#PAGE_PARAMS# #ADS_HEAD_SCRIPTS# #MICRODATA#

Association of FKBP51 with Priming of Autophagy Pathways and Mediation of Antidepressant Treatment Response: Evidence in Cells, Mice, and Humans


Background:
FK506 binding protein 51 (FKBP51) is an Hsp90 co-chaperone and regulator of the glucocorticoid receptor, and consequently of stress physiology. Clinical studies suggest a genetic link between FKBP51 and antidepressant response in mood disorders; however, the underlying mechanisms remain elusive. The objective of this study was to elucidate the role of FKBP51 in the actions of antidepressants, with a particular focus on pathways of autophagy.

Methods and Findings:
Established cell lines, primary neural cells, human blood cells of healthy individuals and patients with depression, and mice were treated with antidepressants. Mice were tested for several neuroendocrine and behavioral parameters. Protein interactions and autophagic pathway activity were mainly evaluated by co-immunoprecipitation and Western blots. We first show that the effects of acute antidepressant treatment on behavior are abolished in FKBP51 knockout (51KO) mice. Autophagic markers, such as the autophagy initiator Beclin1, were increased following acute antidepressant treatment in brains from wild-type, but not 51KO, animals. FKBP51 binds to Beclin1, changes decisive protein interactions and phosphorylation of Beclin1, and triggers autophagic pathways. Antidepressants and FKBP51 exhibited synergistic effects on these pathways. Using chronic social defeat as a depression-relevant stress model in combination with chronic paroxetine (PAR) treatment revealed that the stress response, as well as the effects of antidepressants on behavior and autophagic markers, depends on FKBP51. In human blood cells of healthy individuals, FKBP51 levels correlated with the potential of antidepressants to induce autophagic pathways.

Importantly, the clinical antidepressant response of patients with depression (n = 51) could be predicted by the antidepressant response of autophagic markers in patient-derived peripheral blood lymphocytes cultivated and treated ex vivo (Beclin1/amitriptyline: r = 0.572, p = 0.003; Beclin1/PAR: r = 0.569, p = 0.004; Beclin1/fluoxetine: r = 0.454, p = 0.026; pAkt/amitriptyline: r = −0.416, p = 0.006; pAkt/PAR: r = −0.355, p = 0.021; LC3B-II/PAR: r = 0.453, p = 0.02), as well as by the lymphocytic expression levels of FKBP51 (r = 0.631, p<0.0001), pAkt (r = −0.515, p = 0.003), and Beclin1 (r = 0.521, p = 0.002) at admission. Limitations of the study include the use of male mice only and the relatively low number of patients for protein analyses.

Conclusions:
To our knowledge, these findings provide the first evidence for the molecular mechanism of FKBP51 in priming autophagic pathways; this process is linked to the potency of at least some antidepressants. These newly discovered functions of FKBP51 also provide novel predictive markers for treatment outcome, consistent with physiological and potential clinical relevance.

Please see later in the article for the Editors' Summary


Vyšlo v časopise: Association of FKBP51 with Priming of Autophagy Pathways and Mediation of Antidepressant Treatment Response: Evidence in Cells, Mice, and Humans. PLoS Med 11(11): e32767. doi:10.1371/journal.pmed.1001755
Kategorie: Research Article
prolekare.web.journal.doi_sk: https://doi.org/10.1371/journal.pmed.1001755

Souhrn

Background:
FK506 binding protein 51 (FKBP51) is an Hsp90 co-chaperone and regulator of the glucocorticoid receptor, and consequently of stress physiology. Clinical studies suggest a genetic link between FKBP51 and antidepressant response in mood disorders; however, the underlying mechanisms remain elusive. The objective of this study was to elucidate the role of FKBP51 in the actions of antidepressants, with a particular focus on pathways of autophagy.

Methods and Findings:
Established cell lines, primary neural cells, human blood cells of healthy individuals and patients with depression, and mice were treated with antidepressants. Mice were tested for several neuroendocrine and behavioral parameters. Protein interactions and autophagic pathway activity were mainly evaluated by co-immunoprecipitation and Western blots. We first show that the effects of acute antidepressant treatment on behavior are abolished in FKBP51 knockout (51KO) mice. Autophagic markers, such as the autophagy initiator Beclin1, were increased following acute antidepressant treatment in brains from wild-type, but not 51KO, animals. FKBP51 binds to Beclin1, changes decisive protein interactions and phosphorylation of Beclin1, and triggers autophagic pathways. Antidepressants and FKBP51 exhibited synergistic effects on these pathways. Using chronic social defeat as a depression-relevant stress model in combination with chronic paroxetine (PAR) treatment revealed that the stress response, as well as the effects of antidepressants on behavior and autophagic markers, depends on FKBP51. In human blood cells of healthy individuals, FKBP51 levels correlated with the potential of antidepressants to induce autophagic pathways.

Importantly, the clinical antidepressant response of patients with depression (n = 51) could be predicted by the antidepressant response of autophagic markers in patient-derived peripheral blood lymphocytes cultivated and treated ex vivo (Beclin1/amitriptyline: r = 0.572, p = 0.003; Beclin1/PAR: r = 0.569, p = 0.004; Beclin1/fluoxetine: r = 0.454, p = 0.026; pAkt/amitriptyline: r = −0.416, p = 0.006; pAkt/PAR: r = −0.355, p = 0.021; LC3B-II/PAR: r = 0.453, p = 0.02), as well as by the lymphocytic expression levels of FKBP51 (r = 0.631, p<0.0001), pAkt (r = −0.515, p = 0.003), and Beclin1 (r = 0.521, p = 0.002) at admission. Limitations of the study include the use of male mice only and the relatively low number of patients for protein analyses.

Conclusions:
To our knowledge, these findings provide the first evidence for the molecular mechanism of FKBP51 in priming autophagic pathways; this process is linked to the potency of at least some antidepressants. These newly discovered functions of FKBP51 also provide novel predictive markers for treatment outcome, consistent with physiological and potential clinical relevance.

Please see later in the article for the Editors' Summary


Zdroje

1. KesslerRC, Aguilar-GaxiolaS, AlonsoJ, ChatterjiS, LeeS, et al. (2009) The WHO World Mental Health (WMH) Surveys. Psychiatrie (Stuttg) 6: 5–9.

2. GeddesJR, MiklowitzDJ (2013) Treatment of bipolar disorder. Lancet 381: 1672–1682.

3. KlengelT, MehtaD, AnackerC, Rex-HaffnerM, PruessnerJC, et al. (2013) Allele-specific FKBP5 DNA demethylation mediates gene-childhood trauma interactions. Nat Neurosci 16: 33–41.

4. AttwoodBK, BourgognonJM, PatelS, MuchaM, SchiavonE, et al. (2011) Neuropsin cleaves EphB2 in the amygdala to control anxiety. Nature 473: 372–375.

5. HartmannJ, WagnerKV, LieblC, ScharfSH, WangXD, et al. (2012) The involvement of FK506-binding protein 51 (FKBP5) in the behavioral and neuroendocrine effects of chronic social defeat stress. Neuropharmacology 62: 332–339.

6. ToumaC, GassenNC, HerrmannL, Cheung-FlynnJ, BullDR, et al. (2011) FK506 binding protein 5 shapes stress responsiveness: modulation of neuroendocrine reactivity and coping behavior. Biol Psychiatry 70: 928–936.

7. WochnikGM, RüeggJ, AbelGA, SchmidtU, HolsboerF, et al. (2005) FK506-binding proteins 51 and 52 differentially regulate dynein interaction and nuclear translocation of the glucocorticoid receptor in mammalian cells. J Biol Chem 280: 4609–4616.

8. BinderEB, SalyakinaD, LichtnerP, WochnikGM, IsingM, et al. (2004) Polymorphisms in FKBP5 are associated with increased recurrence of depressive episodes and rapid response to antidepressant treatment. Nat Genet 36: 1319–1325.

9. LekmanM, LajeG, CharneyD, RushAJ, WilsonAF, et al. (2008) The FKBP5-gene in depression and treatment response—an association study in the Sequenced Treatment Alternatives to Relieve Depression (STAR*D) Cohort. Biol Psychiatry 63: 1103–1110.

10. ZouYF, WangF, FengXL, LiWF, TaoJH, et al. (2010) Meta-analysis of FKBP5 gene polymorphisms association with treatment response in patients with mood disorders. Neurosci Lett 484: 56–61.

11. LajeG, PerlisRH, RushAJ, McMahonFJ (2009) Pharmacogenetics studies in STAR*D: strengths, limitations, and results. Psychiatr Serv 60: 1446–1457.

12. De KloetER, JoelsM, HolsboerF (2005) Stress and the brain: from adaptation to disease. Nat Rev Neurosci 6: 463–475.

13. HolsboerF (2000) The corticosteroid receptor hypothesis of depression. Neuropsychopharmacology 23: 477–501.

14. ZschockeJ, ZimmermannN, BerningB, GanalV, HolsboerF, et al. (2011) Antidepressant drugs diversely affect autophagy pathways in astrocytes and neurons—dissociation from cholesterol homeostasis. Neuropsychopharmacology 36: 1754–1768.

15. RossiM, MunarrizER, BartesaghiS, MilaneseM, DinsdaleD, et al. (2009) Desmethylclomipramine induces the accumulation of autophagy markers by blocking autophagic flux. J Cell Sci 122: 3330–3339.

16. TaipaleM, JaroszDF, LindquistS (2010) HSP90 at the hub of protein homeostasis: emerging mechanistic insights. Nat Rev Mol Cell Biol 11: 515–528.

17. MizushimaN, KomatsuM (2011) Autophagy: renovation of cells and tissues. Cell 147: 728–741.

18. NakatogawaH, SuzukiK, KamadaY, OhsumiY (2009) Dynamics and diversity in autophagy mechanisms: lessons from yeast. Nat Rev Mol Cell Biol 10: 458–467.

19. WangRC, WeiY, AnZ, ZouZ, XiaoG, et al. (2012) Akt-mediated regulation of autophagy and tumorigenesis through Beclin 1 phosphorylation. Science 338: 956–959.

20. HarrisH, RubinszteinDC (2012) Control of autophagy as a therapy for neurodegenerative disease. Nat Rev Neurol 8: 108–117.

21. LevineB, KroemerG (2008) Autophagy in the pathogenesis of disease. Cell 132: 27–42.

22. WelbergL (2012) Neurotransmission: autophagy regulates transmission. Nat Rev Neurosci 13: 362–363.

23. CaiQ, LuL, TianJH, ZhuYB, QiaoH, et al. (2010) Snapin-regulated late endosomal transport is critical for efficient autophagy-lysosomal function in neurons. Neuron 68: 73–86.

24. MaJ, HouLN, RongZX, LiangP, FangC, et al. (2013) Antidepressant desipramine leads to C6 glioma cell autophagy: implication for the adjuvant therapy of cancer. Anticancer Agents Med Chem 13: 254–260.

25. ChenJ, KorostyshevskyD, LeeS, PerlsteinEO (2012) Accumulation of an antidepressant in vesiculogenic membranes of yeast cells triggers autophagy. PLoS ONE 7: e34024.

26. KaraNZ, TokerL, AgamG, AndersonGW, BelmakerRH, et al. (2013) Trehalose induced antidepressant-like effects and autophagy enhancement in mice. Psychopharmacology (Berl) 229: 367–375.

27. ClearyC, LindeJA, HiscockKM, HadasI, BelmakerRH, et al. (2008) Antidepressive-like effects of rapamycin in animal models: implications for mTOR inhibition as a new target for treatment of affective disorders. Brain Res Bull 76: 469–473.

28. NestlerEJ, HymanSE (2010) Animal models of neuropsychiatric disorders. Nat Neurosci 13: 1161–1169.

29. DeussingJM (2006) Animal models of depression. Drug Discov Today 3: 375–383.

30. WangXD, ChenY, WolfM, WagnerKV, LieblC, et al. (2011) Forebrain CRHR1 deficiency attenuates chronic stress-induced cognitive deficits and dendritic remodeling. Neurobiol Dis 42: 300–310.

31. BertonO, McClungCA, DileoneRJ, KrishnanV, RenthalW, et al. (2006) Essential role of BDNF in the mesolimbic dopamine pathway in social defeat stress. Science 311: 864–868.

32. SkeenJE, BhaskarPT, ChenCC, ChenWS, PengXD, et al. (2006) Akt deficiency impairs normal cell proliferation and suppresses oncogenesis in a p53-independent and mTORC1-dependent manner. Cancer Cell 10: 269–280.

33. PerisicT, ZimmermannN, KirmeierT, AsmusM, TuortoF, et al. (2010) Valproate and amitriptyline exert common and divergent influences on global and gene promoter-specific chromatin modifications in rat primary astrocytes. Neuropsychopharmacology 35: 792–805.

34. FrankeB, FigielM, EngeleJ (1998) CNS glia are targets for GDNF and neurturin. Histochem Cell Biol 110: 595–601.

35. SchumannBG, JutziP, RoditiI (2011) Genome-wide RNAi screens in bloodstream form trypanosomes identify drug transporters. Mol Biochem Parasitol 175: 91–94.

36. KimD, SunM, HeL, ZhouQH, ChenJ, et al. (2010) A small molecule inhibits Akt through direct binding to Akt and preventing Akt membrane translocation. J Biol Chem 285: 8383–8394.

37. BrunetA, BonniA, ZigmondMJ, LinMZ, JuoP, et al. (1999) Akt promotes cell survival by phosphorylating and inhibiting a Forkhead transcription factor. Cell 96: 857–868.

38. KabeyaY, MizushimaN, UenoT, YamamotoA, KirisakoT, et al. (2000) LC3, a mammalian homologue of yeast Apg8p, is localized in autophagosome membranes after processing. EMBO J 19: 5720–5728.

39. WagnerKV, MarinescuD, HartmannJ, WangXD, LabermaierC, et al. (2012) Differences in FKBP51 regulation following chronic social defeat stress correlate with individual stress sensitivity: influence of paroxetine treatment. Neuropsychopharmacology 37: 2797–2808.

40. WagnerKV, WangXD, LieblC, ScharfSH, MüllerMB, et al. (2011) Pituitary glucocorticoid receptor deletion reduces vulnerability to chronic stress. Psychoneuroendocrinology 36: 579–587.

41. McIlwainKL, MerriweatherMY, Yuva-PaylorLA, PaylorR (2001) The use of behavioral test batteries: effects of training history. Physiol Behav 73: 705–717.

42. GoldenSA, CovingtonHE3rd, BertonO, RussoSJ (2011) A standardized protocol for repeated social defeat stress in mice. Nat Protoc 6: 1183–1191.

43. FluttertM, DalmS, OitzlMS (2000) A refined method for sequential blood sampling by tail incision in rats. Lab Anim 34: 372–378.

44. UhrM, StecklerT, YassouridisA, HolsboerF (2000) Penetration of amitriptyline, but not of fluoxetine, into brain is enhanced in mice with blood-brain barrier deficiency due to mdr1a P-glycoprotein gene disruption. Neuropsychopharmacology 22: 380–387.

45. UhrM, GrauerMT, HolsboerF (2003) Differential enhancement of antidepressant penetration into the brain in mice with abcb1ab (mdr1ab) P-glycoprotein gene disruption. Biol Psychiatry 54: 840–846.

46. HenningsJM, OwashiT, BinderEB, HorstmannS, MenkeA, et al. (2009) Clinical characteristics and treatment outcome in a representative sample of depressed inpatients—findings from the Munich Antidepressant Response Signature (MARS) project. J Psychiatr Res 43: 215–229.

47. American Psychiatric Association (1994) Diagnostic and statistical manual of mental disorders, 4th edition. Washington (District of Columbia): American Psychiatric Association.

48. OverallJE, RhoadesHM (1982) Use of the Hamilton Rating Scale for classification of depressive disorders. Compr Psychiatry 23: 370–376.

49. HiemkeC, BaumannP, BergemannN, ConcaA, DietmaierO, et al. (2011) AGNP consensus guidelines for therapeutic drug monitoring in psychiatry: update 2011. Pharmacopsychiatry 44: 195–235.

50. TaksandeBG, KotagaleNR, TripathiSJ, UgaleRR, ChopdeCT (2009) Antidepressant like effect of selective serotonin reuptake inhibitors involve modulation of imidazoline receptors by agmatine. Neuropharmacology 57: 415–424.

51. ThoeringerCK, ErhardtA, SillaberI, MuellerMB, OhlF, et al. (2010) Long-term anxiolytic and antidepressant-like behavioural effects of tiagabine, a selective GABA transporter-1 (GAT-1) inhibitor, coincide with a decrease in HPA system activity in C57BL/6 mice. J Psychopharmacol 24: 733–743.

52. GaleottiN, GhelardiniC (2012) Selective modulation of the PKCvarepsilon/p38MAP kinase signalling pathway for the antidepressant-like activity of amitriptyline. Neuropharmacology 62: 289–296.

53. PeiH, LiL, FridleyBL, JenkinsGD, KalariKR, et al. (2009) FKBP51 affects cancer cell response to chemotherapy by negatively regulating Akt. Cancer Cell 16: 259–266.

54. KlionskyDJ, AbdallaFC, AbeliovichH, AbrahamRT, Acevedo-ArozenaA, et al. (2012) Guidelines for the use and interpretation of assays for monitoring autophagy. Autophagy 8: 445–544.

55. JeonSH, KimSH, KimY, KimYS, LimY, et al. (2011) The tricyclic antidepressant imipramine induces autophagic cell death in U-87MG glioma cells. Biochem Biophys Res Commun 413: 311–317.

56. HuangW, ZhaoY, ZhuX, CaiZ, WangS, et al. (2013) Fluoxetine upregulates phosphorylated-AKT and phosphorylated-ERK1/2 proteins in neural stem cells: evidence for a crosstalk between AKT and ERK1/2 pathways. J Mol Neurosci 49: 244–249.

57. LiN, LeeB, LiuRJ, BanasrM, DwyerJM, et al. (2010) mTOR-dependent synapse formation underlies the rapid antidepressant effects of NMDA antagonists. Science 329: 959–964.

58. VoletiB, NavarriaA, LiuRJ, BanasrM, LiN, et al. (2013) Scopolamine rapidly increases mammalian target of rapamycin complex 1 signaling, synaptogenesis, and antidepressant behavioral responses. Biol Psychiatry 74: 742–749.

59. HublerTR, ScammellJG (2004) Intronic hormone response elements mediate regulation of FKBP5 by progestins and glucocorticoids. Cell Stress Chaperones 9: 243–252.

60. LaaneE, TammKP, BuentkeE, ItoK, KharazihaP, et al. (2009) Cell death induced by dexamethasone in lymphoid leukemia is mediated through initiation of autophagy. Cell Death Differ 16: 1018–1029.

61. ScammellJG, DennyWB, ValentineDL, SmithDF (2001) Overexpression of the FK506-binding immunophilin FKBP51 is the common cause of glucocorticoid resistance in three New World primates. Gen Comp Endocrinol 124: 152–165.

62. BhuiyanMS, TagashiraH, FukunagaK (2011) Sigma-1 receptor stimulation with fluvoxamine activates Akt-eNOS signaling in the thoracic aorta of ovariectomized rats with abdominal aortic banding. Eur J Pharmacol 650: 621–628.

63. Basta-KaimA, BudziszewskaB, Jaworska-FeilL, TetichM, KuberaM, et al. (2005) Inhibitory effect of imipramine on the human corticotropin-releasing-hormone gene promoter activity operates through a PI3-K/AKT mediated pathway. Neuropharmacology 49: 156–164.

64. ManningBD, CantleyLC (2007) AKT/PKB signaling: navigating downstream. Cell 129: 1261–1274.

65. BertonO, NestlerEJ (2006) New approaches to antidepressant drug discovery: beyond monoamines. Nat Rev Neurosci 7: 137–151.

66. HernandezD, TorresCA, SetlikW, CebrianC, MosharovEV, et al. (2012) Regulation of presynaptic neurotransmission by macroautophagy. Neuron 74: 277–284.

67. KimCS, ChangPY, JohnstonD (2012) Enhancement of dorsal hippocampal activity by knockdown of HCN1 channels leads to anxiolytic- and antidepressant-like behaviors. Neuron 75: 503–516.

68. O'LearyJC3rd, DhariaS, BlairLJ, BradyS, JohnsonAG, et al. (2011) A new anti-depressive strategy for the elderly: ablation of FKBP5/FKBP51. PLoS ONE 6: e24840.

69. JinwalUK, KorenJ3rd, BorysovSI, SchmidAB, AbisambraJF, et al. (2010) The Hsp90 cochaperone, FKBP51, increases Tau stability and polymerizes microtubules. J Neurosci 30: 591–599.

70. BarabasiAL, GulbahceN, LoscalzoJ (2011) Network medicine: a network-based approach to human disease. Nat Rev Genet 12: 56–68.

71. LuoL, CallawayEM, SvobodaK (2008) Genetic dissection of neural circuits. Neuron 57: 634–660.

Štítky
Interné lekárstvo

Článok vyšiel v časopise

PLOS Medicine


2014 Číslo 11
Najčítanejšie tento týždeň
Najčítanejšie v tomto čísle
Kurzy

Zvýšte si kvalifikáciu online z pohodlia domova

Získaná hemofilie - Povědomí o nemoci a její diagnostika
nový kurz

Eozinofilní granulomatóza s polyangiitidou
Autori: doc. MUDr. Martina Doubková, Ph.D.

Všetky kurzy
Prihlásenie
Zabudnuté heslo

Zadajte e-mailovú adresu, s ktorou ste vytvárali účet. Budú Vám na ňu zasielané informácie k nastaveniu nového hesla.

Prihlásenie

Nemáte účet?  Registrujte sa

#ADS_BOTTOM_SCRIPTS#