Potential therapeutic benefit of C1-esterase inhibitor in neuromyelitis optica evaluated in vitro and in an experimental rat model

Lukmanee Tradtrantip, Nithi Asavapanumas, Puay-Wah Phuan, A S Verkman, Lukmanee Tradtrantip, Nithi Asavapanumas, Puay-Wah Phuan, A S Verkman

Abstract

Neuromyelitis optica (NMO) is an autoimmune demyelinating disease of the central nervous system in which binding of anti-aquaporin-4 (AQP4) autoantibodies (NMO-IgG) to astrocytes causes complement-dependent cytotoxicity (CDC) and inflammation resulting in oligodendrocyte and neuronal injury. There is compelling evidence for a central role of complement in NMO pathogenesis. Here, we evaluated the potential of C1-esterase inhibitor (C1-inh) for complement-targeted therapy of NMO. C1-inh is an anti-inflammatory plasma protein with serine protease inhibition activity that has a broad range of biological activities on the contact (kallikrein), coagulation, fibrinolytic and complement systems. C1-inh is approved for therapy of hereditary angioedema (HAE) and has been studied in a small safety trial in acute NMO relapses (NCT 01759602). In vitro assays of NMO-IgG-dependent CDC showed C1-inh inhibition of human and rat complement, but with predicted minimal complement inhibition activity at a dose of 2000 units in humans. Inhibition of complement by C1-inh was potentiated by ∼10-fold by polysulfated macromolecules including heparin and dextran sulfate. In rats, intravenous C1-inh at a dose 30-fold greater than that approved to treat HAE inhibited serum complement activity by <5%, even when supplemented with heparin. Also, high-dose C1-inh did not reduce pathology in a rat model of NMO produced by intracerebral injection of NMO-IgG. Therefore, although C1r and C1s are targets of C1-inh, our in vitro data with human serum and in vivo data in rats suggest that the complement inhibition activity of C1-inh in serum is too low to confer clinical benefit in NMO.

Trial registration: ClinicalTrials.gov NCT01759602.

Conflict of interest statement

Competing Interests: This work described in this manuscript was funded, in part, by a sponsored research agreement from a commercial source (CSL Behring). This does not alter the authors' adherence to PLOS ONE policies on sharing data and materials.

Figures

Figure 1. Action of C1-inh on NMO-IgG-dependent…
Figure 1. Action of C1-inh on NMO-IgG-dependent cytotoxicity involve the classical complement pathway.
NMO-IgG binds to cell surface AQP4 on astrocytes, which activates the classical complement pathway by C1q binding to the Fc region of NMO-IgG leading to formation of the membrane attack complex (MAC) (left). C1-inh binds to and inactivates C1r and C1s, preventing C1q action on C4 and downstream complement activation.
Figure 2. C1-inh inhibits complement-dependent cytotoxicity produced…
Figure 2. C1-inh inhibits complement-dependent cytotoxicity produced by NMO-IgG in AQP4-expressing cell cultures.
A. CDC assay showing 1 h incubation of NMO-IgG, human complement (HC) and C1-INH to CHO cells expressing human M23-AQP4, followed by assay of cytotoxicity with AlamarBlue. B. Percentage cytotoxicity as a function of C1-inh concentration in cells incubated with human complement (2%) and recombinant NMO-IgG rAb-53 (1 µg/ml) (left) or individual or pooled human NMO sera (100 µg/ml) (right). C. Study as in B in cells incubated with rAb-53 (0.5 µg/ml) and rat serum (2%). Data are mean ± S.E. for 4 measurements per condition.
Figure 3. C1-inh inhibition of NMO-IgG-dependent cytotoxicity…
Figure 3. C1-inh inhibition of NMO-IgG-dependent cytotoxicity depends on complement and NMO-IgG concentration.
A. Percentage cytotoxicity as a function of C1-inh concentration in cells incubated with 1 µg/ml rAb-53 and indicated concentration of human complement. Data are mean ± S.E. for 4 measurements per condition. Inset shows percentage inhibition of cytotoxicity as a function of time after pre-incubation of 2 µM C1-inh and 2% human complement prior to addition AQP4-expressing CHO-cell that were pre-incubated with 1 µg/ml rAb-53. B. Study as in A but with 0.5 µg/ml rAb-53.
Figure 4. Polysulfated macromolecules potentiate C1-inh inhibition…
Figure 4. Polysulfated macromolecules potentiate C1-inh inhibition of NMO-IgG-dependent CDC.
A. Percentage cytotoxicity as a function of C1-inh concentration in cells incubated with human complement (2%) and rAb-53 (1 µg/ml) in the absence or presence of 2 µM heparin, 0.5 µM dextran sulfate or 0.02 µM fucoidan (mean ± S.E., n = 4). Insets show chemical structures. B. Percentage cytotoxicity as a function of concentration of polysulfated macromolecules in cells incubated with human complement (2%) and rAb-53 (1 µg/ml) in the absence or presence of 1 µM C1-inh.
Figure 5. Intravenous administration of high-dose C1-inh…
Figure 5. Intravenous administration of high-dose C1-inh in rats does not inhibit complement activity.
A. C1-inh was administered to rats by tail vein injection, and serum samples obtained at specified times were bioassayed for NMO-IgG-dependent CDC by incubation of AQP4-expressing CHO-cells with 2% rat serum with 1 µg/ml rAb-53 on percentage cytotoxicity produced by rat serum at different times after administration of 600 units/kg C1-inh (or vehicle control) (left), and 1.5 mg/kg heparin, without or with 600 units/kg C1-inh (right) (mean ± S.E., 4 rats per condition).
Figure 6. High-dose C1-inh does not reduce…
Figure 6. High-dose C1-inh does not reduce brain pathology in a rat model of NMO produced by intracerebral injection NMO-IgG.
AQP4, GFAP and MBP immunofluorescence at 2 days after injection of 10 µg rAb-53. C1-inh (300 units/kg) (or saline control) was administered intravenously just before and 1 day after intracerebral injection of NMO-IgG. The needle tract is shown in yellow line, the white line demarcates lesion areas with loss of AQP4 and GFAP immunofluorescence, and the white dashed line demarcates the penumbra areas with reduced AQP4 but normal GFAP immunofluorescence (left). Summary of lesion areas showing data for individual rats (mean ± S.E., n = 4). Differences not significant.

References

    1. Wingerchuk DM, Lennon VA, Lucchinetti CF, Pittock SJ, Weinshenker BG (2007) The spectrum of neuromyelitis optica. Lancet Neurol 6: 805–815.
    1. Jacob A, McKeon A, Nakashima I, Sato DK, Elsone L, et al. (2013) Current concept of neuromyelitis optica (NMO) and NMO spectrum disorders. J Neurol Neurosurg Psychiatry 84: 922–930.
    1. Jarius S, Paul F, Franciotta D, Waters P, Zipp F, et al. (2008) Mechanism of disease: aquaporin-4 antibodies in neuromyelitis optica. Nat Clin Pract Neurol 4: 202–214.
    1. Lennon VA, Kryzer TJ, Pittock SJ, Verkman AS, Hinson SR (2005) IgG marker of optic-spinal multiple sclerosis binds to the aquaporin-4 water channel. J Exp Med 202: 473–477.
    1. Papadopoulos MC, Verkman AS (2012) Aquaporin water channels in the nervous system. Nat Rev Neurosci 14: 265–277.
    1. Papadopoulos MC, Verkman AS (2013) Aquaporin 4 and neuromyelitis optica. Lancet Neurol 11: 535–544.
    1. Kira J (2011) Autoimmunity in neuromyelitis optica and opticospinal multiple sclerosis: astrocytopathy as a common denominator in demyelinating disorders. J Neurol Sci 311: 69–77.
    1. Collongues N, de Seze J (2011) Current and future treatment approaches for neuromyelitis optica. Ther Adv Neurol Disord 4: 111–121.
    1. Cree B (2008) Neuromyelitis optica: diagnosis, pathogenesis, and treatment. Curr Neurol Neurosci Rep 8: 427–433.
    1. Kim SH, Kim W, Huh SY, Lee KY, Jung IJ, et al. (2013) Clinical efficacy of plasmapheresis in patients with neuromyelitis optica spectrum disorder and effects on circulating anti-aquaporin-4 antibody level. J Clin Neurol 9: 36–42.
    1. Lucchinetti CF, Mandler RN, McGavern D, Bruck W, Gleich G, et al. (2002) A role for humoral mechanisms in the pathogenesis of Devic's neuromyelitis optica. Brain 125: 1450–1461.
    1. Misu T, Fujihara K, Kakita A, Konno H, Nakamura M, et al. (2007) Loss of aquaporin 4 in lesions of neuromyelitis optica: distinction from multiple sclerosis. Brain 130: 1224–1234.
    1. Roemer SF, Parisi JE, Lennon VA, Benarroch EE, Lassmann H, et al. (2007) Pattern-specific loss of aquaporin-4 immunoreactivity distinguishes neuromyelitis optica from multiple sclerosis. Brain 130: 1194–1205.
    1. Sabater L, Giralt A, Boronat A, Hankiewicz K, Blanco Y, et al. (2009) Cytotoxic effect of neuromyelitis optica antibody (NMO-IgG) to astrocytes: an in vitro study. J Neuroimmunol 215: 31–35.
    1. Hinson SR, Pittock SJ, Lucchinetti CF, Roemer SF, Fryer JP, et al. (2007) Pathogenic potential of IgG binding to water channel extracellular domain in neuromyelitis optica. Neurology 69: 2221–2231.
    1. Hinson SR, McKeon A, Fryer JP, Apiwattanakul M, Lennon VA, et al. (2009) Prediction of neuromyelitis optica attack severity by quantitation of complement-mediated injury to aquaporin-4-expressing cells. Arch Neurol 66: 1164–1167.
    1. Bennett JL, Lam C, Kalluri SR, Saikali P, Bautista K, et al. (2009) Intrathecal pathogenic anti-aquaporin-4 antibodies in early neuromyelitis optica. Ann Neurol 66: 617–629.
    1. Zhang H, Bennett JL, Verkman AS (2011) Ex vivo spinal cord slice model of neuromyelitis optica reveals novel immunopathogenic mechanisms. Ann Neurol 70: 943–954.
    1. Saadoun S, Waters P, Bell BA, Vincent A, Verkman AS, et al. (2010) Intra-cerebral injection of neuromyelitis optica immunoglobulin G and human complement produces neuromyelitis optica lesions in mice. Brain 133: 349–361.
    1. Asavapanumas N, Ratelade J, Verkman AS (2014) Unique neuromyelitis optica pathology produced in naòve rats by intracerebral administration of NMO-IgG. Acta Neuropathol 127: 539–551.
    1. Pittock SJ, Lennon VA, Mckeon A, Mandrekar J, Weinshenker BG, et al. (2013) Eculizumab in AQP4-IgG-positive relapsing neuromyelitis optica spectrum disorders: an open-label pilot study. Lancet Neurol 12: 554–562.
    1. Parker C (2009) Eculizumab for paroxysmal nocturnal haemoglobinuria. Lancet 373: 759–767.
    1. Phuan PW, Zhang H, Asavapanumas N, Leviten M, Rosenthal A, et al. (2013) C1q-targeted monoclonal antibody prevents complement-dependent cytotoxicity and neuropathology in in vitro and mouse models of neuromyelitis optica. Acta Neuropathol 125: 829–840.
    1. Caliezi C, Wuillemin WA, Zeerleder S, Redondo M, Eisele B, et al. (2000) C1-esterase inhibitor: an anti-inflammatory agent and its potential use in the treatment of disease other than hereditary angioedema. Pharmacol Rev 52: 91–112.
    1. Karnaukhova E (2013) C1-esterase inhibitor: biological activities and therapeutic applications. J Hematol Thromb Dis 1: 3 Available: .
    1. Levy M, Mealy MA (2014) Purified human C1-esterase inhibitor is safe in acute relapses of neuromyelitis optica. Neurol Neuroimmunol Neutoinflamm 1 doi
    1. Phuan PW, Ratelade J, Rossi A, Tradtrantip L, Verkman AS (2012) Complement-dependent cytotoxicity in neuromyelitis optica requires aquaporin-4 protein assembly in orthogonal arrays. J Biol Chem 287: 13829–13839.
    1. Gettins PG (2002) Serpin structure, mechanism, and function. Chem Rev 102: 4751–4804.
    1. Ziccardi RJ, Cooper NR (1979) Active disassembly of the first complement component, C1, by C1 inactivator. J Immunol 123: 788–792.
    1. Weiler JM, Edens RE, Linhardt RJ, Kapelanski DP (1992) Heparin and modified heparin inhibit complement activation in vivo. J Immunol 148: 3210–3215.
    1. Garlatti V, Chouquet A, Lunardi T, Vives R, Paidassi H, et al. (2010) Cutting edge: C1q binds deoxyribose and heparin sulfate through neighboring sites of its recognition domain. J Immunol 185: 808–812.
    1. Hughes-Jones NC, Gardner B (1978) The reaction between the complement subcomponent C1q, IgG complexes and polyionic molecules. Immunol 34: 459–463.
    1. Wuillemin WA, Velthuis HT, Lubbers YTP, de Ruig CP, Eldering E, et al. (1997) Potentiation of C1 inhibitor by glycosaminoglycans. J Immunol 159: 1953–1960.
    1. Beinrohr L, Harmat V, Dobo J, Lorincz Z, Gal P, et al. (2007) C1 inhibitor serpin domain structure reveals the likely mechanism of heparin potentiation and conformational disease. J Biol Chem 282: 21100–21109.
    1. Ratelade J, Smith AJ, Verkman AS (2014) Human immunoglobulin G reduces the pathogenicity of aquaporin-4 autoantibodies in neuromyelitis optica. Exper Neurol 255: 145–153.
    1. Crane JM, Lam C, Rossi A, Gupta T, Bennett JL, et al. (2011) Binding affinity and specificity of neuromyelitis optica autoantibodies to aquaporin-4 M1/M23 isoforms and orthogonal arrays. J Biol Chem 286: 16516–16524.
    1. Ayzenberg I, Kleiter I, Schruder A, Hellwig K, Chan A, et al. (2013) Interleukin 6 receptor blockade in patients with neuromyelitis optica nonresponsive to anti-CD20 therapy. JAMA Neurol 70: 394–397.
    1. Tradtrantip L, Zhang H, Saadoun S, Phuan PW, Lam C, et al. (2012) Anti-aquaporin-4 monoclonal antibody blocker therapy for neuromyelitis optica. Ann Neurol 71: 314–322.
    1. Saadoun S, Waters P, MacDonald C, Bell BA, Vincent A, et al. (2012) Neutrophil protease inhibition reduces neuromyelitis optica-immunoglobulin G-induced damage in mouse brain. Ann Neurol 71: 323–33.
    1. Zhang H, Verkman AS (2013) Eosinophil pathogenicity mechanisms and therapeutics in neuromyelitis optica. J Clin Invest 123: 2306–2316.
    1. Kerr FK, Thomas AR, Wijeyewickrema LC, Whisstock JC, Boyd SE, et al. (2008) Elucidation of the substrate specificity of the MASP-2 protease of the lectin complement pathway and identification of the enzyme as a major physiological target of the serpin, C1-inhibitor. Mol Immunol 45: 670–677.
    1. Prematta MJ, Prematta T, Craig TJ (2008) Treatment of hereditary angioedema with plasma-derived C1 inhibitor. Ther Clin Risk Manag 4: 975–982.
    1. Cugno M, Zanichelli A, Foieni F, Caccia S, Cicardi M (2009) C1-inhibitor deficiency and angioedema: molecular mechanisms and clinical progress. Trend Mol Med 15: 69–78.
    1. Ratelade J, Verkman AS (2014) Inhibitors of the classical complement pathway in mouse serum limit the utility of mice as experimental models of neuromyelitis optica. Mol Immunol 62: 104–113.
    1. Cicardi M, Bergamaschini L, Cugno M, Beretta A, Zingale LC, et al. (1998) Pathogenetic and clinical aspects of C1 inhibitor deficiency. Immunobiol 199: 366–376.
    1. Kunschak M, Engl W, Maritsch F, Rosen FS, Eder G, et al. (1998) A randomized, controlled trial to study the efficacy and safety of C1 inhibitor concentrate in treating hereditary angioedema. Transfusion 38: 540–549.
    1. Wouters D, Stephan F, Strengers P, Hass MD, Brouwer C, et al. (2013) C1-esterase inhibitor concentrate rescues erythrocyte from complement-mediated destruction in autoimmune hemolytic anemia. Blood 121: 1242–1244.
    1. Bianchino AC, Poon PH, Schumaker VN (1988) A mechanism for the spontaneous activation of the first component of complement, C1, and its regulation by C1-inhibitor. J Immunol 141: 3930–3936.
    1. Nielsen EW, Waage C, Fure H, Brekke OL, Sfyroera G (2007) Effect of supraphysiologic levels of C1-inhibitor on the classical, lectin and alternative pathways of complement. Mol Imunol 44: 1819–1826.
    1. Tenner AJ, Frank MM (1986) Activator-bound C1 is less susceptible to inactivation by C1 inhibition than is fluid-phase C1. J Immunol 137: 625–630.

Source: PubMed

3
Abonnieren