashman N, Fujisawa H, Drost MR, Sciot R, Bruyninckx F, Hicklin DJ, Ince C, Gressens P, Lupu F, Plate KH, Robberecht W, Herbert JM, et al. Deletion of the hypoxia response element in the vascular endothelial growth factor promoter causes motor neuron degeneration. Nat Genet. 2001;28:131–138. doi: 10.1038/88842.
    1. Storkebaum E, Lambrechts D, Dewerchin M, Moreno-Murciano MP, Appelmans S, Oh H, Van Damme P, Rutten B, Man WY, De Mol M, Wyns S, Manka D, Vermeulen K, Van Den Bosch L, Mertens N, Schmitz C, Robberecht W, Conway EM, Collen D, Moons L, Carmeliet P. Treatment of motoneuron degeneration by intracerebroventricular delivery of VEGF in a rat model of ALS. Nat Neurosci. 2005;8:85–92. doi: 10.1038/nn1360.
    1. Sondell M, Lundborg G, Kanje M. Vascular endothelial growth factor has neurotrophic activity and stimulates axonal outgrowth, enhancing cell survival and Schwann cell proliferation in the peripheral nervous system. J Neurosci. 1999;19:5731–5740.
    1. Argaw AT, Gurfein BT, Zhang Y, Zameer A, John GR. VEGF-mediated disruption of endothelial CLN-5 promotes blood–brain barrier breakdown. Proc Natl Acad Sci U S A. 2009;106:1977–1982. doi: 10.1073/pnas.0808698106.
    1. Rosenstein JM, Krum JM. New roles for VEGF in nervous tissue–beyond blood vessels. Exp Neurol. 2004;187:246–253. doi: 10.1016/j.expneurol.2004.01.022.
    1. Okada S, Nakamura M, Katoh H, Miyao T, Shimazaki T, Ishii K, Yamane J, Yoshimura A, Iwamoto Y, Toyama Y, Okano H. Conditional ablation of Stat3 or Socs3 discloses a dual role for reactive astrocytes after spinal cord injury. Nat Med. 2006;12:829–834. doi: 10.1038/nm1425.
    1. Choi JS, Kim HY, Cha JH, Choi JY, Park SI, Jeong CH, Jeun SS, Lee MY. Upregulation of vascular endothelial growth factor receptors Flt-1 and Flk-1 following acute spinal cord contusion in rats. J Histochem Cytochem. 2007;55:821–830. doi: 10.1369/jhc.6A7139.2007.
    1. Herz J, Reitmeir R, Hagen SI, Reinboth BS, Guo Z, Zechariah A, ElAli A, Doeppner TR, Bacigaluppi M, Pluchino S, Kilic U, Kilic E, Hermann DM. Intracerebroventricularly delivered VEGF promotes contralesional corticorubral plasticity after focal cerebral ischemia via mechanisms involving anti-inflammatory actions. Neurobiol Dis. 2012;45:1077–1085. doi: 10.1016/j.nbd.2011.12.026.
    1. Heil M, Clauss M, Suzuki K, Buschmann IR, Willuweit A, Fischer S, Schaper W. Vascular endothelial growth factor (VEGF) stimulates monocyte migration through endothelial monolayers via increased integrin expression. Eur J Cell Biol. 2000;79:850–857. doi: 10.1078/0171-9335-00113.
    1. Ishizu T, Osoegawa M, Mei FJ, Kikuchi H, Tanaka M, Takakura Y, Minohara M, Murai H, Mihara F, Taniwaki T, Kira J. Intrathecal activation of the IL-17/IL-8 axis in opticospinal multiple sclerosis. Brain. 2005;128:988–1002. doi: 10.1093/brain/awh453.
    1. Cho ML, Jung YO, Moon YM, Min SY, Yoon CH, Lee SH, Park SH, Cho CS, Jue DM, Kim HY. Interleukin-18 induces the production of vascular endothelial growth factor (VEGF) in rheumatoid arthritis synovial fibroblasts via AP-1-dependent pathways. Immunol Lett. 2006;103:159–166. doi: 10.1016/j.imlet.2005.10.020.
    1. Suarez S, Ballmer-Hofer K. VEGF transiently disrupts gap junctional communication in endothelial cells. J Cell Sci. 2001;114:1229–1235.
    1. Beazley-Long N, Hua J, Jehle T, Hulse RP, Dersch R, Lehrling C, Bevan H, Qiu Y, Lagrèze WA, Wynick D, Churchill AJ, Kehoe P, Harper SJ, Bates DO, Donaldson LF. VEGF-A165b is an endogenous neuroprotective splice isoform of vascular endothelial growth factor A in vivo and in vitro. Am J Pathol. 2013;183:918–929. doi: 10.1016/j.ajpath.2013.05.031.
    1. Kawamura H, Li X, Harper SJ, Bates DO, Claesson-Welsh L. Vascular endothelial growth factor (VEGF)-A165b is a weak in vitro agonist for VEGF receptor-2 due to lack of coreceptor binding and deficient regulation of kinase activity. Cancer Res. 2008;68:4683–4692. doi: 10.1158/0008-5472.CAN-07-6577.
    1. Liu Y, Cox SR, Morita T, Kourembanas S. Hypoxia regulates vascular endothelial growth factor gene expression in endothelial cells. Identification of a 5' enhancer. Circ Res. 1995;77:638–643. doi: 10.1161/01.RES.77.3.638.
    1. Lassmann H. Hypoxia-like tissue injury as a component of multiple sclerosis lesions. J Neurol Sci. 2003;206:187–91. doi: 10.1016/S0022-510X(02)00421-5.
    1. Distler JH, Hirth A, Kurowska-Stolarska M, Gay RE, Gay S, Distler O. Angiogenic and angiostatic factors in the molecular control of angiogenesis. Q J Nucl Med. 2003;47:149–161.
    1. Frost EE, Nielsen JA, Le TQ, Armstrong RC. PDGF and FGF2 regulate oligodendrocyte progenitor responses to demyelination. J Neurobiol. 2003;54:457–472. doi: 10.1002/neu.10158.
    1. Van Meir EG. Cytokines and tumors of the central nervous system. Glia. 1995;15:264–288. doi: 10.1002/glia.440150308.
    1. Haufschild T, Shaw SG, Kesselring J, Flammer J. Increased endothelin-1 plasma levels in patients with multiple sclerosis. J Neuroophthalmol. 2001;21:37–38. doi: 10.1097/00041327-200103000-00011.
    1. Shin T, Kang B, Tanuma N, Matsumoto Y, Wie M, Ahn M, Kang J. Intrathecal administration of endothelin-1 receptor antagonist ameliorates autoimmune encephalomyelitis in Lewis rats. Neuroreport. 2001;12:1465–1468. doi: 10.1097/00001756-200105250-00034.
    1. Li L, Welser-Alves J, van der Flier A, Boroujerdi A, Hynes RO, Milner R. An angiogenic role for the α5β1 integrin in promoting endothelial cell proliferation during cerebral hypoxia. Exp Neurol. 2012;237:46–54. doi: 10.1016/j.expneurol.2012.06.005.
    1. Cunnea P, McMahon J, O'Connell E, Mashayekhi K, Fitzgerald U, McQuaid S. Gene expression analysis of the microvascular compartment in multiple sclerosis using laser microdissected blood vessels. Acta Neuropathol. 2010;119:601–15. doi: 10.1007/s00401-009-0618-9.
    1. Jackson JR, Seed MP, Kircher CH, Willoughby DA, Winkler JD. The codependence of angiogenesis and chronic inflammation. FASEB J. 1997;11:457–465.
    1. Ward NL, Lamanna JC. The neurovascular unit and its growth factors: coordinated response in the vascular and nervous systems. Neurol Res. 2004;26:870–883. doi: 10.1179/016164104X3798.
    1. Carmeliet P, Ruiz de Almodovar C. VEGF ligands and receptors: implications in neurodevelopment and neurodegeneration. Cell Mol Life Sci. 2013;70:1763–1778. doi: 10.1007/s00018-013-1283-7.
    1. Carmeliet P, Tessier-Lavigne M. Common mechanisms of nerve and blood vessel wiring. Nature. 2005;436:193–200. doi: 10.1038/nature03875.
    1. Leventhal C, Rafii S, Rafii D, Shahar A, Goldman SA. Endothelial trophic support of neuronal production and recruitment from the adult mammalian subependyma. Mol Cell Neurosci. 1999;13:450–464. doi: 10.1006/mcne.1999.0762.
    1. Shen Q, Goderie SK, Jin L, Karanth N, Sun Y, Abramova N, Vincent P, Pumiglia K, Temple S. Endothelial cells stimulate self-renewal and expand neurogenesis of neural stem cells. Science. 2004;304:1338–1340. doi: 10.1126/science.1095505.
    1. Zacchigna S, Lambrechts D, Carmeliet P. Neurovascular signalling defects in neurodegeneration. Nat Rev Neurosci. 2008;9:169–181. doi: 10.1038/nrn2336.
    1. Jin K, Zhu Y, Sun Y, Mao XO, Xie L, Greenberg DA. Vascular endothelial growth factor (VEGF) stimulates neurogenesis in vitro and in vivo. Proc Natl Acad Sci U S A. 2002;99:11946–11950. doi: 10.1073/pnas.182296499.
    1. Zhao C, Deng W, Gage FH. Mechanisms and functional implications of adult neurogenesis. Cell. 2008;132:645–660. doi: 10.1016/j.cell.2008.01.033.
    1. Ma YY, Li KY, Wang JJ, Huang YL, Huang Y, Sun FY. Vascular endothelial growth factor acutely reduces calcium influx via inhibition of the Ca2+ channels in rat hippocampal neurons. J Neurosci Res. 2009;87:393–402. doi: 10.1002/jnr.21859.
    1. Hayakawa K, Pham LD, Som AT, Lee BJ, Guo S, Lo EH, Arai K. Vascular endothelial growth factor regulates the migration of oligodendrocyte precursor cells. J Neurosci. 2011;31:10666–10670. doi: 10.1523/JNEUROSCI.1944-11.2011.
    1. Gao P, Shen F, Gabriel RA, Law D, Yang E, Yang GY, Young WL, Su H. Attenuation of brain response to vascular endothelial growth factor-mediated angiogenesis and neurogenesis in aged mice. Stroke. 2009;40:3596–3600. doi: 10.1161/STROKEAHA.109.561050.
    1. Kazanis I, Lathia JD, Vadakkan TJ, Raborn E, Wan R, Mughal MR, Eckley DM, Sasaki T, Patton B, Mattson MP, Hirschi KK, Dickinson ME, Ffrench-Constant C. Quiescence and activation of stem and precursor cell populations in the subependymal zone of the mammalian brain are associated with distinct cellular and extracellular matrix signals. J Neurosci. 2010;30:9771–9781. doi: 10.1523/JNEUROSCI.0700-10.2010.
    1. Kokovay E, Goderie S, Wang Y, Lotz S, Lin G, Sun Y, Roysam B, Shen Q, Temple S. Adult SVZ lineage cells home to and leave the vascular niche via differential responses to SDF1/CXCR4 signaling. Cell Stem Cell. 2010;7:163–173. doi: 10.1016/j.stem.2010.05.019.
    1. Patel JR, McCandless EE, Dorsey D, Klein RS. CXCR4 promotes differentiation of oligodendrocyte progenitors and remyelination. Proc Natl Acad Sci U S A. 2010;107:11062–11067. doi: 10.1073/pnas.1006301107.
    1. Kerever A, Schnack J, Vellinga D, Ichikawa N, Moon C, Arikawa-Hirasawa E, Efird JT, Mercier F. Novel extracellular matrix structures in the neural stem cell niche capture the neurogenic factor fibroblast growth factor 2 from the extracellular milieu. Stem Cells. 2007;25:2146–2157. doi: 10.1634/stemcells.2007-0082.
    1. Clemente D, Ortega MC, Arenzana FJ, de Castro F. FGF-2 and Anosmin-1 are selectively expressed in different types of multiple sclerosis lesions. J Neurosci. 2011;31:14899–14909. doi: 10.1523/JNEUROSCI.1158-11.2011.
    1. Sohn J, Selvaraj V, Wakayama K, Orosco L, Lee E, Crawford SE, Guo F, Lang J, Horiuchi M, Zarbalis K, Itoh T, Deng W, Pleasure D. PEDF is a novel oligodendrogenic morphogen acting on the adult SVZ and corpus callosum. J Neurosci. 2012;32:12152–12164. doi: 10.1523/JNEUROSCI.0628-12.2012.
    1. Aguirre A, Rubio ME, Gallo V. Notch and EGFR pathway interaction regulates neural stem cell number and self-renewal. Nature. 2010;467:323–327. doi: 10.1038/nature09347.
    1. Rash BG, Lim HD, Breunig JJ, Vaccarino FM. FGF signaling expands embryonic cortical surface area by regulating Notch-dependent neurogenesis. J Neurosci. 2011;31:15604–15617. doi: 10.1523/JNEUROSCI.4439-11.2011.
    1. Cha YK, Kim YH, Ahn YH, Koh JY. Epidermal growth factor induces oxidative neuronal injury in cortical culture. J Neurochem. 2000;75:298–303. doi: 10.1046/j.1471-4159.2000.0750298.x.
    1. Levy YA, Fainberg KM, Amidror T, Regev K, Auriel E, Karni A. High and dysregulated secretion of epidermal growth factor from immune cells of patients with relapsing-remitting multiple sclerosis. J Neuroimmunol. 2013;257:82–89. doi: 10.1016/j.jneuroim.2013.01.012.
    1. Calza L, Giuliani A, Fernandez M, Pirondi S, D’Intino G, Aloe L, Giardino L. Neural stem cells and cholinergic neurons: regulation by immunolesion and treatment with mitogens, retinoic acid, and nerve growth factor. Proc Natl Acad Sci U S A. 2003;100:7325–7330. doi: 10.1073/pnas.1132092100.
    1. Kim H, Li Q, Hempstead BL, Madri JA. Paracrine and autocrine functions of brain-derived neurotrophic factor (BDNF) and nerve growth factor (NGF) in brain-derived endothelial cells. J Biol Chem. 2004;279:33538–33546. doi: 10.1074/jbc.M404115200.
    1. Mashayekhi F, Salehi Z, Jamalzadeh HR. Quantitative analysis of cerebrospinal fluid brain derived neurotrophic factor in the patients with multiple sclerosis. Acta Med (Hradec Kralove) 2012;55:83–86. doi: 10.14712/18059694.2015.60.
    1. Laudiero LB, Aloe L, Levi-Montalcini R, Buttinelli C, Schilter D, Gillessen S, Otten U. Multiple sclerosis patients express increased levels of beta-nerve growth factor in cerebrospinal fluid. Neurosci Lett. 1992;147:9–12. doi: 10.1016/0304-3940(92)90762-V.
    1. Stadelmann C, Kerschensteiner M, Misgeld T, Brück W, Hohlfeld R, Lassmann H. BDNF and gp145trkB in multiple sclerosis brain lesions: neuroprotective interactions between immune and neuronal cells? Brain. 2002;125:75–85. doi: 10.1093/brain/awf015.
    1. Kalinowska-Lyszczarz A, Losy J. The role of neurotrophins in multiple sclerosis-pathological and clinical implications. Int J Mol Sci. 2012;13:13713–13725. doi: 10.3390/ijms131013713.
    1. Larrivée B, Freitas C, Suchting S, Brunet I, Eichmann A. Guidance of vascular development: lessons from the nervous system. Circ Res. 2009;104:428–441. doi: 10.1161/CIRCRESAHA.108.188144.
    1. Mizutani K, Yoon K, Dang L, Tokunaga A, Gaiano N. Differential Notch signalling distinguishes neural stem cells from intermediate progenitors. Nature. 2007;449:351–355. doi: 10.1038/nature06090.
    1. Kume T. Novel insights into the differential functions of Notch ligands in vascular formation. J Angiogenes Res. 2009;1:8. doi: 10.1186/2040-2384-1-8.
    1. Shimojo H, Ohtsuka T, Kageyama R. Oscillations in notch signaling regulate maintenance of neural progenitors. Neuron. 2008;58:52–64. doi: 10.1016/j.neuron.2008.02.014.
    1. Tavazoie M, Van der Veken L, Silva-Vargas V, Louissaint M, Colonna L, Zaidi B, Garcia-Verdugo JM, Doetsch F. A specialized vascular niche for adult neural stem cells. Cell Stem Cell. 2008;3:279–288. doi: 10.1016/j.stem.2008.07.025.
    1. Juryńczyk M, Selmaj K. Notch: a new player in MS mechanisms. J Neuroimmunol. 2010;218:3–11. doi: 10.1016/j.jneuroim.2009.08.010.
    1. Hashimoto M, Ishii K, Nakamura Y, Watabe K, Kohsaka S, Akazawa C. Neuroprotective effect of sonic hedgehog up-regulated in Schwann cells following sciatic nerve injury. J Neurochem. 2008;107:918–927. doi: 10.1111/j.1471-4159.2008.05731.x.
    1. Hirsch C, Campano LM, Wohrle S, Hecht A. Canonical Wnt signaling transiently stimulates proliferation and enhances neurogenesis in neonatal neural progenitor cultures. Exp Cell Res. 2007;313:572–587. doi: 10.1016/j.yexcr.2006.11.002.
    1. Xie C, Li Z, Zhang GX, Guan Y. Wnt signaling in Remyelination in multiple sclerosis: friend or foe? Mol Neurobiol. 2013
    1. Alvarez JI, Dodelet-Devillers A, Kebir H, Ifergan I, Fabre PJ, Terouz S, Sabbagh M, Wosik K, Bourbonnière L, Bernard M, van Horssen J, de Vries HE, Charron F, Prat A. The Hedgehog pathway promotes blood–brain barrier integrity and CNS immune quiescence. Science. 2011;334:1727–1731. doi: 10.1126/science.1206936.
    1. Wälchli T, Pernet V, Weinmann O, Shiu JY, Guzik-Kornacka A, Decrey G, Yüksel D, Schneider H, Vogel J, Ingber DE, Vogel V, Frei K, Schwab ME. Nogo-A is a negative regulator of CNS angiogenesis. Proc Natl Acad Sci U S A. 2013;110:E1943–1952. doi: 10.1073/pnas.1216203110.
    1. Karnezis T, Mandemakers W, McQualter JL, Zheng B, Ho PP, Jordan KA, Murray BM, Barres B, Tessier-Lavigne M, Bernard CC. The neurite outgrowth inhibitor Nogo A is involved in autoimmune-mediated demyelination. Nat Neurosci. 2004;7:736–744. doi: 10.1038/nn1261.
    1. Bin JM, Rajasekharan S, Kuhlmann T, Hanes I, Marcal N, Han D, Rodrigues SP, Leong SY, Newcombe J, Antel JP, Kennedy TE. Full-length and fragmented netrin-1 in multiple sclerosis plaques are inhibitors of oligodendrocyte precursor cell migration. Am J Pathol. 2013;183:673–680. doi: 10.1016/j.ajpath.2013.06.004.
    1. Sobel RA. Ephrin A receptors and ligands in lesions and normal-appearing white matter in multiple sclerosis. Brain Pathol. 2005;15:35–45. doi: 10.1111/j.1750-3639.2005.tb00098.x.
    1. Munro KM, Dixon KJ, Gresle MM, Jonas A, Kemper D, Doherty W, Fabri LJ, Owczarek CM, Pearse M, Boyd AW, Kilpatrick TJ, Butzkueven H, Turnley AM. EphA4 receptor tyrosine kinase is a modulator of onset and disease severity of experimental autoimmune encephalomyelitis (EAE) PLoS One. 2013;8:e55948. doi: 10.1371/journal.pone.0055948.
    1. Syed YA, Hand E, Möbius W, Zhao C, Hofer M, Nave KA, Kotter MR. Inhibition of CNS remyelination by the presence of semaphorin 3A. J Neurosci. 2011;31:3719–3728. doi: 10.1523/JNEUROSCI.4930-10.2011.
    1. Williams A, Piaton G, Aigrot MS, Belhadi A, Théaudin M, Petermann F, Thomas JL, Zalc B, Lubetzki C. Semaphorin 3A and 3 F: key players in myelin repair in multiple sclerosis? Brain. 2007;130:2554–2565. doi: 10.1093/brain/awm202.
    1. Okuno T, Nakatsuji Y, Kumanogoh A. The role of immune semaphorins in multiple sclerosis. FEBS Lett. 2011;585:3829–3835. doi: 10.1016/j.febslet.2011.03.033.
    1. Solomon BD, Mueller C, Chae WJ, Alabanza LM, Bynoe MS. Neuropilin-1 attenuates autoreactivity in experimental autoimmune encephalomyelitis. Proc Natl Acad Sci U S A. 2011;108:2040–2045. doi: 10.1073/pnas.1008721108.
    1. Liu XS, Chopp M, Zhang RL, Hozeska-Solgot A, Gregg SC, Buller B, Lu M, Zhang ZG. Angiopoietin 2 mediates the differentiation and migration of neural progenitor cells in the subventricular zone after stroke. J Biol Chem. 2009;284:22680–22689. doi: 10.1074/jbc.M109.006551.
    1. Wang L, Zhang Z, Wang Y, Zhang R, Chopp M. Treatment of stroke with erythropoietin enhances neurogenesis and angiogenesis and improves neurological function in rats. Stroke. 2004;35:1732–1737. doi: 10.1161/01.STR.0000132196.49028.a4.
    1. Spence RD, Voskuhl RR. Neuroprotective effects of estrogens and androgens in CNS inflammation and neurodegeneration. Front Neuroendocrinol. 2012;33:105–115. doi: 10.1016/j.yfrne.2011.12.001.
    1. Suzuki S, Gerhold LM, Böttner M, Rau SW, Dela Cruz C, Yang E, Zhu H, Yu J, Cashion AB, Kindy MS, Merchenthaler I, Gage FH, Wise PM. Estradiol enhances neurogenesis following ischemic stroke through estrogen receptors alpha and beta. J Comp Neurol. 2007;500:1064–1075. doi: 10.1002/cne.21240.
    1. Crawford DK, Mangiardi M, Song B, Patel R, Du S, Sofroniew MV, Voskuhl RR, Tiwari-Woodruff SK. Oestrogen receptor beta ligand: a novel treatment to enhance endogenous functional remyelination. Brain. 2010;133:2999–3016. doi: 10.1093/brain/awq237.
    1. Zhang R, Wang L, Zhang L, Chen J, Zhu Z, Zhang Z, Chopp M. Nitric oxide enhances angiogenesis via the synthesis of vascular endothelial growth factor and cGMP after stroke in the rat. Circ Res. 2003;92:308–313. doi: 10.1161/01.RES.0000056757.93432.8C.
    1. Zhang R, Zhang L, Zhang Z, Wang Y, Lu M, Lapointe M, Chopp M. A nitric oxide donor induces neurogenesis and reduces functional deficits after stroke in rats. Ann Neurol. 2001;50:602–611. doi: 10.1002/ana.1249.
    1. Li Q, Ford MC, Lavik EB, Madri JA. Modeling the neurovascular niche: VEGF- and BDNF-mediated cross-talk between neural stem cells and endothelial cells: an in vitro study. J Neurosci Res. 2006;84:1656–1668. doi: 10.1002/jnr.21087.
    1. Fong GH. Mechanisms of adaptive angiogenesis to tissue hypoxia. Angiogenesis. 2008;11:121–140. doi: 10.1007/s10456-008-9107-3.
    1. Mohyeldin A, Garzon-Muvdi T, Quinones-Hinojosa A. Oxygen in stem cell biology: a critical component of the stem cell niche. Cell Stem Cell. 2010;7:150–161. doi: 10.1016/j.stem.2010.07.007.
    1. Imitola J, Raddassi K, Park KI, Mueller FJ, Nieto M, Teng YD, Frenkel D, Li J, Sidman RL, Walsh CA, Snyder EY, Khoury SJ. Directed migration of neural stem cells to sites of CNS injury by the stromal cell-derived factor 1alpha/CXC chemokine receptor 4 pathway. Proc Natl Acad Sci U S A. 2004;101:18117–18122. doi: 10.1073/pnas.0408258102.
    1. Arai K, Lo EH. An oligovascular niche: cerebral endothelial cells promote the survival and proliferation of oligodendrocyte precursor cells. J Neurosci. 2009;29:4351–4355. doi: 10.1523/JNEUROSCI.0035-09.2009.
    1. Chang A, Nishiyama A, Peterson J, Prineas J, Trapp BD. NG2-positive oligodendrocyte progenitor cells in adult human brain and multiple sclerosis lesions. J Neurosci. 2000;20:6404–6412.
    1. Chang A, Tourtellotte WW, Rudick R, Trapp BD. Premyelinating oligodendrocytes in chronic lesions of multiple sclerosis. N Engl J Med. 2002;346:165–173. doi: 10.1056/NEJMoa010994.
    1. Gadea A, Aguirre A, Haydar TF, Gallo V. Endothelin-1 regulates oligodendrocyte development. J Neurosci. 2009;29:10047–10062. doi: 10.1523/JNEUROSCI.0822-09.2009.
    1. Greenberg DA, Jin K. From angiogenesis to neuropathology. Nature. 2005;438:954–959. doi: 10.1038/nature04481.
    1. Sanai N, Tramontin AD, Quinones-Hinojosa A, Barbaro NM, Gupta N, Kunwar S, Lawton MT, McDermott MW, Parsa AT, Manuel-Garcia Verdugo J, Berger MS, Alvarez-Buylla A. Unique astrocyte ribbon in adult human brain contains neural stem cells but lacks chain migration. Nature. 2004;427:740–744. doi: 10.1038/nature02301.
    1. Pluchino S, Muzio L, Imitola J, Deleidi M, Alfaro-Cervello C, Salani G, Porcheri C, Brambilla E, Cavasinni F, Bergamaschi A, Garcia-Verdugo JM, Comi G, Khoury SJ, Martino G. Persistent inflammation alters the function of the endogenous brain stem cell compartment. Brain. 2008;131:2564–2578. doi: 10.1093/brain/awn198.
    1. Rasmussen S, Imitola J, Ayuso-Sacido A, Wang Y, Starossom SC, Kivisäkk P, Zhu B, Meyer M, Bronson RT, Garcia-Verdugo JM, Khoury SJ. Reversible neural stem cell niche dysfunction in a model of multiple sclerosis. Ann Neurol. 2011;69:878–891. doi: 10.1002/ana.22299.
    1. Lehtinen MK, Zappaterra MW, Chen X, Yang YJ, Hill AD, Lun M, Maynard T, Gonzalez D, Kim S, Ye P, D'Ercole AJ, Wong ET, LaMantia AS, Walsh CA. The cerebrospinal fluid provides a proliferative niche for neural progenitor cells. Neuron. 2011;69:893–905. doi: 10.1016/j.neuron.2011.01.023.
    1. Sun X, Tanaka M, Kondo S, Okamoto K, Hirai S. Clinical significance of reduced cerebral metabolism in multiple sclerosis: a combined PET and MRI study. Ann Nucl Med. 1998;12:89–94. doi: 10.1007/BF03164835.
    1. Swank RL, Roth JG, Woody DC., Jr Cerebral blood flow and red cell delivery in normal subjects and in multiple sclerosis. Neurol Res. 1983;5:37–59.
    1. Law M, Saindane AM, Ge Y, Babb JS, Johnson G, Mannon LJ, Herbert J, Grossman RI. Microvascular abnormality in relapsing-remitting multiple sclerosis: perfusion MR imaging findings in normal-appearing white matter. Radiology. 2004;231:645–652. doi: 10.1148/radiol.2313030996.
    1. Varga AW, Johnson G, Babb JS, Herbert J, Grossman RI, Inglese M. White matter hemodynamic abnormalities precede sub-cortical gray matter changes in multiple sclerosis. J Neurol Sci. 2009;282:28–33. doi: 10.1016/j.jns.2008.12.036.
    1. Adhya S, Johnson G, Herbert J, Jaggi H, Babb JS, Grossman RI, Inglese M. Pattern of hemodynamic impairment in multiple sclerosis: dynamic susceptibility contrast perfusion MR imaging at 3.0 T. Neuroimage. 2006;33:1029–1035. doi: 10.1016/j.neuroimage.2006.08.008.
    1. Rashid W, Parkes LM, Ingle GT, Chard DT, Toosy AT, Altmann DR, Symms MR, Tofts PS, Thompson AJ, Miller DH. Abnormalities of cerebral perfusion in multiple sclerosis. J Neurol Neurosurg Psychiatry. 2004;75:1288–1293. doi: 10.1136/jnnp.2003.026021.
    1. Juurlink BH. The multiple sclerosis lesion: initiated by a localized hypoperfusion in a central nervous system where mechanisms allowing leukocyte infiltration are readily upregulated? Med Hypotheses. 1998;51:299–303. doi: 10.1016/S0306-9877(98)90052-4.
    1. Haselhorst R, Kappos L, Bilecen D, Scheffler K, Möri D, Radü EW, Seelig J. Dynamic susceptibility contrast MR imaging of plaque development in multiple sclerosis: application of an extended blood–brain barrier leakage correction. J Magn Reson Imaging. 2000;11:495–505. doi: 10.1002/(SICI)1522-2586(200005)11:5<495::AID-JMRI5>;2-S.
    1. Wuerfel J, Bellmann-Strobl J, Brunecker P, Aktas O, McFarland H, Villringer A, Zipp F. Changes in cerebral perfusion precede plaque formation in multiple sclerosis: a longitudinal perfusion MRI study. Brain. 2004;127:111–119. doi: 10.1093/brain/awh007.
    1. Jiang Q, Zhang ZG, Ding GL, Zhang L, Ewing JR, Wang L, Zhang R, Li L, Lu M, Meng H, Arbab AS, Hu J, Li QJ, Pourabdollah Nejad DS, Athiraman H, Chopp M. Investigation of neural progenitor cell induced angiogenesis after embolic stroke in rat using MRI. Neuroimage. 2005;28:698–707. doi: 10.1016/j.neuroimage.2005.06.063.
    1. Hiehle JF, Jr, Lenkinski RE, Grossman RI, Dousset V, Ramer KN, Schnall MD, Cohen JA, Gonzalez-Scarano F. Correlation of spectroscopy and magnetization transfer imaging in the evaluation of demyelinating lesions and normal appearing white matter in multiple sclerosis. Magn Reson Med. 1994;32:285–293. doi: 10.1002/mrm.1910320303.
    1. De Keyser J, Steen C, Mostert JP, Koch MW. Hypoperfusion of the cerebral white matter in multiple sclerosis: possible mechanisms and pathophysiological significance. J Cereb Blood Flow Metab. 2008;28:1645–1651. doi: 10.1038/jcbfm.2008.72.
    1. Steen C, Wilczak N, Hoogduin JM, Koch M, De Keyser J. Reduced creatine kinase B activity in multiple sclerosis normal appearing white matter. PLoS One. 2010;5:e10811. doi: 10.1371/journal.pone.0010811.
    1. D'haeseleer M, Cambron M, Vanopdenbosch L, De Keyser J. Vascular aspects of multiple sclerosis. Lancet Neurol. 2011;10:657–666. doi: 10.1016/S1474-4422(11)70105-3.
    1. Brück W, Lucchinetti C, Lassmann H. The pathology of primary progressive multiple sclerosis. Mult Scler. 2002;8:93–97. doi: 10.1191/1352458502ms785rr.
    1. Dore-Duffy P, Wencel M, Katyshev V, Cleary K. Chronic mild hypoxia ameliorates chronic inflammatory activity in myelin oligodendrocyte glycoprotein (MOG) peptide induced experimental autoimmune encephalomyelitis (EAE) Adv Exp Med Biol. 2011;701:165–173. doi: 10.1007/978-1-4419-7756-4_23.
    1. Esen N, Serkin Z, Dore-Duffy P. Induction of vascular remodeling: a novel therapeutic approach in EAE. J Neurol Sci. 2013;333:88–92. doi: 10.1016/j.jns.2013.06.004.
    1. Lu F, Selak M, O'Connor J, Croul S, Lorenzana C, Butunoi C, Kalman B. Oxidative damage to mitochondrial DNA and activity of mitochondrial enzymes in chronic active lesions of multiple sclerosis. J Neurol Sci. 2000;177:95–103. doi: 10.1016/S0022-510X(00)00343-9.
    1. Haider L, Fischer MT, Frischer JM, Bauer J, Höftberger R, Botond G, Esterbauer H, Binder CJ, Witztum JL, Lassmann H. Oxidative damage in multiple sclerosis lesions. Brain. 2011;134:1914–1924. doi: 10.1093/brain/awr128.
    1. Gironi M, Borgiani B, Mariani E, Cursano C, Mendozzi L, Cavarretta R, Saresella M, Clerici M, Comi G, Rovaris M, Furlan R. Oxidative stress is differentially present in multiple sclerosis courses, early evident, and unrelated to treatment. J Immunol Res. 2014;2014:961863. doi: 10.1155/2014/961863.
    1. Bolanos JP, Almeida A, Stewart V, Peuchen S, Land JM, Clark JB, Heales SJ. Nitric oxide-mediated mitochondrial damage in the brain: mechanisms and implications for neurodegenerative diseases. J Neurochem. 1997;68:2227–2240. doi: 10.1046/j.1471-4159.1997.68062227.x.
    1. Aboul-Enein F, Rauschka H, Kornek B, Stadelmann C, Stefferl A, Brück W, Lucchinetti C, Schmidbauer M, Jellinger K, Lassmann H. 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. doi: 10.1093/jnen/62.1.25.
    1. Redford EJ, Kapoor R, Smith KJ. Nitric oxide donors reversibly block axonal conduction: demyelinated axons are especially susceptible. Brain. 1997;120:2149–2157. doi: 10.1093/brain/120.12.2149.
    1. Amorini AM, Nociti V, Petzold A, Gasperini C, Quartuccio E, Lazzarino G, Di Pietro V, Belli A, Signoretti S, Vagnozzi R, Lazzarino G, Tavazzi B. Serum lactate as a novel potential biomarker in multiple sclerosis. Biochim Biophys Acta. 2014;1842:1137–1143. doi: 10.1016/j.bbadis.2014.04.005.
    1. Simone IL, Federico F, Trojano M, Tortorella C, Liguori M, Giannini P, Picciola E, Natile G, Livrea P. High resolution proton MR spectroscopy of cerebrospinal fluid in MS patients. Comparison with biochemical changes in demyelinating plaques. J Neurol Sci. 1996;144:182–190. doi: 10.1016/S0022-510X(96)00224-9.
    1. Fischer MT, Sharma R, Lim JL, Haider L, Frischer JM, Drexhage J, Mahad D, Bradl M, van Horssen J, Lassmann H. NADPH oxidase expression in active multiple sclerosis lesions in relation to oxidative tissue damage and mitochondrial injury. Brain. 2012;135:886–899. doi: 10.1093/brain/aws012.
    1. Campbell GR, Kraytsberg Y, Krishnan KJ, Ohno N, Ziabreva I, Reeve A, Trapp BD, Newcombe J, Reynolds R, Lassmann H, Khrapko K, Turnbull DM, Mahad DJ. Clonally expanded mitochondrial DNA deletions within the choroid plexus in multiple sclerosis. Acta Neuropathol. 2012;124:209–220. doi: 10.1007/s00401-012-1001-9.
    1. Pandit A, Vadnal J, Houston S, Freeman E, McDonough J. Impaired regulation of electron transport chain subunit genes by nuclear respiratory factor 2 in multiple sclerosis. J Neurol Sci. 2009;279:14–20. doi: 10.1016/j.jns.2009.01.009.
    1. Pache M, Kaiser HJ, Akhalbedashvili N, Lienert C, Dubler B, Kappos L, Flammer J. Extraocular blood flow and endothelin-1 plasma levels in patients with multiple sclerosis. Eur Neurol. 2003;49:164–168. doi: 10.1159/000069085.
    1. Speciale L, Sarasella M, Ruzzante S, Caputo D, Mancuso R, Calvo MG, Guerini FR, Ferrante P. Endothelin and nitric oxide levels in cerebrospinal fluid of patients with multiple sclerosis. J Neurovirol. 2000;6:S62–66.
    1. Matsushita T, Isobe N, Kawajiri M, Mogi M, Tsukuda K, Horiuchi M, Ohyagi Y, Kira J. CSF angiotensin II and angiotensin-converting enzyme levels in anti-aquaporin-4 autoimmunity. J Neurol Sci. 2010;295:41–45. doi: 10.1016/j.jns.2010.05.014.
    1. Constantinescu CS, Goodman DB, Grossman RI, Mannon LJ, Cohen JA. Serum angiotensin-converting enzyme in multiple sclerosis. Arch Neurol. 1997;54:1012–1015. doi: 10.1001/archneur.1997.00550200068012.
    1. Platten M, Youssef S, Hur EM, Ho PP, Han MH, Lanz TV, Phillips LK, Goldstein MJ, Bhat R, Raine CS, Sobel RA, Steinman L. Blocking angiotensin-converting enzyme induces potent regulatory T cells and modulates TH1- and TH17-mediated autoimmunity. Proc Natl Acad Sci U S A. 2009;106:14948–14953. doi: 10.1073/pnas.0903958106.
    1. Griffioen AW, Molema G. Angiogenesis: potentials for pharmacologic intervention in the treatment of cancer, cardiovascular diseases, and chronic inflammation. Pharmacol Rev. 2000;52:237–268.
    1. Krum JM, Khaibullina A. Inhibition of endogenous VEGF impende revascularization and astroglial proliferation: roles for VEGF in brain repair. Exp Neurol. 2003;181:241–257. doi: 10.1016/S0014-4886(03)00039-6.
    1. Bouérat L, Fensholdt J, Liang X, Havez S, Nielsen SF, Hansen JR, Bolvig S, Andersson C. Indolin-2-ones with high in vivo efficacy in a model for multiple sclerosis. J Med Chem. 2005;48:5412–5414. doi: 10.1021/jm0504151.
    1. Mohty M, Brissot E, Savani BN, Gaugler B. Effects of bortezomib on the immune system: a focus on immune regulation. Biol Blood Marrow Transplant. 2013;19:1416–1420. doi: 10.1016/j.bbmt.2013.05.011.
    1. Dredge K, Marriott JB, Dalgleish AG. Immunological effects of thalidomide and its chemical and functional analogs. Crit Rev Immunol. 2002;22:425–437. doi: 10.1615/CritRevImmunol.v22.i5-6.40.
    1. Sastry PS. Inhibition of TNF-alpha synthesis with thalidomide for prevention of acute exacerbations and altering the natural history of multiple sclerosis. Med Hypotheses. 1999;53:76–77. doi: 10.1054/mehy.1997.0719.
    1. Sharief MK, Thompson EJ. In vivo relationship of tumor necrosis factor-alpha to blood–brain barrier damage in patients with active multiple sclerosis. J Neuroimmunol. 1992;38:27–33. doi: 10.1016/0165-5728(92)90087-2.
    1. Faure M, Lejeune JP, Gaucherand M, Thivolet J. PMN leukocytes chemotaxis: inhibition by thalidomide. Pathol Biol (Paris) 1981;9:601–604.
    1. McHugh SM, Rifkin IR, Deighton J, Wilson AB, Lachmann PJ, Lockwood CM, Ewan PW. The immunosuppressive drug thalidomide induces T helper cell type 2 (Th2) and concomitantly inhibits Th1 cytokine production in mitogen- and antigen-stimulated human peripheral blood mononuclear cell cultures. Clin Exp Immunol. 1995;99:160–167. doi: 10.1111/j.1365-2249.1995.tb05527.x.
    1. Haslett PA, Corral LG, Albert M, Kaplan G. Thalidomide costimulates primary human T lymphocytes, preferentially inducing proliferation, cytokine production, and cytotoxic responses in the CD8+ subset. J Exp Med. 1998;187:1885–1892. doi: 10.1084/jem.187.11.1885.
    1. Ryu JK, Jantaratnotai N, McLarnon JG. Thalidomide inhibition of vascular remodeling and inflammatory reactivity in the quinolinic acid-injected rat striatum. Neuroscience. 2009;163:601–608. doi: 10.1016/j.neuroscience.2009.07.006.
    1. Contino-Pépin C, Parat A, Périno S, Lenoir C, Vidal M, Galons H, Karlik S, Pucci B. Preliminary biological evaluations of new thalidomide analogues for multiple sclerosis application. Bioorg Med Chem Lett. 2009;19:878–881. doi: 10.1016/j.bmcl.2008.11.118.
    1. MacLean HJ, Freedman MS. Immunologic therapy for relapsing – remitting multiple sclerosis. Curr Neurol Neurosci Rep. 2001;1:277–85. doi: 10.1007/s11910-001-0031-9.
    1. Folkman J, Langer R, Linhardt RJ, Haudenschild C, Taylor S. Angiogenesis inhibition and tumor regression caused by heparin or a heparin fragment in the presence of cortisone. Science. 1983;221:719–725. doi: 10.1126/science.6192498.
    1. Nauck M, Karakiulakis G, Perruchoud AP, Papakonstantinou E, Roth M. Corticosteroids inhibit the expression of the vascular endothelial growth factor gene in human vascular smooth muscle cells. Eur J Pharmacol. 1998;341:309–315. doi: 10.1016/S0014-2999(97)01464-7.
    1. Jablonska J, Leschner S, Westphal K, Lienenklaus S, Weiss S. Neutrophils responsive to endogenous IFN-beta regulate tumor angiogenesis and growth in a mouse tumor model. J Clin Invest. 2010;120:1151–1164. doi: 10.1172/JCI37223.
    1. Taylor KL, Leaman DW, Grane R, Mechti N, Borden EC, Lindner DJ. Identification of interferon-beta-stimulated genes that inhibit angiogenesis in vitro. J Interferon Cytokine Res. 2008;28:733–740. doi: 10.1089/jir.2008.0030.
    1. Ewalt KL, Schimmel P. Activation of angiogenic signaling pathways by two human tRNA synthetases. Biochemistry. 2002;41:13344–13349. doi: 10.1021/bi020537k.
    1. Thamilarasan M, Hecker M, Goertsches RH, Paap BK, Schröder I, Koczan D, Thiesen HJ, Zettl UK. Glatiramer acetate treatment effects on gene expression in monocytes of multiple sclerosis patients. J Neuroinflammation. 2013;10:126. doi: 10.1186/1742-2094-10-126.
    1. Jin H, Su J, Garmy-Susini B, Kleeman J, Varner J. Integrin alpha4beta1 promotes monocyte trafficking and angiogenesis in tumors. Cancer Res. 2006;66:2146–2152. doi: 10.1158/0008-5472.CAN-05-2704.
    1. Podar K, Zimmerhackl A, Fulciniti M, Tonon G, Hainz U, Tai YT, Vallet S, Halama N, Jäger D, Olson DL, Sattler M, Chauhan D, Anderson KC. The selective adhesion molecule inhibitor Natalizumab decreases multiple myeloma cell growth in the bone marrow microenvironment: therapeutic implications. Br J Haematol. 2011;155:438–448. doi: 10.1111/j.1365-2141.2011.08864.x.
    1. Foster CA, Mechtcheriakova D, Storch MK, Balatoni B, Howard LM, Bornancin F, Wlachos A, Sobanov J, Kinnunen A, Baumruker T. FTY720 rescue therapy in the dark agouti rat model of experimental autoimmune encephalomyelitis: expression of central nervous system genes and reversal of blood–brain-barrier damage. Brain Pathol. 2009;19:254–266. doi: 10.1111/j.1750-3639.2008.00182.x.
    1. Miron VE, Schubart A, Antel JP. Central nervous system-directed effects of FTY720 (fingolimod) J Neurol Sci. 2008;274:13–17. doi: 10.1016/j.jns.2008.06.031.
    1. Mousseau Y, Mollard S, Richard L, Nizou A, Faucher-Durand K, Cook-Moreau J, Qiu H, Baaj Y, Funalot B, Fourcade L, Sturtz FG. Fingolimod inhibits PDGF-B-induced migration of vascular smooth muscle cell by down-regulating the S1PR1/S1PR3 pathway. Biochimie. 2012;94:2523–2531. doi: 10.1016/j.biochi.2012.07.002.
    1. Pulaski HL, Spahlinger G, Silva IA, McLean K, Kueck AS, Reynolds RK, Coukos G, Conejo-Garcia JR, Buckanovich RJ. Identifying alemtuzumab as an anti-myeloid cell antiangiogenic therapy for the treatment of ovarian cancer. J Transl Med. 2009;7:49. doi: 10.1186/1479-5876-7-49.
    1. Arnold P, Mojumder D, Detoledo J, Lucius R, Wilms H. Pathophysiological processes in multiple sclerosis: focus on nuclear factor erythroid-2-related factor 2 and emerging pathways. Clin Pharmacol. 2014;6:35–42.
    1. García-Caballero M, Marí-Beffa M, Medina MÁ, Quesada AR. Dimethylfumarate inhibits angiogenesis in vitro and in vivo: a possible role for its antipsoriatic effect? J Invest Dermatol. 2011;131:1347–1355. doi: 10.1038/jid.2010.416.
    1. Billington DC. Angiogenesis and its inhibition: potential new therapies in oncology and non-neoplastic diseases. Drug Des Discov. 1991;8:3–35.
    1. Patten SG, Adamcic U, Lacombe K, Minhas K, Skowronski K, Coomber BL. VEGFR2 heterogeneity and response to anti-angiogenic low dose metronomic cyclophosphamide treatment. BMC Cancer. 2010;10:683. doi: 10.1186/1471-2407-10-683.
    1. Gora-Tybor J, Blonski JZ, Robak T. Cladribine decreases the level of angiogenic factors in patients with chronic lymphocytic leukemia. Neoplasma. 2002;49:145–148.
    1. Wu X, Zhong H, Song J, Damoiseaux R, Yang Z, Lin S. Mycophenolic acid is a potent inhibitor of angiogenesis. Arterioscler Thromb Vasc Biol. 2006;26:2414–2416. doi: 10.1161/01.ATV.0000238361.07225.fc.
    1. Brundula V, Rewcastle NB, Metz LM, Bernard CC, Yong VW. Targeting leukocyte MMPs and transmigration Minocycline as a potential therapy for multiple sclerosis. Brain. 2002;125:1297–1308. doi: 10.1093/brain/awf133.
    1. Weingart JD, Sipos EP, Brem H. The role of minocycline in the treatment of intracranial 9 L glioma. J Neurosurg. 1995;82:635–640. doi: 10.3171/jns.1995.82.4.0635.
    1. Tamargo RJ, Bok RA, Brem H. Angiogenesis inhibition by minocycline. Cancer Res. 1991;51:672–675.
    1. Yao JS, Chen Y, Zhai W, Xu K, Young WL, Yang GY. Minocycline exerts multiple inhibitory effects on vascular endothelial growth factor-induced smooth muscle cell migration: the role of ERK1/2, PI3K, and matrix metalloproteinases. Circ Res. 2004;95:364–371. doi: 10.1161/01.RES.0000138581.04174.2f.
    1. Garrido-Mesa N, Zarzuelo A, Gálvez J. Minocycline: far beyond an antibiotic. Br J Pharmacol. 2013;169:337–352. doi: 10.1111/bph.12139.
    1. Hauser SL, Oksenberg JR. The neurobiology of multiple sclerosis: genes, inflammation, and neurodegeneration. Neuron. 2006;52:61–76. doi: 10.1016/j.neuron.2006.09.011.
    1. Rovaris M, Confavreux C, Furlan R, Kappos L, Comi G, Filippi M. Secondary progressive multiple sclerosis: current knowledge and future challenges. Lancet Neurol. 2006;5:343–354. doi: 10.1016/S1474-4422(06)70410-0.
    1. Herrán E, Pérez-González R, Igartua M, Pedraz JL, Carro E, Hernández RM. VEGF-releasing biodegradable nanospheres administered by craniotomy: a novel therapeutic approach in the APP/Ps1 mouse model of Alzheimer's disease. J Control Release. 2013;170:111–119. doi: 10.1016/j.jconrel.2013.04.028.
    1. Hwang DH, Lee HJ, Park IH, Seok JI, Kim BG, Joo IS, Kim SU. Intrathecal transplantation of human neural stem cells overexpressing VEGF provide behavioral improvement, disease onset delay and survival extension in transgenic ALS mice. Gene Ther. 2009;16:1234–1244. doi: 10.1038/gt.2009.80.
    1. Tian YY, Tang CJ, Wang JN, Feng Y, Chen XW, Wang L, Qiao X, Sun SG. Favorable effects of VEGF gene transfer on a rat model of Parkinson’s disease using adeno-associated viral vectors. Neurosci Lett. 2007;421:239–244. doi: 10.1016/j.neulet.2007.05.033.
    1. Azzouz M, Ralph GS, Storkebaum E, Walmsley LE, Mitrophanous KA, Kingsman SM, Carmeliet P, Mazarakis ND. VEGF delivery with retrogradely transported lentivector prolongs survival in a mouse ALS model. Nature. 2004;429:413–417. doi: 10.1038/nature02544.
    1. Sun Y, Jin K, Xie L, Childs J, Mao XO, Logvinova A, Greenberg DA. VEGF-induced neuroprotection, neurogenesis, and angiogenesis after focal cerebral ischemia. J Clin Invest. 2003;111:1843–1851. doi: 10.1172/JCI200317977.
    1. Tang Z, Yin JX, Han P, Gan Y, Coons SW, Wang C, Maalouf M, Shi J. Pertussis toxin attenuates experimental autoimmune encephalomyelitis by upregulating neuronal vascular endothelial growth factor. Neuroreport. 2013;24:469–475. doi: 10.1097/WNR.0b013e3283619fc8.
    1. Nowak DG, Woolard J, Amin EM, Konopatskaya O, Saleem MA, Churchill AJ, Ladomery MR, Harper SJ, Bates DO. Expression of pro- and anti-angiogenic isoforms of VEGF is differentially regulated by splicing and growth factors. J Cell Sci. 2008;121:3487–3495. doi: 10.1242/jcs.016410.
    1. Magnussen AL, Rennel ES, Hua J, Bevan HS, Beazley Long N, Lehrling C, Gammons M, Floege J, Harper SJ, Agostini HT, Bates DO, Churchill AJ. VEGF-A165b is cytoprotective and antiangiogenic in the retina. Invest Ophthalmol Vis Sci. 2010;51:4273–4281. doi: 10.1167/iovs.09-4296.
    1. MacMillan CJ, Doucette CD, Warford J, Furlong SJ, Hoskin DW, Easton AS. Murine experimental autoimmune encephalomyelitis is diminished by treatment with the angiogenesis inhibitors B20-4.1.1 and angiostatin (K1-3) PLoS One. 2014;9:e89770. doi: 10.1371/journal.pone.0089770.
    1. Lopez-Ramirez MA, Wu D, Pryce G, Simpson JE, Reijerkerk A, King-Robson J, Kay O, de Vries HE, Hirst MC, Sharrack B, Baker D, Male DK, Michael GJ, Romero IA. MicroRNA-155 negatively affects blood–brain barrier function during neuroinflammation. FASEB J. 2014
    1. Munger KL, Zhang SM, O'Reilly E, Hernán MA, Olek MJ, Willett WC, Ascherio A. Vitamin D intake and incidence of multiple sclerosis. Neurology. 2004;62:60–65. doi: 10.1212/01.WNL.0000101723.79681.38.
    1. Grundmann M, Haidar M, Placzko S, Niendorf R, Darashchonak N, Hubel CA, von Versen-Höynck F. Vitamin D improves the angiogenic properties of endothelial progenitor cells. Am J Physiol Cell Physiol. 2012;303:C954–962. doi: 10.1152/ajpcell.00030.2012.
    1. Gazzerro P, Proto MC, Gangemi G, Malfitano AM, Ciaglia E, Pisanti S, Santoro A, Laezza C, Bifulco M. Pharmacological actions of statins: a critical appraisal in the management of cancer. Pharmacol Rev. 2012;64:102–146. doi: 10.1124/pr.111.004994.
    1. Wang B, Sun L, Tian Y, Li Z, Wei H, Wang D, Yang Z, Chen J, Zhang J, Jiang R. Effects of atorvastatin in the regulation of circulating EPCs and angiogenesis in traumatic brain injury in rats. J Neurol Sci. 2012;319:117–123. doi: 10.1016/j.jns.2012.04.015.
    1. Birnbaum G, Cree B, Altafullah I, Zinser M, Reder AT. Combining beta interferon and atorvastatin may increase disease activity in multiple sclerosis. Neurology. 2008;71:1390–1395. doi: 10.1212/01.wnl.0000319698.40024.1c.
    1. Kamm CP, El-Koussy M, Humpert S, Findling O, Burren Y, Schwegler G, Donati F, Müller M, Müller F, Slotboom J, Kappos L, Naegelin Y, Mattle HP, SWABIMS Study Group Atorvastatin added to interferon beta for relapsing multiple sclerosis: 12-month treatment extension of the randomized multicenter SWABIMS Trial. PLoS One. 2014;9:e86663. doi: 10.1371/journal.pone.0086663.
    1. Stüve O, Youssef S, Weber MS, Nessler S, von Büdingen HC, Hemmer B, Prod'homme T, Sobel RA, Steinman L, Zamvil SS. Immunomodulatory synergy by combination of atorvastatin and glatiramer acetate in treatment of CNS autoimmunity. J Clin Invest. 2006;116:1037–1044. doi: 10.1172/JCI25805.
    1. Sorensen PS, Lycke J, Erälinna JP, Edland A, Wu X, Frederiksen JL, Oturai A, Malmeström C, Stenager E, Sellebjerg F, Sondergaard HB, SIMCOMBIN study investigators Simvastatin as add-on therapy to interferon β-1a for relapsing-remitting multiple sclerosis (SIMCOMBIN study): a placebo-controlled randomised phase 4 trial. Lancet Neurol. 2011;10:691–701. doi: 10.1016/S1474-4422(11)70144-2.
    1. Dell'Acqua ML, Lorenzini L, D'Intino G, Sivilia S, Pasqualetti P, Panetta V, Paradisi M, Filippi MM, Baiguera C, Pizzi M, Giardino L, Rossini PM, Calzà L. Functional and molecular evidence of myelin- and neuroprotection by thyroid hormone administration in experimental allergic encephalomyelitis. Neuropathol Appl Neurobiol. 2012;38:454–470. doi: 10.1111/j.1365-2990.2011.01228.x.
    1. Bartels C, Späte K, Krampe H, Ehrenreich H. Recombinant human Erythropoietin: novel strategies for neuroprotective/neuro-regenerative treatment of multiple sclerosis. Ther Adv Neurol Disord. 2008;1:193–206. doi: 10.1177/1756285608098422.
    1. Créange A, Lefaucheur JP, Balleyguier MO, Galactéros F. Iron depletion induced by bloodletting and followed by rhEPO administration as a therapeutic strategy in progressive multiple sclerosis: a pilot, open-label study with neurophysiological measurements. Neurophysiol Clin. 2013;43:303–312. doi: 10.1016/j.neucli.2013.09.004.
    1. Ehrenreich H, Fischer B, Norra C, Schellenberger F, Stender N, Stiefel M, Sirén AL, Paulus W, Nave KA, Gold R, Bartels C. Exploring recombinant human erythropoietin in chronic progressive multiple sclerosis. Brain. 2007;130:2577–2588. doi: 10.1093/brain/awm203.
    1. Najmi Varzaneh F, Najmi Varzaneh F, Azimi AR, Rezaei N, Sahraian MA. Efficacy of combination therapy with erythropoietin and methylprednisolone in clinical recovery of severe relapse in multiple sclerosis. Acta Neurol Belg. 2014;ᅟ:114.
    1. Balashov KE, Aung LL, Dhib-Jalbut S, Keller IA. Acute multiple sclerosis lesion: conversion of restricted diffusion due to vasogenic edema. J Neuroimaging. 2011;21:202–204. doi: 10.1111/j.1552-6569.2009.00443.x.
    1. Tievsky AL, Ptak T, Farkas J. Investigation of apparent diffusion coefficient and diffusion tensor anisotrophy in acute and chronic multiple sclerosis lesions. AJNR Am J Neuroradiol. 1999;20:1491–1499.
    1. Zlokovic BV. The blood–brain barrier in health and chronic neurodegenerative disorders. Neuron. 2008;57:178–201. doi: 10.1016/j.neuron.2008.01.003.
    1. Muramatsu R, Takahashi C, Miyake S, Fujimura H, Mochizuki H, Yamashita T. Angiogenesis induced by CNS inflammation promotes neuronal remodeling through vessel-derived prostacyclin. Nat Med. 2012;18:1658–1664. doi: 10.1038/nm.2943.
    1. Li L, Welser JV, Dore-Duffy P, del Zoppo GJ, Lamanna JC, Milner R. In the hypoxic central nervous system, endothelial cell proliferation is followed by astrocyte activation, proliferation, and increased expression of the alpha 6 beta 4 integrin and dystroglycan. Glia. 2010;58:1157–1167.
    1. Palavra F, Marado D, Mascarenhas-Melo F, Sereno J, Teixeira-Lemos E, Nunes CC, Gonçalves G, Teixeira F, Reis F. New markers of early cardiovascular risk in multiple sclerosis patients: oxidized-LDL correlates with clinical staging. Dis Markers. 2013;34:341–348. doi: 10.1155/2013/567162.
    1. Chaitanya GV, Omura S, Sato F, Martinez NE, Minagar A, Ramanathan M, Guttman BW, Zivadinov R, Tsunoda I, Alexander JS. Inflammation induces neuro-lymphatic protein expression in multiple sclerosis brain neurovasculature. J Neuroinflammation. 2013;10:125. doi: 10.1186/1742-2094-10-125.
    1. Sarchielli P, Di Filippo M, Ercolani MV, Chiasserini D, Mattioni A, Bonucci M, Tenaglia S, Eusebi P, Calabresi P. Fibroblast growth factor-2 levels are elevated in the cerebrospinal fluid of multiple sclerosis patients. Neurosci Lett. 2008;435:223–228. doi: 10.1016/j.neulet.2008.02.040.
    1. Rieckmann P, Albrecht M, Ehrenreich H, Weber T, Michel U. Semi-quantitative analysis of cytokine gene expression in blood and cerebrospinal fluid cells by reverse transcriptase polymerase chain reaction. Res Exp Med (Berl) 1995;195:17–29. doi: 10.1007/BF02576770.
    1. Giovannoni G. Cerebrospinal fluid and serum nitric oxide metabolites in patients with multiple sclerosis. Mult Scler. 1998;4:27–30. doi: 10.1191/135245898678909178.
    1. Sarchielli P, Orlacchio A, Vicinanza F, Pelliccioli GP, Tognoloni M, Saccardi C, Gallai V. Cytokine secretion and nitric oxide production by mononuclear cells of patients with multiple sclerosis. J Neuroimmunol. 1997;80:76–86. doi: 10.1016/S0165-5728(97)00136-7.
    1. Maimone D, Gregory S, Arnason BG, Reder AT. Cytokine levels in the cerebrospinal fluid and serum of patients with multiple sclerosis. J Neuroimmunol. 1991;32:67–74. doi: 10.1016/0165-5728(91)90073-G.
    1. Hauser SL, Doolittle TH, Lincoln R, Brown RH, Dinarello CA. Cytokine accumulations in CSF of multiple sclerosis patients: frequent detection of interleukin-1 and tumor necrosis factor but not interleukin-6. Neurology. 1990;40:1735–1739. doi: 10.1212/WNL.40.11.1735.
    1. Sharief MK, Hentges R. Association between tumor necrosis factor-alpha and disease progression in patients with multiple sclerosis. N Engl J Med. 1991;325:467–472. doi: 10.1056/NEJM199108153250704.
    1. Kahl KG, Kruse N, Toyka KV, Rieckmann P. Serial analysis of cytokine mRNA profiles in whole blood samples from patients with early multiple sclerosis. J Neurol Sci. 2002;200:53–55. doi: 10.1016/S0022-510X(02)00136-3.
    1. Link J. Interferon-gamma, interleukin-4 and transforming growth factor-beta mRNA expression in multiple sclerosis and myasthenia gravis. Acta Neurol Scand Suppl. 1994;158:1–58.
    1. Rieckmann P, Albrecht M, Kitze B, Weber T, Tumani H, Broocks A, Lüer W, Helwig A, Poser S. Tumor necrosis factor-alpha messenger RNA expression in patients with relapsing-remitting multiple sclerosis is associated with disease activity. Ann Neurol. 1995;37:82–88. doi: 10.1002/ana.410370115.
    1. Nicoletti F, Di Marco R, Patti F, Reggio E, Nicoletti A, Zaccone P, Stivala F, Meroni PL, Reggio A. Blood levels of transforming growth factor-beta 1 (TGF-beta1) are elevated in both relapsing remitting and chronic progressive multiple sclerosis (MS) patients and are further augmented by treatment with interferon-beta 1b (IFN-beta1b) Clin Exp Immunol. 1998;113:96–99. doi: 10.1046/j.1365-2249.1998.00604.x.
    1. Rollnik JD, Sindern E, Schweppe C, Malin JP. Biologically active TGF-beta 1 is increased in cerebrospinal fluid while it is reduced in serum in multiple sclerosis patients. Acta Neurol Scand. 1997;96:101–105. doi: 10.1111/j.1600-0404.1997.tb00248.x.
    1. Hollifield RD, Harbige LS, Pham-Dinh D, Sharief MK. Evidence for cytokine dysregulation in multiple sclerosis: peripheral blood mononuclear cell production of pro-inflammatory and anti-inflammatory cytokines during relapse and remission. Autoimmunity. 2003;36:133–141. doi: 10.1080/0891693031000089427.
    1. Hohnoki K, Inoue A, Koh CS. Elevated serum levels of IFN-gamma, IL-4 and TNF-alpha/unelevated serum levels of IL-10 in patients with demyelinating diseases during the acute stage. J Neuroimmunol. 1998;87:27–32. doi: 10.1016/S0165-5728(98)00053-8.
    1. Avolio C, Ruggieri M, Giuliani F, Liuzzi GM, Leante R, Riccio P, Livrea P, Trojano M. Serum MMP-2 and MMP-9 are elevated in different multiple sclerosis subtypes. J Neuroimmunol. 2003;136:46–53. doi: 10.1016/S0165-5728(03)00006-7.
    1. Fainardi E, Castellazzi M, Tamborino C, Trentini A, Manfrinato MC, Baldi E, Tola MR, Dallocchio F, Granieri E, Bellini T. Potential relevance of cerebrospinal fluid and serum levels and intrathecal synthesis of active matrix metalloproteinase-2 (MMP-2) as markers of disease remission in patients with multiple sclerosis. Mult Scler. 2009;15:547–554. doi: 10.1177/1352458509102372.
    1. Bar-Or A, Nuttall RK, Duddy M, Alter A, Kim HJ, Ifergan I, Pennington CJ, Bourgoin P, Edwards DR, Yong VW. Analyses of all matrix metalloproteinase members in leukocytes emphasize monocytes as major inflammatory mediators in multiple sclerosis. Brain. 2003;126:2738–2749. doi: 10.1093/brain/awg285.
    1. Lee MA, Palace J, Stabler G, Ford J, Gearing A, Miller K. Serum gelatinase B, TIMP-1 and TIMP-2 levels in multiple sclerosis. A longitudinal clinical and MRI study. Brain. 1999;122:191–197. doi: 10.1093/brain/122.2.191.
    1. Fainardi E, Castellazzi M, Bellini T, Manfrinato MC, Baldi E, Casetta I, Paolino E, Granieri E, Dallocchio F. Cerebrospinal fluid and serum levels and intrathecal production of active matrix metalloproteinase-9 (MMP-9) as markers of disease activity in patients with multiple sclerosis. Mult Scler. 2006;12:294–301. doi: 10.1191/135248506ms1274oa.
    1. Leppert D, Ford J, Stabler G, Grygar C, Lienert C, Huber S, Miller KM, Hauser SL, Kappos L. Matrix metalloproteinase-9 (gelatinase B) is selectively elevated in CSF during relapses and stable phases of multiple sclerosis. Brain. 1998;121:2327–2734. doi: 10.1093/brain/121.12.2327.
    1. Boz C, Ozmenoglu M, Velioglu S, Kilinc K, Orem A, Alioglu Z, Altunayoglu V. Matrix metalloproteinase-9 (MMP-9) and tissue inhibitor of matrix metalloproteinase (TIMP-1) in patients with relapsing-remitting multiple sclerosis treated with interferon beta. Clin Neurol Neurosurg. 2006;108:124–1248. doi: 10.1016/j.clineuro.2005.01.005.
    1. Bomprezzi R, Ringnér M, Kim S, Bittner ML, Khan J, Chen Y, Elkahloun A, Yu A, Bielekova B, Meltzer PS, Martin R, McFarland HF, Trent JM. Gene expression profile in multiple sclerosis patients and healthy controls: identifying pathways relevant to disease. Hum Mol Genet. 2003;12:2191–2199. doi: 10.1093/hmg/ddg221.
    1. Duan H, Luo Y, Hao H, Feng L, Zhang Y, Lu D, Xing S, Feng J, Yang D, Song L, Yan X. Soluble CD146 in cerebrospinal fluid of active multiple sclerosis. Neuroscience. 2013;235:16–26. doi: 10.1016/j.neuroscience.2013.01.020.

Source: PubMed

3
S'abonner