Rheumatoid arthritis (RA) is mediated in large part by two major effector cells; effector memory T (TEM) lymphocytes and fibroblast-like synoviocytes (FLS). During RA, these cells have phenotypes that result in joint destruction through a variety of mechanisms. TEM cells induce inflammation throughout the synovial joints mediated by the release of a variety of pro-inflammatory cytokines. FLS are resident joint cells that normally function to maintain synovial homeostasis. During RA, their phenotype becomes altered in that they become highly invasive, lose contact inhibition, induce angiogenesis, function as professional antigen-presenting cells, and release pro-inflammatory cytokines. Both of these phenotypes in TEM cells and FLS result in inflammation and joint degradation. It is also likely that these cells interact with each other during RA, either directly through the FLS ability to act as antigen-presenting cells or indirectly through cytokine-induced signaling. The K+ channels Kv1.3 on TEM cells and KCa1.1 on FLS regulate each cell's pathogenic phenotypes. Therefore, I hypothesize that Kv1.3 and KCa1.1 can serve as regulators of the interactions between TEM cells and FLS and a combined therapy of Kv1.3 and KCa1.1 blockers can dramatically reduce disease severity in an in vivo RA model through a synergistic effect. The overall goal of this project is to determine if the interactions of FLS and TEM cells that results in their activation cn be hindered by modulating Kv1.3 and KCa1.1. This will be beneficial as it will provide evidence regarding the benefits of combined therapies targeting both TEM cells and FLS for treating RA. The first objective of this project is to determine if a combined therapy of KCa1.1 and Kv1.3 blockers is more efficacious than single blocker treatments at ameliorating disease severity in a rat model of RA. Rats with a model of RA will be treated with either a single channel blocker, both blockers, and vehicle. Clinical signs of disease will be monitored in each group and histology and x-rays will be used to assess joint damage. Then, ex vivo functional analyses of T cells and FLS isolated from animals in each treatment group will be completed. The second objective is to determine if the cyclical interactions and activations of TEM cells and FLS are mediated by Kv1.3 and KCa1.1 in vitro. This will involve examining the phenotype of FLS and TEM cells co- cultured in the presence or absence of KCa1.1 or Kv1.3 blockers, or from cells with either channel knocked down by siRNA. Proliferation and cytokine production will be examined in both cell-types. FLS expression of molecules needed to directly interact with a T cell will also be examined. Furthermore, it will be determined if FLS can induce differentiation of central memory T cells into TEM cells and if this is dependent on KCa1.1 activity. Overall, this work will validate KCa1.1 and Kv1.3 as regulators of the interactions between FLS and TEM cells and define the benefits of a combined therapy for treating a rat model of RA.

Public Health Relevance

Rheumatoid arthritis is a chronic autoimmune disease that affects approximately 1% of the population in developed regions of the world. Most current medications for treating RA focus on broadly targeting the immune system, which is not beneficial for many people and causes many side effects. We believe that specifically targeting certain immune cells and pathogenic joint cells will provide the basis for more beneficial RA therapies in the future.

Agency
National Institute of Health (NIH)
Institute
National Institute of Arthritis and Musculoskeletal and Skin Diseases (NIAMS)
Type
Predoctoral Individual National Research Service Award (F31)
Project #
1F31AR069960-01
Application #
9117804
Study Section
Special Emphasis Panel (ZRG1-F10B-B (20)L)
Program Officer
Mao, Su-Yau
Project Start
2016-06-01
Project End
2019-05-31
Budget Start
2016-06-01
Budget End
2017-05-31
Support Year
1
Fiscal Year
2016
Total Cost
$40,776
Indirect Cost
Name
Baylor College of Medicine
Department
Physiology
Type
Schools of Medicine
DUNS #
051113330
City
Houston
State
TX
Country
United States
Zip Code
77030
Tanner, Mark R; Beeton, Christine (2018) Differences in ion channel phenotype and function between humans and animal models. Front Biosci (Landmark Ed) 23:43-64
Tanner, Mark R; Pennington, Michael W; Chamberlain, Brayden H et al. (2018) Targeting KCa1.1 Channels with a Scorpion Venom Peptide for the Therapy of Rat Models of Rheumatoid Arthritis. J Pharmacol Exp Ther 365:227-236
Tanner, Mark R; Pennington, Michael W; Laragione, Teresina et al. (2017) KCa1.1 channels regulate ?1-integrin function and cell adhesion in rheumatoid arthritis fibroblast-like synoviocytes. FASEB J 31:3309-3320
Tanner, Mark R; Tajhya, Rajeev B; Huq, Redwan et al. (2017) Prolonged immunomodulation in inflammatory arthritis using the selective Kv1.3 channel blocker HsTX1[R14A] and its PEGylated analog. Clin Immunol 180:45-57
Fortune, Ryan D; Grill, Raymond J; Beeton, Christine et al. (2017) Changes in Gene Expression and Metabolism in the Testes of the Rat following Spinal Cord Injury. J Neurotrauma 34:1175-1186
Tajhya, Rajeev B; Hu, Xueyou; Tanner, Mark R et al. (2016) Functional KCa1.1 channels are crucial for regulating the proliferation, migration and differentiation of human primary skeletal myoblasts. Cell Death Dis 7:e2426
Huq, Redwan; Samuel, Errol L G; Sikkema, William K A et al. (2016) Preferential uptake of antioxidant carbon nanoparticles by T lymphocytes for immunomodulation. Sci Rep 6:33808