Neurostimulation of the cholinergic anti-inflammatory pathway ameliorates disease in rat collagen-induced arthritis

Yaakov A Levine, Frieda A Koopman, Michael Faltys, April Caravaca, Alison Bendele, Ralph Zitnik, Margriet J Vervoordeldonk, Paul Peter Tak, Yaakov A Levine, Frieda A Koopman, Michael Faltys, April Caravaca, Alison Bendele, Ralph Zitnik, Margriet J Vervoordeldonk, Paul Peter Tak

Abstract

Introduction: The inflammatory reflex is a physiological mechanism through which the nervous system maintains immunologic homeostasis by modulating innate and adaptive immunity. We postulated that the reflex might be harnessed therapeutically to reduce pathological levels of inflammation in rheumatoid arthritis by activating its prototypical efferent arm, termed the cholinergic anti-inflammatory pathway. To explore this, we determined whether electrical neurostimulation of the cholinergic anti-inflammatory pathway reduced disease severity in the collagen-induced arthritis model.

Methods: Rats implanted with vagus nerve cuff electrodes had collagen-induced arthritis induced and were followed for 15 days. Animals underwent active or sham electrical stimulation once daily from day 9 through the conclusion of the study. Joint swelling, histology, and levels of cytokines and bone metabolism mediators were assessed.

Results: Compared with sham treatment, active neurostimulation of the cholinergic anti-inflammatory pathway resulted in a 52% reduction in ankle diameter (p = 0.02), a 57% reduction in ankle diameter (area under curve; p = 0.02) and 46% reduction overall histological arthritis score (p = 0.01) with significant improvements in inflammation, pannus formation, cartilage destruction, and bone erosion (p = 0.02), accompanied by numerical reductions in systemic cytokine levels, not reaching statistical significance. Bone erosion improvement was associated with a decrease in serum levels of receptor activator of NF-κB ligand (RANKL) from 132±13 to 6±2 pg/mL (mean±SEM, p = 0.01).

Conclusions: The severity of collagen-induced arthritis is reduced by neurostimulation of the cholinergic anti-inflammatory pathway delivered using an implanted electrical vagus nerve stimulation cuff electrode, and supports the rationale for testing this approach in human inflammatory disorders.

Conflict of interest statement

Competing Interests: YAL, MF, AC and RZ are employees of SetPoint Medical Corporation. PPT is a consultant to and has received research grants from SetPoint Medical. PPT has become an employee of GlaxoSmithKline (GSK) after completion of the study. GSK has obtained an equity interest in Setpoint Medical Corporation. FAK, AB, and MJV have no competing interests to declare. This does not alter the authors' adherence to all PLOS ONE policies on sharing data and materials.

Figures

Figure 1. NCAP Delivery System.
Figure 1. NCAP Delivery System.
(A) Schematic drawing of the percutaneous vagus nerve cuff electrode (B) Surgical implantation of the lead in the neck of a supine animal is shown. The slotted belt (asterisk) is wound around the entire carotid sheath (small arrow) containing the vagus nerve, bringing the electrode pair in close apposition to and in proper orientation on the nerve, allowing effective induction of vagus depolarization. The distal portion of the lead (large arrow) is tunneled subcutaneously to the back, and externalized between the scapulae. (C) Following surgical healing, the distal end of the lead is externalized and protected under a jacket with a Velcro closure. The distal lead is intermittently connected using alligator clips to an external pulse generator for daily active or sham NCAP delivery. (D) The waveform of one cycle of the charge-balanced biphasic pulse delivered by the pulse generator is illustrated schematically. Rats were stimulated once daily for 60 seconds from study day 9 to 15 with a pulse waveform amplitude (PA) of 3 mA, pulsewidth (PW) of 200 microseconds, pulse frequency of 10 Hz, and a 50 microsecond inter-pulse interval (IPI).
Figure 2. NCAP Reduces Clinical Signs of…
Figure 2. NCAP Reduces Clinical Signs of Joint Inflammation.
Rats were placed into four treatment groups, three of which received vagus nerve lead implants, with a fourth group of unimplanted controls. The three implanted groups had CIA induction or sham CIA induction, and active versus sham electrical neurostimulation on study days 9–15 as outlined in Table 1 (Control/No Implant n = 4; Control/NCAP n = 4; CIA/NCAP n = 9; CIA/Sham NCAP n = 12). Ankle diameter over time is shown as mean+SE (A), and AUC of ankle diameter over Day 9–16 is shown as mean+SE (B), *p≤0.05 ANOVA versus CIA/Sham NCAP.
Figure 3. NCAP Reduces Histological Measures of…
Figure 3. NCAP Reduces Histological Measures of Joint Damage.
(A–B) Ankle joints were harvested on study day 16, stained with Toluidine Blue, and scored on a scale of 0-5 for inflammation, pannus formation, cartilage damage and bone resorption (A), with a composite summated score of 0–20 (B). Data are shown as mean+SE score. *p≤0.05 t-test versus CIA/Sham NCAP. (C–D) Representative 50X photomicrographs of ankle joints are shown which have the approximate mean summed score as that of the entire treatment group. Ankle from CIA/Sham NCAP group (C) demonstrates marked inflammation and synovitis (S) and mild cartilage damage (large arrow) and bone resorption (small arrow). Ankle from CIA/NCAP group (D) demonstrates mild inflammation and synovitis (S) and minimal cartilage damage (large arrow) and minimal bone resorption (small arrow).
Figure 4. NCAP Effect on Circulating Cytokines.
Figure 4. NCAP Effect on Circulating Cytokines.
Study day 16 serum was assayed for IL-1α, IL-1β, IL-2, IL-6, IFN-γ and TNF. Data are shown as mean+SE level, t-test p = NS for all individual cytokine comparisons between CIA/NCAP and CIA/Sham NCAP.
Figure 5. NCAP Effect on Mediators of…
Figure 5. NCAP Effect on Mediators of Bone Metabolism.
Study day 16 serum was assayed for RANKL (A), and OPG (B), and the ratio of OPG/RANKL group means calculated (C). Data are shown as mean+SE level, *p≤0.05, t-test versus CIA/Sham NCAP. Osteocalcin (D), P1NP (E), TRAP-5b (F) and CTX-1 (G) data are shown as mean+SE level, *p≤0.05 t-test versus CIA/Sham NCAP.

References

    1. Andersson U, Tracey KJ (2012) Reflex principles of immunological homeostasis. Annu Rev Immunol 30: 313–335.
    1. Huston JM, Ochani M, Rosas-Ballina M, Liao H, Ochani K, et al. (2006) Splenectomy inactivates the cholinergic antiinflammatory pathway during lethal endotoxemia and polymicrobial sepsis. J Exp Med 203: 1623–1628.
    1. Rosas-Ballina M, Olofsson PS, Ochani M, Valdes-Ferrer SI, Levine YA, et al. (2011) Acetylcholine-synthesizing T cells relay neural signals in a vagus nerve circuit. Science 334: 98–101.
    1. Borovikova LV, Ivanova S, Zhang M, Yang H, Botchkina GI, et al. (2000) Vagus nerve stimulation attenuates the systemic inflammatory response to endotoxin. Nature 405: 458–462.
    1. Van Maanen M, Vervoordeldonk M, Tak P (2009) The cholinergic anti-inflammatory pathway: towards innovative treatment of rheumatoid arthritis. Nat Rev Rheumatol 5: 229–232.
    1. van Maanen M, Lebre M, van der Poll T, LaRosa G, Elbaum D, et al. (2009) Stimulation of nicotinic acetylcholine receptors attenuates collagen-induced arthritis in mice. Arthritis Rheum 60: 114–122.
    1. van Maanen MA, Stoof SP, Larosa GJ, Vervoordeldonk MJ, Tak PP (2010) Role of the cholinergic nervous system in rheumatoid arthritis: aggravation of arthritis in nicotinic acetylcholine receptor alpha7 subunit gene knockout mice. Ann Rheum Dis 69: 1717–1723.
    1. Beekwilder JP, Beems T (2010) Overview of the clinical applications of vagus nerve stimulation. J Clin Neurophysiol 27: 130–138.
    1. National Research Council (U.S.). Committee for the Update of the Guide for the Care and Use of Laboratory Animals., Institute for Laboratory Animal Research (U.S.), National Academies Press (U.S.) (2011) Guide for the care and use of laboratory animals. Washington, D.C.: National Academies Press. xxv, 220 p. p.
    1. Bendele A, McAbee T, Woodward M, Scherrer J, Collins D, et al. (1998) Effects of interleukin-1 receptor antagonist in a slow-release hylan vehicle on rat type II collagen arthritis. Pharm Res 15: 1557–1561.
    1. Bendele AM, Chlipala ES, Scherrer J, Frazier J, Sennello G, et al. (2000) Combination benefit of treatment with the cytokine inhibitors interleukin-1 receptor antagonist and PEGylated soluble tumor necrosis factor receptor type I in animal models of rheumatoid arthritis. Arthritis Rheum 43: 2648–2659.
    1. Huston JM, Rosas-Ballina M, Xue X, Dowling O, Ochani K, et al. (2009) Cholinergic neural signals to the spleen down-regulate leukocyte trafficking via CD11b. J Immunol 183: 552–559.
    1. de Jonge WJ, van der Zanden EP, The FO, Bijlsma MF, van Westerloo DJ, et al. (2005) Stimulation of the vagus nerve attenuates macrophage activation by activating the Jak2-STAT3 signaling pathway. Nat Immunol 6: 844–851.
    1. Saeed R, Varma S, Peng-Nemeroff T (2005) Cholinergic stimulation blocks endothelial cell activation and leukocyte recruitment during inflammation. J Exp Med 201: 1113–1123.
    1. Karimi K, Bienenstock J, Wang L, Forsythe P (2010) The vagus nerve modulates CD4+ T cell activity. Brain Behav Immun 24: 316–323.
    1. Galitovskiy V, Qian J, Chernyavsky AI, Marchenko S, Gindi V, et al. (2011) Cytokine-Induced Alterations of {alpha}7 Nicotinic Receptor in Colonic CD4 T Cells Mediate Dichotomous Response to Nicotine in Murine Models of Th1/Th17- versus Th2-Mediated Colitis. J Immunol 187: 2677–2687.
    1. Ghia JE, Blennerhassett P, Collins SM (2007) Vagus nerve integrity and experimental colitis. Am J Physiol Gastrointest Liver Physiol 293: G560–567.
    1. Ghia JE, Blennerhassett P, Kumar-Ondiveeran H, Verdu EF, Collins SM (2006) The vagus nerve: a tonic inhibitory influence associated with inflammatory bowel disease in a murine model. Gastroenterology 131: 1122–1130.
    1. Mina-Osorio P, Rosas-Ballina M, Valdes-Ferrer SI, Al-Abed Y, Tracey KJ, et al. (2012) Neural signaling in the spleen controls B-cell responses to blood-borne antigen. Mol Med 18: 618–627.
    1. van Maanen M, Stoof S, van der Zanden E, de Jonge W, Janssen R, et al. (2009) The alpha7 nicotinic acetylcholine receptor on fibroblast-like synoviocytes and in synovial tissue from rheumatoid arthritis patients: a possible role for a key neurotransmitter in synovial inflammation. Arthritis Rheum 60: 1272–1281.
    1. Waldburger JM, Boyle DL, Pavlov VA, Tracey KJ, Firestein GS (2008) Acetylcholine regulation of synoviocyte cytokine expression by the alpha7 nicotinic receptor. Arthritis Rheum 58: 3439–3449.
    1. Zhang P, Han D, Tang T, Zhang X, Dai K (2008) Inhibition of the development of collagen-induced arthritis in Wistar rats through vagus nerve suspension: a 3-month observation. Inflamm Res 57: 322–328.
    1. Heck C, Helmers SL, DeGiorgio CM (2002) Vagus nerve stimulation therapy, epilepsy, and device parameters: scientific basis and recommendations for use. Neurology 59: S31–37.
    1. Koo B, Ham SD, Sood S, Tarver B (2001) Human vagus nerve electrophysiology: a guide to vagus nerve stimulation parameters. J Clin Neurophysiol 18: 429–433.
    1. The Vagus Nerve Stimulation Study Group (1995) A randomized controlled trial of chronic vagus nerve stimulation for treatment of medically intractable seizures. Neurology 45: 224–230.
    1. Huston JM, Gallowitsch-Puerta M, Ochani M, Ochani K, Yuan R, et al. (2007) Transcutaneous vagus nerve stimulation reduces serum high mobility group box 1 levels and improves survival in murine sepsis. Crit Care Med 35: 2762–2768.
    1. Levine YA, Caravaca A, Faltys M, Arnold A, Zitnik R (2013) Vagus nerve stimulation provides prolonged and spleen-independent protection against indomethacin-induced intestinal inflammation. Gastroenterology 144: S67 (abstract)..
    1. Lacey DL, Boyle WJ, Simonet WS, Kostenuik PJ, Dougall WC, et al. (2012) Bench to bedside: elucidation of the OPG-RANK-RANKL pathway and the development of denosumab. Nat Rev Drug Discov 11: 401–419.
    1. Saidenberg-Kermanac'h N, Corrado A, Lemeiter D, deVernejoul MC, Boissier MC, et al. (2004) TNF-alpha antibodies and osteoprotegerin decrease systemic bone loss associated with inflammation through distinct mechanisms in collagen-induced arthritis. Bone 35: 1200–1207.
    1. Stolina M, Schett G, Dwyer D, Vonderfecht S, Middleton S, et al. (2009) RANKL inhibition by osteoprotegerin prevents bone loss without affecting local or systemic inflammation parameters in two rat arthritis models: comparison with anti-TNFalpha or anti-IL-1 therapies. Arthritis Res Ther 11: R187.
    1. Dore RK, Cohen SB, Lane NE, Palmer W, Shergy W, et al. (2010) Effects of denosumab on bone mineral density and bone turnover in patients with rheumatoid arthritis receiving concurrent glucocorticoids or bisphosphonates. Ann Rheum Dis 69: 872–875.
    1. Redlich K, Smolen JS (2012) Inflammatory bone loss: pathogenesis and therapeutic intervention. Nat Rev Drug Discov 11: 234–250.
    1. Stolina M, Adamu S, Ominsky M, Dwyer D, Asuncion F, et al. (2005) RANKL is a Marker and Mediator of Local and Systemic Bone Loss in Two Rat Models of Inflammatory Arthritis. Journal of Bone and Mineral Research 20: 1756–1765.
    1. Bajayo A, Bar A, Denes A, Bachar M, Kram V, et al. (2012) Skeletal parasympathetic innervation communicates central IL-1 signals regulating bone mass accrual. Proc Natl Acad Sci U S A 109: 15455–15460.
    1. Famm K, Litt B, Tracey KJ, Boyden ES, Slaoui M (2013) Drug discovery: a jump-start for electroceuticals. Nature 496: 159–161.

Source: PubMed

3
Prenumerera