Abstract

BackgroundExpression of chemokine CCL2 in the normal central nervous system (CNS) is nearly undetectable, but is significantly upregulated and drives neuroinflammation during experimental autoimmune encephalomyelitis (EAE), an animal model of multiple sclerosis which is considered a contributing factor in the human disease. As astrocytes and brain microvascular endothelial cells (BMEC) forming the blood–brain barrier (BBB) are sources of CCL2 in EAE and other neuroinflammatory conditions, it is unclear if one or both CCL2 pools are critical to disease and by what mechanism(s).MethodsMice with selective CCL2 gene knockout (KO) in astrocytes (Astro KO) or endothelial cells (Endo KO) were used to evaluate the respective contributions of these sources to neuroinflammation, i.e., clinical disease progression, BBB damage, and parenchymal leukocyte invasion in a myelin oligodendrocyte glycoprotein peptide (MOG35-55)-induced EAE model. High-resolution 3-dimensional (3D) immunofluorescence confocal microscopy and colloidal gold immuno-electron microscopy were employed to confirm sites of CCL2 expression, and 3D immunofluorescence confocal microscopy utilized to assess inflammatory responses along the CNS microvasculature.ResultsCell-selective loss of CCL2 immunoreactivity was demonstrated in the respective KO mice. Compared to wild-type (WT) mice, Astro KO mice showed reduced EAE severity but similar onset, while Endo KO mice displayed near normal severity but significantly delayed onset. Neither of the KO mice showed deficits in T cell proliferation, or IL-17 and IFN-γ production, following MOG35-55 exposure in vitro, or altered MOG-major histocompatibility complex class II tetramer binding. 3D confocal imaging further revealed distinct actions of the two CCL2 pools in the CNS. Astro KOs lacked the CNS leukocyte penetration and disrupted immunostaining of CLN-5 at the BBB seen during early EAE in WT mice, while Endo KOs uniquely displayed leukocytes stalled in the microvascular lumen.ConclusionsThese results point to astrocyte and endothelial pools of CCL2 each regulating different stages of neuroinflammation in EAE, and carry implications for drug delivery in neuroinflammatory disease.

Highlights

  • Expression of chemokine Chemokine ligand 2 (CCL2) in the normal central nervous system (CNS) is nearly undetectable, but is significantly upregulated and drives neuroinflammation during experimental autoimmune encephalomyelitis (EAE), an animal model of multiple sclerosis which is considered a contributing factor in the human disease

  • Elevated CNS expression of CCL2 has been a consistent observation among the different paradigms of experimental autoimmune encephalomyelitis (EAE) [6,7,8], a CNS demyelinating inflammatory disease that serves as a model for multiple sclerosis

  • This chemokine’s singular importance in driving EAE was demonstrated by global CCL2 knockout (KO) mice (CCL2−/−), which showed diminished severity and delay in onset of disease in C57BL/6 mice actively immunized with myelin oligodendrocyte glycoprotein35-55 (MOG35-55) [9]

Read more

Summary

Introduction

Expression of chemokine CCL2 in the normal central nervous system (CNS) is nearly undetectable, but is significantly upregulated and drives neuroinflammation during experimental autoimmune encephalomyelitis (EAE), an animal model of multiple sclerosis which is considered a contributing factor in the human disease. Elevated CNS expression of CCL2 has been a consistent observation among the different paradigms of experimental autoimmune encephalomyelitis (EAE) [6,7,8], a CNS demyelinating inflammatory disease that serves as a model for multiple sclerosis This chemokine’s singular importance in driving EAE was demonstrated by global CCL2 knockout (KO) mice (CCL2−/−), which showed diminished severity and delay in onset of disease in C57BL/6 mice actively immunized with myelin oligodendrocyte glycoprotein (MOG35-55) [9]. Bone marrow chimera studies further showed active immunization EAE was markedly reduced when bone marrow from WT mice was engrafted into lethally irradiated CCL2−/− mice, but not when bone marrow from CCL2−/− mice was transferred into WT recipients [10] These findings of induced CNS expression of CCL2 during EAE, together with the adoptive transfer and bone marrow chimera studies, are consistent with a prominent role for CNS CCL2 in mediating EAE and diminish or negate the pathogenic impact of CCL2 from the peripheral leukocyte compartment

Methods
Results
Discussion
Conclusion
Full Text
Paper version not known

Talk to us

Join us for a 30 min session where you can share your feedback and ask us any queries you have

Schedule a call

Disclaimer: All third-party content on this website/platform is and will remain the property of their respective owners and is provided on "as is" basis without any warranties, express or implied. Use of third-party content does not indicate any affiliation, sponsorship with or endorsement by them. Any references to third-party content is to identify the corresponding services and shall be considered fair use under The CopyrightLaw.