Skip to main content
Top
Published in: Arthritis Research & Therapy 1/2018

Open Access 01-12-2018 | Research article

Highly selective inhibition of Bruton’s tyrosine kinase attenuates skin and brain disease in murine lupus

Authors: Samantha A. Chalmers, Jing Wen, Jessica Doerner, Ariel Stock, Carla M. Cuda, Hadijat M. Makinde, Harris Perlman, Todd Bosanac, Deborah Webb, Gerald Nabozny, Jay S. Fine, Elliott Klein, Meera Ramanujam, Chaim Putterman

Published in: Arthritis Research & Therapy | Issue 1/2018

Login to get access

Abstract

Background

Systemic lupus erythematosus (SLE) is a systemic autoimmune disease that affects different end organs, including skin and brain. We and others have previously shown the importance of macrophages in the pathogenesis of cutaneous and neuropsychiatric lupus. Additionally, autoantibodies produced by autoreactive B cells are thought to play a role in both the skin and central nervous system pathologies associated with SLE.

Methods

We used a novel inhibitor of Bruton’s tyrosine kinase (BTK), BI-BTK-1, to target both macrophage and B cell function in the MRL-lpr/lpr murine model of SLE, and examined the effect of treatment on skin and brain disease.

Results

We found that treatment with BI-BTK-1 significantly attenuated the lupus associated cutaneous and neuropsychiatric disease phenotypes in MRL/lpr mice. Specifically, BI-BTK-1 treated mice had fewer macroscopic and microscopic skin lesions, reduced cutaneous cellular infiltration, and diminished inflammatory cytokine expression compared to control mice. BTK inhibition also significantly improved cognitive function, and decreased accumulation of T cells, B cells, and macrophages within the central nervous system, specifically the choroid plexus.

Conclusions

Directed therapies may improve the response rate in lupus-driven target organ involvement, and decrease the dangerous side effects associated with global immunosuppression. Overall, our results suggest that inhibition of BTK may be a promising therapeutic option for cutaneous and neuropsychiatric disease associated with SLE.
Appendix
Available only for authorised users
Literature
1.
go back to reference Crispín JC, Liossis S-NC, Kis-Toth K, Lieberman LA, Kyttaris VC, Juang Y-T, Tsokos GC. Pathogenesis of human systemic lupus erythematosus: recent advances. Trends Mol Med. 2010;16(2):47–57.CrossRefPubMedPubMedCentral Crispín JC, Liossis S-NC, Kis-Toth K, Lieberman LA, Kyttaris VC, Juang Y-T, Tsokos GC. Pathogenesis of human systemic lupus erythematosus: recent advances. Trends Mol Med. 2010;16(2):47–57.CrossRefPubMedPubMedCentral
2.
go back to reference Deng GM, Tsokos GC. Pathogenesis and targeted treatment of skin injury in SLE. Nat Rev Rheumatol. 2015;11(11):663–9.CrossRefPubMed Deng GM, Tsokos GC. Pathogenesis and targeted treatment of skin injury in SLE. Nat Rev Rheumatol. 2015;11(11):663–9.CrossRefPubMed
3.
go back to reference Meszaros ZS, Perl A, Faraone SV. Psychiatric symptoms in systemic lupus erythematosus: a systematic review. J Clin Psychiatry. 2012;73(7):993–1001.CrossRefPubMed Meszaros ZS, Perl A, Faraone SV. Psychiatric symptoms in systemic lupus erythematosus: a systematic review. J Clin Psychiatry. 2012;73(7):993–1001.CrossRefPubMed
4.
go back to reference Xiong W, Lahita RG. Pragmatic approaches to therapy for systemic lupus erythematosus. Nat Rev Rheumatol. 2014;10(2):97–107.CrossRefPubMed Xiong W, Lahita RG. Pragmatic approaches to therapy for systemic lupus erythematosus. Nat Rev Rheumatol. 2014;10(2):97–107.CrossRefPubMed
6.
go back to reference Govoni M, Bortoluzzi A, Padovan M, Silvagni E, Borrelli M, Donelli F, Ceruti S, Trotta F. The diagnosis and clinical management of the neuropsychiatric manifestations of lupus. J Autoimmun. 2016;74:41–72.CrossRefPubMed Govoni M, Bortoluzzi A, Padovan M, Silvagni E, Borrelli M, Donelli F, Ceruti S, Trotta F. The diagnosis and clinical management of the neuropsychiatric manifestations of lupus. J Autoimmun. 2016;74:41–72.CrossRefPubMed
7.
go back to reference Chalmers S, Doerner J, Wen J, Putterman C. Macrophage depletion attenuates skin and kidney disease in lupus mice (BA7P.143). J Immunol. 2015;194(1 Supplement):115.113. Chalmers S, Doerner J, Wen J, Putterman C. Macrophage depletion attenuates skin and kidney disease in lupus mice (BA7P.143). J Immunol. 2015;194(1 Supplement):115.113.
8.
go back to reference Chalmers SA, Wen J, Shum J, Doerner J, Herlitz L, Putterman C. CSF-1R inhibition attenuates renal and neuropsychiatric disease in murine lupus. Clin Immunol. 2016;185:100–108 Chalmers SA, Wen J, Shum J, Doerner J, Herlitz L, Putterman C. CSF-1R inhibition attenuates renal and neuropsychiatric disease in murine lupus. Clin Immunol. 2016;185:100–108
9.
go back to reference Doerner J, Chalmers SA, Friedman A, Putterman C. Fn14 deficiency protects lupus-prone mice from histological lupus erythematosus-like skin inflammation induced by ultraviolet light. Exp Dermatol. 2016;25(12):969–76.CrossRefPubMedPubMedCentral Doerner J, Chalmers SA, Friedman A, Putterman C. Fn14 deficiency protects lupus-prone mice from histological lupus erythematosus-like skin inflammation induced by ultraviolet light. Exp Dermatol. 2016;25(12):969–76.CrossRefPubMedPubMedCentral
10.
go back to reference Menke J, Hsu MY, Byrne KT, Lucas JA, Rabacal WA, Croker BP, Zong XH, Stanley ER, Kelley VR. Sunlight triggers cutaneous lupus through a CSF-1-dependent mechanism in MRL-Fas(lpr) mice. J Immunol. 2008;181(10):7367–79.CrossRefPubMedPubMedCentral Menke J, Hsu MY, Byrne KT, Lucas JA, Rabacal WA, Croker BP, Zong XH, Stanley ER, Kelley VR. Sunlight triggers cutaneous lupus through a CSF-1-dependent mechanism in MRL-Fas(lpr) mice. J Immunol. 2008;181(10):7367–79.CrossRefPubMedPubMedCentral
11.
12.
go back to reference Jongstra-Bilen J, Puig Cano A, Hasija M, Xiao H, Smith CIE, Cybulsky MI. Dual functions of Bruton’s tyrosine kinase and Tec kinase during Fcγ receptor-induced signaling and phagocytosis. J Immunol. 2008;181(1):288–98.CrossRefPubMed Jongstra-Bilen J, Puig Cano A, Hasija M, Xiao H, Smith CIE, Cybulsky MI. Dual functions of Bruton’s tyrosine kinase and Tec kinase during Fcγ receptor-induced signaling and phagocytosis. J Immunol. 2008;181(1):288–98.CrossRefPubMed
13.
go back to reference Ni Gabhann J, Hams E, Smith S, Wynne C, Byrne JC, Brennan K, Spence S, Kissenpfennig A, Johnston JA, Fallon PG, et al. Btk regulates macrophage polarization in response to lipopolysaccharide. PLoS One. 2014;9(1):e85834.CrossRefPubMedPubMedCentral Ni Gabhann J, Hams E, Smith S, Wynne C, Byrne JC, Brennan K, Spence S, Kissenpfennig A, Johnston JA, Fallon PG, et al. Btk regulates macrophage polarization in response to lipopolysaccharide. PLoS One. 2014;9(1):e85834.CrossRefPubMedPubMedCentral
14.
15.
go back to reference Chalmers SA, Doerner J, Bosanac T, Khalil S, Smith D, Harcken C, Dimock J, Der E, Herlitz L, Webb D, et al. Therapeutic blockade of immune complex-mediated glomerulonephritis by highly selective inhibition of Bruton’s tyrosine kinase. Sci Rep. 2016;6:26164.CrossRefPubMedPubMedCentral Chalmers SA, Doerner J, Bosanac T, Khalil S, Smith D, Harcken C, Dimock J, Der E, Herlitz L, Webb D, et al. Therapeutic blockade of immune complex-mediated glomerulonephritis by highly selective inhibition of Bruton’s tyrosine kinase. Sci Rep. 2016;6:26164.CrossRefPubMedPubMedCentral
16.
go back to reference Sakic B. The MRL, model: an invaluable tool in studies of autoimmunity-brain interactions. Methods Mol Biol. 2012;934:277–99.CrossRefPubMed Sakic B. The MRL, model: an invaluable tool in studies of autoimmunity-brain interactions. Methods Mol Biol. 2012;934:277–99.CrossRefPubMed
17.
go back to reference Gulinello M, Putterman C. The MRL/lpr mouse strain as a model for neuropsychiatric systemic lupus erythematosus. J Biomed Biotechnol. 2011;2011:207504.CrossRefPubMedPubMedCentral Gulinello M, Putterman C. The MRL/lpr mouse strain as a model for neuropsychiatric systemic lupus erythematosus. J Biomed Biotechnol. 2011;2011:207504.CrossRefPubMedPubMedCentral
18.
go back to reference Wen J, Xia Y, Stock A, Michaelson JS, Burkly LC, Gulinello M. Neuropsychiatric disease in murine lupus is dependent on the TWEAK/Fn14 pathway. J Autoimmun. 2013;43:44–54.CrossRefPubMedPubMedCentral Wen J, Xia Y, Stock A, Michaelson JS, Burkly LC, Gulinello M. Neuropsychiatric disease in murine lupus is dependent on the TWEAK/Fn14 pathway. J Autoimmun. 2013;43:44–54.CrossRefPubMedPubMedCentral
19.
go back to reference Lin JH, Dutz JP, Sontheimer RD, Werth VP. Pathophysiology of cutaneous lupus erythematosus. Clin Rev Allergy Immunol. 2007;33(1-2):85–106.CrossRefPubMed Lin JH, Dutz JP, Sontheimer RD, Werth VP. Pathophysiology of cutaneous lupus erythematosus. Clin Rev Allergy Immunol. 2007;33(1-2):85–106.CrossRefPubMed
20.
go back to reference Stock AD, Wen J, Doerner J, Herlitz LC, Gulinello M, Putterman C. Neuropsychiatric systemic lupus erythematosus persists despite attenuation of systemic disease in MRL/lpr mice. J Neuroinflammation. 2015;12:205.CrossRefPubMedPubMedCentral Stock AD, Wen J, Doerner J, Herlitz LC, Gulinello M, Putterman C. Neuropsychiatric systemic lupus erythematosus persists despite attenuation of systemic disease in MRL/lpr mice. J Neuroinflammation. 2015;12:205.CrossRefPubMedPubMedCentral
21.
go back to reference Sciascia S, Bertolaccini ML, Roccatello D, Khamashta MA, Sanna G. Autoantibodies involved in neuropsychiatric manifestations associated with systemic lupus erythematosus: a systematic review. J Neurol. 2014;261(9):1706–14.CrossRefPubMed Sciascia S, Bertolaccini ML, Roccatello D, Khamashta MA, Sanna G. Autoantibodies involved in neuropsychiatric manifestations associated with systemic lupus erythematosus: a systematic review. J Neurol. 2014;261(9):1706–14.CrossRefPubMed
22.
23.
go back to reference Hutcheson J, Vanarsa K, Bashmakov A, Grewal S, Sajitharan D, Chang BY, Buggy JJ, Zhou XJ, Du Y, Satterthwaite AB, et al. Modulating proximal cell signaling by targeting Btk ameliorates humoral autoimmunity and end-organ disease in murine lupus. Arthritis Res Ther. 2012;14(6):R243.CrossRefPubMedPubMedCentral Hutcheson J, Vanarsa K, Bashmakov A, Grewal S, Sajitharan D, Chang BY, Buggy JJ, Zhou XJ, Du Y, Satterthwaite AB, et al. Modulating proximal cell signaling by targeting Btk ameliorates humoral autoimmunity and end-organ disease in murine lupus. Arthritis Res Ther. 2012;14(6):R243.CrossRefPubMedPubMedCentral
24.
go back to reference Mina-Osorio P, LaStant J, Keirstead N, Whittard T, Ayala J, Stefanova S, Garrido R, Dimaano N, Hilton H, Giron M, et al. Suppression of glomerulonephritis in lupus-prone NZB x NZW mice by RN486, a selective inhibitor of Bruton’s tyrosine kinase. Arthritis Rheum. 2013;65(9):2380–91.CrossRefPubMed Mina-Osorio P, LaStant J, Keirstead N, Whittard T, Ayala J, Stefanova S, Garrido R, Dimaano N, Hilton H, Giron M, et al. Suppression of glomerulonephritis in lupus-prone NZB x NZW mice by RN486, a selective inhibitor of Bruton’s tyrosine kinase. Arthritis Rheum. 2013;65(9):2380–91.CrossRefPubMed
25.
go back to reference Rankin AL, Seth N, Keegan S, Andreyeva T, Cook TA, Edmonds J, Mathialagan N, Benson MJ, Syed J, Zhan Y, et al. Selective inhibition of BTK prevents murine lupus and antibody-mediated glomerulonephritis. J Immunol. 2013;191(9):4540–50.CrossRefPubMed Rankin AL, Seth N, Keegan S, Andreyeva T, Cook TA, Edmonds J, Mathialagan N, Benson MJ, Syed J, Zhan Y, et al. Selective inhibition of BTK prevents murine lupus and antibody-mediated glomerulonephritis. J Immunol. 2013;191(9):4540–50.CrossRefPubMed
26.
go back to reference Bender AT, Pereira A, Fu K, Samy E, Wu Y, Liu-Bujalski L, Caldwell R, Chen YY, Tian H, Morandi F, et al. Btk inhibition treats TLR7/IFN driven murine lupus. Clin Immunol. 2016;164:65–77.CrossRefPubMed Bender AT, Pereira A, Fu K, Samy E, Wu Y, Liu-Bujalski L, Caldwell R, Chen YY, Tian H, Morandi F, et al. Btk inhibition treats TLR7/IFN driven murine lupus. Clin Immunol. 2016;164:65–77.CrossRefPubMed
27.
go back to reference Byrd JC, Furman RR, Coutre SE, Flinn IW, Burger JA, Blum KA, Grant B, Sharman JP, Coleman M, Wierda WG, et al. Targeting BTK with ibrutinib in relapsed chronic lymphocytic leukemia. N Engl J Med. 2013;369(1):32–42.CrossRefPubMedPubMedCentral Byrd JC, Furman RR, Coutre SE, Flinn IW, Burger JA, Blum KA, Grant B, Sharman JP, Coleman M, Wierda WG, et al. Targeting BTK with ibrutinib in relapsed chronic lymphocytic leukemia. N Engl J Med. 2013;369(1):32–42.CrossRefPubMedPubMedCentral
28.
go back to reference Sakuma C, Sato M, Takenouchi T, Chiba J, Kitani H. Critical Roles of the WASP N-terminal domain and Btk in LPS-induced inflammatory response in macrophages. PLoS One. 2012;7(1):e30351.CrossRefPubMedPubMedCentral Sakuma C, Sato M, Takenouchi T, Chiba J, Kitani H. Critical Roles of the WASP N-terminal domain and Btk in LPS-induced inflammatory response in macrophages. PLoS One. 2012;7(1):e30351.CrossRefPubMedPubMedCentral
29.
go back to reference Amoras AL, Kanegane H, Miyawaki T, Vilela MM. Defective Fc-, CR1- and CR3-mediated monocyte phagocytosis and chemotaxis in common variable immunodeficiency and X-linked agammaglobulinemia patients. J Investig Allergol Clin Immunol. 2003;13(3):181–8.PubMed Amoras AL, Kanegane H, Miyawaki T, Vilela MM. Defective Fc-, CR1- and CR3-mediated monocyte phagocytosis and chemotaxis in common variable immunodeficiency and X-linked agammaglobulinemia patients. J Investig Allergol Clin Immunol. 2003;13(3):181–8.PubMed
30.
go back to reference Honigberg LA, Smith AM, Sirisawad M, Verner E, Loury D, Chang B, Li S, Pan Z, Thamm DH, Miller RA, et al. The Bruton tyrosine kinase inhibitor PCI-32765 blocks B-cell activation and is efficacious in models of autoimmune disease and B-cell malignancy. Proc Natl Acad Sci U S A. 2010;107(29):13075–80.CrossRefPubMedPubMedCentral Honigberg LA, Smith AM, Sirisawad M, Verner E, Loury D, Chang B, Li S, Pan Z, Thamm DH, Miller RA, et al. The Bruton tyrosine kinase inhibitor PCI-32765 blocks B-cell activation and is efficacious in models of autoimmune disease and B-cell malignancy. Proc Natl Acad Sci U S A. 2010;107(29):13075–80.CrossRefPubMedPubMedCentral
31.
go back to reference Katewa A, Wang Y, Hackney JA, Huang T, Suto E, Ramamoorthi N, Austin CD, Bremer M, Chen JZ, Crawford JJ, et al. Btk-specific inhibition blocks pathogenic plasma cell signatures and myeloid cell-associated damage in IFNalpha-driven lupus nephritis. JCI insight. 2017;2(7):e90111.CrossRefPubMedPubMedCentral Katewa A, Wang Y, Hackney JA, Huang T, Suto E, Ramamoorthi N, Austin CD, Bremer M, Chen JZ, Crawford JJ, et al. Btk-specific inhibition blocks pathogenic plasma cell signatures and myeloid cell-associated damage in IFNalpha-driven lupus nephritis. JCI insight. 2017;2(7):e90111.CrossRefPubMedPubMedCentral
32.
33.
go back to reference Volmering S, Block H, Boras M, Lowell Clifford A, Zarbock A. The neutrophil Btk signalosome regulates integrin activation during sterile inflammation. Immunity. 2016;44:73–87.CrossRefPubMedPubMedCentral Volmering S, Block H, Boras M, Lowell Clifford A, Zarbock A. The neutrophil Btk signalosome regulates integrin activation during sterile inflammation. Immunity. 2016;44:73–87.CrossRefPubMedPubMedCentral
34.
go back to reference Ito M, Shichita T, Okada M, Komine R, Noguchi Y, Yoshimura A, et al. Bruton’s tyrosine kinase is essential for NLRP3 inflammasome activation and contributes to ischaemic brain injury. Nat Commun. 2015;6:7360.CrossRefPubMedPubMedCentral Ito M, Shichita T, Okada M, Komine R, Noguchi Y, Yoshimura A, et al. Bruton’s tyrosine kinase is essential for NLRP3 inflammasome activation and contributes to ischaemic brain injury. Nat Commun. 2015;6:7360.CrossRefPubMedPubMedCentral
35.
go back to reference Broides A, Hadad N, Levy J, Levy R. The effects of Bruton tyrosine kinase inhibition on chemotaxis and superoxide generation in human neutrophils. J Clin Immunol. 2014;34:555–60.CrossRefPubMed Broides A, Hadad N, Levy J, Levy R. The effects of Bruton tyrosine kinase inhibition on chemotaxis and superoxide generation in human neutrophils. J Clin Immunol. 2014;34:555–60.CrossRefPubMed
Metadata
Title
Highly selective inhibition of Bruton’s tyrosine kinase attenuates skin and brain disease in murine lupus
Authors
Samantha A. Chalmers
Jing Wen
Jessica Doerner
Ariel Stock
Carla M. Cuda
Hadijat M. Makinde
Harris Perlman
Todd Bosanac
Deborah Webb
Gerald Nabozny
Jay S. Fine
Elliott Klein
Meera Ramanujam
Chaim Putterman
Publication date
01-12-2018
Publisher
BioMed Central
Published in
Arthritis Research & Therapy / Issue 1/2018
Electronic ISSN: 1478-6362
DOI
https://doi.org/10.1186/s13075-017-1500-0

Other articles of this Issue 1/2018

Arthritis Research & Therapy 1/2018 Go to the issue