Noninvasive ultrasound stimulation of the spleen to treat inflammatory arthritis

Daniel P Zachs, Sarah J Offutt, Rachel S Graham, Yohan Kim, Jerel Mueller, Jennifer L Auger, Nathaniel J Schuldt, Claire R W Kaiser, Abigail P Heiller, Raini Dutta, Hongsun Guo, Jamu K Alford, Bryce A Binstadt, Hubert H Lim, Daniel P Zachs, Sarah J Offutt, Rachel S Graham, Yohan Kim, Jerel Mueller, Jennifer L Auger, Nathaniel J Schuldt, Claire R W Kaiser, Abigail P Heiller, Raini Dutta, Hongsun Guo, Jamu K Alford, Bryce A Binstadt, Hubert H Lim

Abstract

Targeted noninvasive control of the nervous system and end-organs may enable safer and more effective treatment of multiple diseases compared to invasive devices or systemic medications. One target is the cholinergic anti-inflammatory pathway that consists of the vagus nerve to spleen circuit, which has been stimulated with implantable devices to improve autoimmune conditions such as rheumatoid arthritis. Here we report that daily noninvasive ultrasound (US) stimulation targeting the spleen significantly reduces disease severity in a mouse model of inflammatory arthritis. Improvements are observed only with specific parameters, in which US can provide both protective and therapeutic effects. Single cell RNA sequencing of splenocytes and experiments in genetically-immunodeficient mice reveal the importance of both T and B cell populations in the anti-inflammatory pathway. These findings demonstrate the potential for US stimulation of the spleen to treat inflammatory diseases.

Conflict of interest statement

The authors declare no competing interests.

Figures

Fig. 1
Fig. 1
Modulation of the cholinergic anti-inflammatory pathway through the vagus nerve, splenic nerve and spleen. a Electrical stimulation of the vagus nerve or US stimulation of the spleen is thought to modulate the neural communication with T Cells and Macrophages, blocking the production of inflammatory cytokines and promoting an anti-inflammatory state. b Timeline of a typical experiment performed in the presented study in which animals were injected with 300 μl of K/BxN serum on day 0 and treated with focused US that targeted the spleen on days -1 through 6
Fig. 2
Fig. 2
US treatment modulates arthritis severity. Results from 7-day arthritis experiments are presented. ad Pooled data from three experiments (total n = 32 mice), using identical experimental conditions (1 MHz US at 350 kPa, 1 s on/5 s off bursts for 2 min per day; shallow US-focusing cone to target the spleen, see Supplementary Fig. 2a) are shown. On the final day of the experiment, change in ankle thickness (a) and clinical score (b) of US treated animals are significantly reduced compared to sham-US controls (p = 0.0017 and p = 0.0060, respectively, using a Mann–Whitney Test). For comparison, normalized data from the same set of experiments are shown in c, d. A normalized value of zero indicates that the animal’s arthritis outcome was similar to the average untreated (sham-US) animal, a positive value indicates clinical worsening, and a negative value indicates clinical improvement. e, f Dose-response curves of US pressure reveal an optimal US amplitude between 333–350 kPa. Each point represents the normalized mean change of US treatment compared to sham at day 7. 1 MHz US stimulation with 1 s on/5 s off bursts for 2 min per day was applied in each experiment. Circles indicate that the shallow US-focusing cone was used, and triangles indicate that the deep US-focusing cone was used. Results are pooled from 108 mice across 9 experiments (see Mice section of Methods). g Additional US stimulation parameters and body locations were also tested, revealing consistent and effective therapeutic effects when specifically using 350 kPa, 1 s on/5 s off bursts with the shallow US-focusing cone targeting the spleen. One hundred and three mice were used over 9 experiments. All conditions consisted of 2 min of US stimulation per day, except for the right leg conditions that consisted of 4 min per day. Contra-spleen refers to right abdominal stimulation contralateral to the spleen. Tcra KO and muMt- were spleen stimulation experiments performed in T cell and B cell knockout mice, respectively. Mean and SEM are shown for all figures. Double asterisks denote p < 0.01. kPa kilopascal
Fig. 3
Fig. 3
US treatment is effective before or after arthritis onset and depends on duration of stimulation period (page 18). All experiments in this figure used the 1 MHz US at 350 kPa, 1 s on/5 s off bursts in animals with arthritic serum transfer at day 0. a, b Full 7-day time course pooling data from the three experiments shown previously (Fig. 2a, b), which used identical experimental conditions for 2 min per day with US administered daily from day -1 through 6 (total n = 32). c, d Ultrasound duration causes a dose-dependent effect on arthritis. 6, 12, and 20 minute US stimulation was tested and is shown with the 2 min US and sham results from a, b for comparison. Figures show the 7-day progression of the disease and mice were stimulated with US from day -1 through day 6. For the 6, 12, and 20 min experiments, there were 6 animals per cohort (total n = 18) and for the 2 minute US and control data there were 16 animals per cohort (total n = 32). e, f US treatment is also effective at reducing symptoms after onset of arthritis. Treatment was not initiated until day 3 after arthritis had manifested. US was administered for 20 min per day. Data was pooled from two experiments with a total n = 23. g, h Normalized 7th day values of ankle thickness (g) and clinical score (h) for the different durations of ultrasound treatment shown in 3a–f. Error bars are SEM. 6, 12, and 20 minute values were normalized to the pooled sham control data from a, b. The bar “20 min US initiated on day 3” shows the improvement in ankle thickness and clinical score compared to the first day of treatment on day 3. In af, whisker plots are presented for better visualization of the data over time in which the endpoints of the vertical lines span the minimum to maximum values, the midline of each box is the median value and each box extends from the 25th to 75th percentile. In g, h, mean and SEM are plotted for each condition. kPa kilopascal
Fig. 4
Fig. 4
Splenic T and B cells demonstrate induction of genes following US treatment. The dot plot displays the statistically significantly differentially expressed genes (DEGs) in T cells (top list) and B cells (bottom list) from mice given arthritogenic serum comparing US versus sham-US stimulation. The size of each circle represents the percent of cells within each cell type (T cells = T, B cells = B) which express the gene listed, and the color of each circle represents average scaled expression. Gray bars denote mouse treatment groups; genes that are in bold are statistically significantly DEG in both T cells and B cells, and genes found to also be DEG in non-arthritic mice with US stimulation (see Supplementary Fig. 8) are denoted with a superscript x. Adjusted p-values for each gene in either T or B cells (top or bottom gene lists) are shown in shades of green. All DEGs listed in this Figure range in statistical significance from p = 2.39e-02 to p = 3.95e-230 using a Wilcox rank sum test with Bonferroni correction

References

    1. Borovikova LV, et al. Vagus nerve stimulation attenuates the systemic inflammatory response to endotoxin. Nature. 2000;405:458–462. doi: 10.1038/35013070.
    1. Bai A, Guo Y, Lu N. The effect of the cholinergic anti-inflammatory pathway on experimental colitis. Scand. J. Immunol. 2007;66:538–545. doi: 10.1111/j.1365-3083.2007.02011.x.
    1. The F, et al. Central activation of the cholinergic anti-inflammatory pathway reduces surgical inflammation in experimental post-operative ileus. Br. J. Pharmacol. 2011;163:1007–1016. doi: 10.1111/j.1476-5381.2011.01296.x.
    1. Andersson U, Tracey KJ. Neural reflexes in inflammation and immunity. J. Exp. Med. 2012;209:1057–1068. doi: 10.1084/jem.20120571.
    1. Huston JM, et al. Splenectomy inactivates the cholinergic antiinflammatory pathway during lethal endotoxemia and polymicrobial sepsis. J. Exp. Med. 2006;203:1623–1628. doi: 10.1084/jem.20052362.
    1. Rosas-Ballina M, et al. Splenic nerve is required for cholinergic antiinflammatory pathway control of TNF in endotoxemia. Proc. Natl Acad. Sci. USA. 2008;105:11008–11013. doi: 10.1073/pnas.0803237105.
    1. Ji H, et al. Central cholinergic activation of a vagus nerve-to-spleen circuit alleviates experimental colitis. Mucosal Immunol. 2014;7:335–347. doi: 10.1038/mi.2013.52.
    1. Levine YA, et al. Neurostimulation of the cholinergic anti-inflammatory pathway ameliorates disease in rat collagen-induced arthritis. PLoS One. 2014;9:e104530. doi: 10.1371/journal.pone.0104530.
    1. van Maanen MA, Vervoordeldonk MJ, Tak PP. The cholinergic anti-inflammatory pathway: towards innovative treatment of rheumatoid arthritis. Nat. Rev. Rheumatol. 2009;5:229–232. doi: 10.1038/nrrheum.2009.31.
    1. Koopman FA, et al. Vagus nerve stimulation inhibits cytokine production and attenuates disease severity in rheumatoid arthritis. Proc. Natl Acad. Sci. USA. 2016;113:8284–8289. doi: 10.1073/pnas.1605635113.
    1. Gigliotti JC, et al. Ultrasound prevents renal ischemia-reperfusion injury by stimulating the splenic cholinergic anti-inflammatory pathway. J. Am. Soc. Nephrol. 2013;24:1451–1460. doi: 10.1681/ASN.2013010084.
    1. Gigliotti JC, et al. Ultrasound modulates the splenic neuroimmune axis in attenuating AKI. J. Am. Soc. Nephrol. 2015;26:2470–2481. doi: 10.1681/ASN.2014080769.
    1. Cotero, V. et al. Noninvasive sub-organ ultrasound stimulation for targeted neuromodulation. Nat Commun. 2019
    1. Stangenberg L, et al. Denervation protects limbs from inflammatory arthritis via an impact on the microvasculature. Proc. Natl Acad. Sci. USA. 2014;111:11419–11424. doi: 10.1073/pnas.1410854111.
    1. Lee DM, et al. Mast cells: a cellular link between autoantibodies and inflammatory arthritis. Science. 2002;297:1689–1692. doi: 10.1126/science.1073176.
    1. Binstadt BA, et al. The same systemic autoimmune disease provokes arthritis and endocarditis via distinct mechanisms. Proc. Natl Acad. Sci. USA. 2009;106:16758–16763. doi: 10.1073/pnas.0909132106.
    1. Kim H, et al. Noninvasive transcranial stimulation of rat abducens nerve by focused ultrasound. Ultrasound Med. Biol. 2012;38:1568–1575. doi: 10.1016/j.ultrasmedbio.2012.04.023.
    1. Downs ME, et al. Non-invasive peripheral nerve stimulation via focused ultrasound in vivo. Phys. Med. Biol. 2018;63:035011. doi: 10.1088/1361-6560/aa9fc2.
    1. Lele PP. Effects of focused ultrasonic radiation on peripheral nerve, with observations on local heating. Exp. Neurol. 1963;8:47–83. doi: 10.1016/0014-4886(63)90008-6.
    1. Tyler WJ, et al. Remote excitation of neuronal circuits using low-intensity, low-frequency ultrasound. PLoS One. 2008;3:e3511. doi: 10.1371/journal.pone.0003511.
    1. Wright CJ, Haqshenas SR, Rothwell J, Saffari N. Unmyelinated peripheral nerves can be stimulated in vitro using pulsed ultrasound. Ultrasound Med. Biol. 2017;43:2269–2283. doi: 10.1016/j.ultrasmedbio.2017.05.008.
    1. Legon W, Rowlands A, Opitz A, Sato TF, Tyler WJ. Pulsed ultrasound differentially stimulates somatosensory circuits in humans as indicated by EEG and FMRI. PLoS One. 2012;7:e51177. doi: 10.1371/journal.pone.0051177.
    1. Kubanek J, Shukla P, Das A, Baccus SA, Goodman MB. Ultrasound elicits behavioral responses through mechanical effects on neurons and ion channels in a simple nervous system. J. Neurosci. 2018;38:3081–3091. doi: 10.1523/JNEUROSCI.1458-17.2018.
    1. Deng CX, Sieling F, Pan H, Cui J. Ultrasound-induced cell membrane porosity. Ultrasound Med. Biol. 2004;30:519–526. doi: 10.1016/j.ultrasmedbio.2004.01.005.
    1. Cesare P, Moriondo A, Vellani V, McNaughton PA. Ion channels gated by heat. Proc. Natl Acad. Sci. USA. 1999;96:7658–7663. doi: 10.1073/pnas.96.14.7658.
    1. Shapiro MG, Homma K, Villarreal S, Richter CP, Bezanilla F. Infrared light excites cells by changing their electrical capacitance. Nat. Commun. 2012;3:736. doi: 10.1038/ncomms1742.
    1. Chinenov Y, Kerppola TK. Close encounters of many kinds: Fos-Jun interactions that mediate transcription regulatory specificity. Oncogene. 2001;20:2438–2452. doi: 10.1038/sj.onc.1204385.
    1. Han S, Brunet A. Histone methylation makes its mark on longevity. Trends Cell Biol. 2012;22:42–49. doi: 10.1016/j.tcb.2011.11.001.
    1. Hannemann N, et al. The AP-1 transcription factor c-Jun promotes arthritis by regulating cyclooxygenase-2 and arginase-1 expression in macrophages. J. Immunol. 2017;198:3605–3614. doi: 10.4049/jimmunol.1601330.
    1. Bunnell TM, Burbach BJ, Shimizu Y, Ervasti JM. β-Actin specifically controls cell growth, migration, and the G-actin pool. Mol. Biol. Cell. 2011;22:4047–4058. doi: 10.1091/mbc.e11-06-0582.
    1. Mina-Osorio P, et al. Neural signaling in the spleen controls B-cell responses to blood-borne antigen. Mol. Med. 2012;18:618–627. doi: 10.2119/molmed.2012.00027.
    1. Mazzone SB, Undem BJ. Vagal afferent innervation of the airways in health and disease. Physiol. Rev. 2016;96:975–1024. doi: 10.1152/physrev.00039.2015.
    1. Breit S, Kupferberg A, Rogler G, Hasler G. Vagus nerve as modulator of the brain-gut axis in psychiatric and inflammatory disorders. Front Psychiatry. 2018;9:44. doi: 10.3389/fpsyt.2018.00044.
    1. Garamendi-Ruiz, I. & Gómez-Esteban, J. C. Cardiovascular autonomic effects of vagus nerve stimulation. Clin. Auton. Res. 10.1007/s10286-017-0477-8 (2017).
    1. Oliveira TVHF, Francisco AN, Demartini Z, Stebel SL. The role of vagus nerve stimulation in refractory epilepsy. Arq. Neuropsiquiatr. 2017;75:657–666. doi: 10.1590/0004-282x20170113.
    1. Elliott RE, et al. Vagus nerve stimulation in 436 consecutive patients with treatment-resistant epilepsy: long-term outcomes and predictors of response. Epilepsy Behav. 2011;20:57–63. doi: 10.1016/j.yebeh.2010.10.017.
    1. Monach, P. A., Mathis, D. & Benoist, C. The K/BxN arthritis model. Curr. Protoc. Immunol. doi: 10.1002/0471142735.im1522s81 (2008).
    1. Satija R, Farrell JA, Gennert D, Schier AF, Regev A. Spatial reconstruction of single-cell gene expression data. Nat. Biotechnol. 2015;33:495–502. doi: 10.1038/nbt.3192.

Source: PubMed

3
Subscribe