Skip to main content

Advertisement

Log in

Selective Knockdown of TASK3 Potassium Channel in Monoamine Neurons: a New Therapeutic Approach for Depression

  • Published:
Molecular Neurobiology Aims and scope Submit manuscript

Abstract

Current pharmacological treatments for major depressive disorder (MDD) are severely compromised by both slow action and limited efficacy. RNAi strategies have been used to evoke antidepressant-like effects faster than classical drugs. Using small interfering RNA (siRNA), we herein show that TASK3 potassium channel knockdown in monoamine neurons induces antidepressant-like responses in mice. TASK3-siRNAs were conjugated to cell-specific ligands, sertraline (Ser) or reboxetine (Reb), to promote their selective accumulation in serotonin (5-HT) and norepinephrine (NE) neurons, respectively, after intranasal delivery. Following neuronal internalization of conjugated TASK3-siRNAs, reduced TASK3 mRNA and protein levels were found in the brainstem 5-HT and NE cell groups. Moreover, Ser-TASK3-siRNA induced robust antidepressant-like behaviors, enhanced the hippocampal plasticity, and potentiated the fluoxetine-induced increase on extracellular 5-HT. Similar responses, yet of lower magnitude, were detected for Reb-TASK3-siRNA. These findings provide substantial support for TASK3 as a potential target, and RNAi-based strategies as a novel therapeutic approach to treat MDD.

This is a preview of subscription content, log in via an institution to check access.

Access this article

Price excludes VAT (USA)
Tax calculation will be finalised during checkout.

Instant access to the full article PDF.

Fig. 1
Fig. 2
Fig. 3
Fig. 4
Fig. 5
Fig. 6

Similar content being viewed by others

References

  1. Murray CJ, Vos T, Lozano R, Naghavi M, Flaxman AD, Michaud C et al (2012) Disability-adjusted life years (DALYs) for 291 diseases and injuries in 21 regions, 1990-2010: a systematic analysis for the global burden of disease study 2010. Lancet 380:2197–2223

    Article  Google Scholar 

  2. Whiteford HA, Degenhardt L, Rehm J, Baxter AJ, Ferrari AJ, Erskine HE, Charlson FJ, Norman RE et al (2013) Global burden of disease attributable to mental and substance use disorders: findings from the global burden of disease study 2010. Lancet 382:1575–1586

    Article  Google Scholar 

  3. Global Burden of Disease Study 2013 Collaborators (2015) Global, regional, and national incidence, prevalence, and years lived with disability for 301 acute and chronic diseases and injuries in 188 countries, 1990-2013: a systematic analysis for the global burden of disease study 2013. Lancet 386:743–800

    Article  Google Scholar 

  4. Trivedi MH, Rush AJ, Wisniewski SR, Nierenberg AA, Warden D, Ritz L, Norquist G, Howland RH et al (2006) Evaluation of outcomes with citalopram for depression using measurement-based care in STAR*D: implications for clinical practice. Am J Psychiatry 163:28–40

    Article  Google Scholar 

  5. Trivedi MH, Fava M, Wisniewski SR, Thase ME, Quitkin F, Warden D, Ritz L, Nierenberg AA et al (2006) Medication augmentation after the failure of SSRIs for depression. N Engl J Med 354:1243–1252

    Article  CAS  Google Scholar 

  6. Rush AJ, Trivedi MH, Wisniewski SR, Nierenberg AA, Stewart JW, Warden D et al (2006) Acute and longer-term outcomes in depressed outpatients requiring one or several treatment steps: a STAR*D report. Am J Psychiatry 163:1905–1917

    Article  Google Scholar 

  7. Artigas F, Bortolozzi A (2017) Therapeutic potential of conjugated siRNAs for the treatment of major depressive disorder. Neuropsychopharmacol 42:371

    Article  CAS  Google Scholar 

  8. Artigas F, Celada P, Bortolozzi A (2018) Can we increase the speed and efficacy of antidepressant treatments? Part II Glutamatergic and RNA interference strategies. Eur Neuropsychopharmacol 28:457–482. https://doi.org/10.1016/j.euroneuro.2018.01.005

    Article  CAS  PubMed  Google Scholar 

  9. Bortolozzi A, Castañé A, Semakova J, Santana N, Alvarado G, Cortés R, Ferrés-Coy A, Fernández G et al (2012) Selective siRNA-mediated suppression of 5-HT1A autoreceptors evokes strong anti-depressant-like effects. Mol Psychiatry 17:612–623

    Article  CAS  Google Scholar 

  10. Ferrés-Coy A, Galofré M, Pilar-Cuéllar F, Vidal R, Paz V, Ruiz-Bronchal E, Campa L, Pazos Á et al (2016) Therapeutic antidepressant potential of a conjugated siRNA silencing the serotonin transporter after intranasal administration. Mol Psychiatry 21:328–338

    Article  Google Scholar 

  11. Ferrés-Coy A, Santana N, Castañé A, Cortés R, Carmona MC, Toth M, Montefeltro A, Artigas F et al (2013) Acute 5-HT1A autoreceptor knockdown increases antidepressant responses and serotonin release in stressful conditions. Psychopharmacology 225:61–74

    Article  Google Scholar 

  12. Ferrés-Coy A, Pilar-Cuellar F, Vidal R, Paz V, Masana M, Cortés R et al (2013) RNAi-mediated serotonin transporter suppression rapidly increases serotonergic neurotransmission and hippocampal neurogenesis. Transl Psychiatry 3:11e211

    Article  Google Scholar 

  13. Rajan S, Wischmeyer E, Xin Liu G, Preisig-Müller R, Daut J, Karschin A, Derst C (2000) TASK-3, a novel tandem pore domain acid-sensitive K+ channel. An extracellular histiding as pH sensor. J Biol Chem 275:16650–16657

    Article  CAS  Google Scholar 

  14. Bayliss DA, Barrett PQ (2008) Emerging roles for two-pore-domain potassium channels and their potential therapeutic impact. Trends Pharmacol Sci 29:566–575

    Article  CAS  Google Scholar 

  15. Russo SJ, Murrough JW, Han MH, Charney DS, Nestler EJ (2012) Neurobiology of resilience. Nat Neurosci 15:1475–1484

    Article  CAS  Google Scholar 

  16. Borsotto M, Veyssiere J, Moha Ou Maati H, Devader C, Mazella J, Heurteaux C (2015) Targeting two-pore domain K(+) channels TREK-1 and TASK-3 for the treatment of depression: a new therapeutic concept. Br J Pharmacol 172:771–784

    Article  CAS  Google Scholar 

  17. Gotter AL, Santarelli VP, Doran SM, Tannenbaum PL, Kraus RL, Rosahl TW, Meziane H, Montial M et al (2011) TASK-3 as a potential antidepressant target. Brain Res 1416:69–79

    Article  CAS  Google Scholar 

  18. Coburn CA, Luo Y, Cui M, Wang J, Soll R, Dong J, Hu B, Lyon MA et al (2012) Discovery of a pharmacologically active antagonist of the two-pore-domain potassium channel K2P9.1 (TASK-3). Chem Med Chem 7:123–133

    Article  CAS  Google Scholar 

  19. Karschin C, Wischmeyer E, Preisig-Müller R, Rajan S, Derst C, Grzeschik KH, Daut J, Karschin A (2001) Expression pattern in brain of TASK-1, TASK-3, and a tandem pore domain K+ channel subunit, TASK-5, associated with the central auditory nervous system. Mol Cell Neurosci 18:632–648

    Article  CAS  Google Scholar 

  20. Meadows HJ, Randall AD (2001) Functional characterisation of human TASK-3, an acid-sensitive two-pore domain potassium channel. Neuropharmacology 40:551–559

    Article  CAS  Google Scholar 

  21. Medhurst A, Rennie G, Chapman C, Meadows H, Duckworth M, Kelsell R et al (2001) Distribution analysis of human two pore domain potassium channels in tissues of the central nervous system and periphery. Mol Brain Res 86:101–114

    Article  CAS  Google Scholar 

  22. Talley EM, Solorzano G, Lei Q, Kim D, Bayliss DA (2001) CNS distribution of members of the two-pore-domain (KCNK) potassium channel family. J Neurosci 21:7491–7505

    Article  CAS  Google Scholar 

  23. Marinc C, Preisig-Müller R, Prüss H, Derst C, Veh RW (2011) Immunocytochemical localization of TASK-3 (K2P 9.1) channels in monoaminergic and cholinergic neurons. Cell Mol Neurobiol 31:323–335

    Article  CAS  Google Scholar 

  24. Linden AM, Sandu C, Aller MI, Vekovischeva OY, Rosenberg PH, Wisden W, Korpi ER (2007) TASK-3 knockout mice exhibit exaggerated nocturnal activity, impairments in cognitive functions, and reduced sensitivity to inhalation anesthetics. J Pharmacol Exp Ther 323:924–934

    Article  CAS  Google Scholar 

  25. Alarcón-Arís D, Recasens A, Galofré M, Carballo-Carbajal I, Zacchi N, Ruiz-Bronchal E, Pavia-Collado R, Chica R et al (2018) Selective α-synuclein knockdown in monoamine neurons by intranasal oligonucleotide delivery: potential therapy for Parkinson’s disease. Mol Ther 26:550–567

    Article  Google Scholar 

  26. Franklin KBJ, Paxinos G (2008) The mouse brain in stereotaxic coordinates. Academic Press, New York

    Google Scholar 

  27. Mateo Y, Meana JJ (1999) Determination of the somatodendritic alpha2-adrenoceptor subtype located in rat locus coeruleus that modulates cortical noradrenaline release in vivo. Eur J Pharmacol 379:53–57

    Article  CAS  Google Scholar 

  28. Mateo Y, Fernández-Pastor B, Meana JJ (2001) Acute and chronic effects of desipramine and clorgyline on alpha(2)-adrenoceptors regulating noradrenergic transmission in the rat brain: a dual-probe microdialysis study. Br J Pharmacol 133:1362–1370

    Article  CAS  Google Scholar 

  29. Ortega JE, Katner J, Davis R, Wade M, Nisenbaum L, Nomikos GG, Svensson KA, Perry KW (2012) Modulation of neurotransmitter release in orexin/hypocretin-2 receptor knockout mice: a microdialysis study. J Neurosci Res 90:588–596

    Article  CAS  Google Scholar 

  30. Samuels BA, Hen R (2011) Novelty-suppressed feeding in the mouse. In: Gould TD (ed) Mood and anxiety related phenotypes in mice: characterization using behavioral test, Volume II. Springer, New York, pp. 107–121

    Chapter  Google Scholar 

  31. Zhao C, Deng W, Gage FH (2008) Mechanisms and functional implications of adult neurogenesis. Cell 132:645–660

    Article  CAS  Google Scholar 

  32. Duman RS, Voleti B (2012) Signaling pathways underlying the pathophysiology and treatment of depression: novel mechanisms for rapid-acting agents. Trends Neurosci 35:47–56

    Article  CAS  Google Scholar 

  33. Javitch JA, Strittmatter SM, Snyder SH (1985) Differential visualization of dopamine and norepinephrine uptake sites in rat brain using [3H]mazindol autoradiography. J Neurosci 5:1513–1521

    Article  CAS  Google Scholar 

  34. Cortés R, Soriano E, Pazos A, Probst A, Palacios JM (1988) Autoradiography of antidepressant binding sites in the human brain: localization using [3H]imipramine and [3H]paroxetine. Neuroscience 27:473–496

    Article  Google Scholar 

  35. Wang JW, David DJ, Monckton JE, Battaglia F, Hen R (2008) Chronic fluoxetine stimulates maturation and synaptic plasticity of adult-born hippocampal granule cells. J Neurosci 28:1374–1384

    Article  CAS  Google Scholar 

  36. Brachman RA, McGowan JC, Perusini JN, Lim SC, Pham TH, Faye C et al (2016) Ketamine as a prophylactic against stress-induced depressive-like behavior. Biol Psychiatry 79:776–786

    Article  CAS  Google Scholar 

  37. Redrobe JP, Bourin M (1998) Dose-dependent influence of buspirone on the activities of selective serotonin reuptake inhibitors in the mouse forced swimming test. Psychopharmacology 138:198–206

    Article  CAS  Google Scholar 

  38. Wong EH, Sonders MS, Amara SG, Tinholt PM, Piercey MF, Hoffmann WP et al (2000) Reboxetine: a pharmacologically potent, selective, and specific norepinephrine reuptake inhibitor. Biol Psychiatry 47:818–829

    Article  CAS  Google Scholar 

  39. Cryan JF, O’Leary OF, Jin SH, Friedland JC, Ouyang M, Hirsch BR et al (2004) Norepinephrine-deficient mice lack responses to antidepressant drugs, including selective serotonin reuptake inhibitors. Proc Natl Acad Sci U S A 101:8186–8891

    Article  CAS  Google Scholar 

  40. O’Leary OF, Bechtholt AJ, Crowley JJ, Hill TE, Page ME, Lucki I (2007) Depletion of serotonin and catecholamines block the acute behavioral response to different classes of antidepressant drugs in the mouse tail suspension test. Psychopharmacology 192:357–371

    Article  Google Scholar 

  41. Roni MA, Rahman S (2015) Effects of lobeline and reboxetine, fluoxetine, or bupropion combination on depression-like behaviors in mice. Pharmacol Biochem Behav 139(Pt A):1–6

    Article  CAS  Google Scholar 

  42. Artigas F, Romero L, de Montigny C, Blier P (1996) Acceleration of the effect of selected antidepressant drugs in major depression by 5-HT1A antagonists. Trends Neurosci 19:378–383

    Article  CAS  Google Scholar 

  43. Hervás I, Artigas F (1998) Effect of fluoxetine on extracellular 5-hydroxytryptamine in rat brain. Role of 5-HT autoreceptors. Eur J Pharmacol 358:9–18

    Article  Google Scholar 

  44. Mateo Y, Pineda J, Meana JJ (1998) Somatodendritic alpha2-adrenoceptors in the locus coeruleus are involved in the in vivo modulation of cortical noradrenaline release by the antidepressant desipramine. J Neurochem 71:790–798

    Article  CAS  Google Scholar 

  45. Ortega JE, Fernández-Pastor B, Callado LF, Meana JJ (2010) In vivo potentiation of reboxetine and citalopram effect on extracellular noradrenaline in rat brain by α2-adrenoceptor antagonism. Eur Neuropsychopharmacol 20:813–822

    Article  CAS  Google Scholar 

  46. Washburn CP, Sirois JE, Talley EM, Guyenet PG, Bayliss DA (2002) Serotonergic raphe neurons express TASK channel transcripts and a TASK-like pH- and halothane-sensitive K+ conductance. J Neurosci 22:1256–1265

    Article  CAS  Google Scholar 

  47. Gordon JA, Hen R (2006) TREKing toward new antidepressants. Nat Neurosci 9:1081–1083

    Article  CAS  Google Scholar 

  48. Mazella J, Pétrault O, Lucas G, Deval E, Béraud-Dufour S, Gandin C, el-Yacoubi M, Widmann C et al (2010) Spadin, a sortilin-derived peptide, targeting rodent TREK-1 channels: a new concept in the antidepressant drug design. PLoS Biol 8:e1000355

    Article  Google Scholar 

  49. Kennard LE, Chumbley JR, Ranatunga KM, Armstrong SJ, Veale EL, Mathie A (2005) Inhibition of the human two-pore domain potassium channel, TREK-1, by fluoxetine and its metabolite norfluoxetine. Br J Pharmacol 144:821–829

    Article  CAS  Google Scholar 

  50. Sandoz G, Bell SC, Isacoff EY (2011) Optical probing of a dynamic membrane interaction that regulates the TREK1 channel. Proc Natl Acad Sci U S A 108:2605–2610

    Article  CAS  Google Scholar 

  51. Hajdu P, Ulens C, Panyi G, Tytgat J (2003) Drug- and mutagenesis-induced changes in the selectivity filter of a cardiac two-pore background K+ channel. Cardiovasc Res 58:46–54

    Article  CAS  Google Scholar 

  52. Stockmeier CA, Shapiro LA, Dilley GE, Kolli TN, Friedman L, Rajkowska G (1998) Increase in serotonin-1A autoreceptors in the midbrain of suicide victims with major depression-postmortem evidence for decreased serotonin activity. J Neurosci 18:7394–7401

    Article  CAS  Google Scholar 

  53. Lemonde S, Turecki G, Bakish D, Du L, Hrdina PD, Bown CD et al (2003) Impaired repression at a 5-hydroxytryptamine 1A receptor gene polymorphism associated with major depression and suicide. J Neurosci 23:8788–8799

    Article  CAS  Google Scholar 

  54. Lemonde S, Du L, Bakish D, Hrdina P, Albert PR (2004) Association of the C(-1019)G 5-HT1A functional promoter polymorphism with antidepressant response. Int J Neuropsychopharmacol 7:501–506

    Article  CAS  Google Scholar 

  55. Neff CD, Abkevich V, Packer JC, Chen Y, Potter J, Riley R et al (2009) Evidence for HTR1A and LHPP as interacting genetic risk factors in major depression. Mol Psychiatry 14:621–630

    Article  CAS  Google Scholar 

  56. Richardson-Jones JW, Craige CP, Guiard BP, Stephen A, Metzger KL, Kung HF, Gardier AM, Dranovsky A et al (2010) 5-HT1A autoreceptor levels determine vulnerability to stress and response to antidepressants. Neuron 65:40–52

    Article  CAS  Google Scholar 

  57. Nibuya M, Morinobu S, Duman RS (1995) Regulation of BDNF and trkB mRNA in rat brain by chronic electroconvulsive seizure and antidepressant drug treatments. J Neurosci 15:7539–7547

    Article  CAS  Google Scholar 

  58. Pei Q, Zetterström TS, Sprakes M, Tordera R, Sharp T (2003) Antidepressant drug treatment induces Arc gene expression in the rat brain. Neuroscience 121:975–982

    Article  CAS  Google Scholar 

  59. David DJ, Samuels BA, Rainer Q, Wang JW, Marsteller D, Mendez I, Drew M, Craig DA et al (2009) Neurogenesis-dependent and -independent effects of fluoxetine in an animal model of anxiety/depression. Neuron 62:479–493

    Article  CAS  Google Scholar 

  60. Dranovsky A, Hen R (2006) Hippocampal neurogenesis: regulation by stress and antidepressants. Biol Psychiatry 59:1136–1143

    Article  CAS  Google Scholar 

  61. Page ME (2003) The promises and pitfalls of reboxetine. CNS Drug Rev 9:327–342

    Article  CAS  Google Scholar 

  62. Sanchez C, Reines EH, Montgomery SA (2014) A comparative review of escitalopram, paroxetine, and sertraline: Are they all alike? Int Clin Psychopharmacol 29:185–196

    Article  Google Scholar 

Download references

Acknowledgements

We thank María Calvo, Elisenda Coll, and Anna Bosch for outstanding technical support in the confocal microscopy unit (CCiT-UB); and Mireia Galofré and Letizia Campa for their outstanding technical assistance. We also thank J Pablo Salvador and Núria Pascual for the TASK3 antibody production and purification (Institut de Quimica Avançada de Catalunya, CSIC; Parc Cientific de Barcelona, UB; and CIBER in Bioengineering, Biomaterials, and Nanomedicine), and to Nlife Therapeutics S.L. for advice on the design of conjugated siRNA molecules.

Funding

This work was supported by the following grants: SAF2015-68346-P (F.A.); SAF2013-48586-R (J.M.); SAF2016-75797-R (A.B.); Retos-Colaboración Subprograms RTC-2014-2812-1 and RTC-2015-3309-1 (A.B.); Ministry of Economy and Competitiveness (MINECO)—European Regional Development Fund (ERDF), UE; PI13/01390, Instituto de Salud Carlos III co-financed by ERDF (A.B.); IT616-13 Basque Government—ERDF (J.M.); 20003 NARSAD Independent Investigator (A.B.); and Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM). CERCA Programme/Generalitat de Catalunya is also acknowledged. M.N.F. and A.F-C. are recipients of a fellowship from the Spanish Ministry of Education, Culture and Sport.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Analia Bortolozzi.

Ethics declarations

Conflict of Interest

F.A. has received consulting honoraria on antidepressant drugs from Lundbeck and he has been PI of grants from Lundbeck. A.B. has been PI of grants from Nlife Therapeutics. S.L., F.A., and A.B. are coauthors of the patent WO/2011/131693 for the siRNA and ASO (antisense oligonucleotides) molecules and the targeting approach related to this work. The rest of authors declare no competing financial interest.

Electronic Supplementary Material

ESM 1

(DOCX 2522 kb)

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Fullana, M.N., Ferrés-Coy, A., Ortega, J.E. et al. Selective Knockdown of TASK3 Potassium Channel in Monoamine Neurons: a New Therapeutic Approach for Depression. Mol Neurobiol 56, 3038–3052 (2019). https://doi.org/10.1007/s12035-018-1288-1

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s12035-018-1288-1

Keywords

Navigation