Abstract

Histamine H3 receptor (Hrh3/H3R) is primarily expressed by neurons in the central nervous system (CNS) where it functions as a presynaptic inhibitory autoreceptor and heteroreceptor. Previously, we identified an H3R-mediated central component in susceptibility to experimental allergic encephalomyelitis (EAE), the principal autoimmune model of multiple sclerosis (MS), related to neurogenic control of blood brain barrier permeability and peripheral T cell effector responses. Furthermore, we identified Hrh3 as a positional candidate for the EAE susceptibility locus Eae8. Here, we characterize Hrh3 polymorphisms between EAE-susceptible and resistant SJL and B10.S mice, respectively, and show that Hrh3 isoform expression in the CNS is differentially regulated by acute peripheral inflammatory stimuli in an allele-specific fashion. Next, we show that Hrh3 is not expressed in any subpopulations of the immune compartment, and that secondary lymphoid tissue is anatomically poised to be regulated by central H3R signaling. Accordingly, using transcriptome analysis, we show that, inflammatory stimuli elicit unique transcriptional profiles in the lymph nodes of H3RKO mice compared to WT mice, which is indicative of negative regulation of peripheral immune responses by central H3R signaling. These results further support a functional link between the neurogenic control of T cell responses and susceptibility to CNS autoimmune disease coincident with acute and/or chronic peripheral inflammation. Pharmacological targeting of H3R may therefore be useful in preventing the development and formation of new lesions in MS, thereby limiting disease progression.

Highlights

  • Multiple sclerosis (MS), a chronic inflammatory disease of the central nervous system (CNS), is the most common disabling neurologic disease of young adults and adolescents affecting,350,000 individuals in the United States and more than 1 million individuals worldwide [1]

  • An examination of 18 different inbred strains of mice using restriction fragment length polymorphism (RFLP) analysis confirmed the existence of two alleles segregating among the various inbred strains (Fig. 1)

  • To exclude the possibility that H3R is influencing immune responses directly through its expression in either secondary lymphoid tissues and/or hematopoietic cells including T cells, we examined its expression in the spleen and lymph node (LN) of naive animals, and by macrophages, mast cells, neutrophils, bone marrow-derived dendritic cells, B cells (B220+), effector CD8+ (TCRb+CD8+CD42), naive CD4+ (TCRb+ CD4+CD82CD25lowCD45RhighCD44lowCD1d-tetramer-), memory CD4+ (TCRb+CD4+CD25lowCD45RBlowCD44highCD1d-tetramer), NKT (CD1d tetramer+), and Treg cells (TCRb+CD4+Foxp3+) by RT-PCR. mRNA for any of the known Hrh3 isoforms was undetectable in both whole spleen and LN, as well as in all cells of the innate and adaptive immune systems studied

Read more

Summary

Introduction

Multiple sclerosis (MS), a chronic inflammatory disease of the central nervous system (CNS), is the most common disabling neurologic disease of young adults and adolescents affecting ,350,000 individuals in the United States and more than 1 million individuals worldwide [1]. We characterize Hrh3 polymorphisms between EAE-susceptible and resistant SJL and B10.S mice, respectively, and show that Hrh3 isoform expression in the CNS is differentially regulated by acute peripheral inflammatory stimuli in an allele-specific fashion.

Results
Conclusion
Full Text
Published version (Free)

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