Skip to main content

Advertisement

Log in

Recent Research Progress on the Antidepressant-like Effect and Neuropharmacological Potential of Gastrodia elata Blume

  • Natural Products: From Chemistry to Pharmacology (C Ho, Section Editor)
  • Published:
Current Pharmacology Reports Aims and scope Submit manuscript

Abstract

Purpose of Review

Depression is a difficult and currently incurable mood disorder and the conventional therapies used as treatment are unsatisfactory nowadays. Therefore, there is an urgent demand to find alternative or complementary approaches to deal with depression. Gastrodia elata Blume (GE) is not only a traditional Chinese medicine but also a food material that has been widely used in treating numerous neurological disorders in East Asia, including migraine, seizure, and mood disorders. In the present review article, we summarized the antidepressant-like effects of GE that have been reported in animal studies and elucidate its cellular mechanisms.

Recent Findings

GE and its active compounds, gastrodin, 4-hydroxybenzyl alcohol, vanillin, and their derivatives have recently been reported to have intensive antidepressant-like effects in several animal models of depression, such as the forced swim test, the tail suspension test, and the chronic mild stress. The molecular mechanisms of GE-induced antidepressant-like effect are likely through regulation of neurotransmitters, antioxidation, antiinflammation, modulation of hypothalamic-pituitary-adrenal axis, neurotrophic effects, regulation of stem cell, and enhancement of neuroplasticity and neuroprotection.

Summary

GE and its bioactive components have been shown to possess antidepressant-like effects in several well-conducted animal studies. Additionally, GE has been applied to manipulate mood disorders in Chinese medicine for many years. However, more accurate and specific initial targeting of the mechanism of action of GE, as well as the application of clinical trials, are warranted and deserve further investigation.

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

Similar content being viewed by others

Abbreviations

AD:

Alzheimer’s disease

Akt:

Protein kinase B

AP-1:

Activator protein-1

B2:

N(6)-(3-methoxyl-4-hydroxybenzyl) adenine riboside

Bax:

Bcl-2-associated X

BBB:

Blood-brain barrier

BDNF:

Brain-derived neurotrophic factor

BrdU:

Bromodeoxyuridine

Clic4:

Chloride intracellular channel protein 4

COX-2:

Cyclooxygenase-2

CREB:

cAMP responsive element binding protein

CSDS:

Chronic social defeat stress

DA:

Dopamine

Dbnl:

Drebrin-like protein

DCX:

Doublecortin

DG:

Dentate gyrus

DOPAC:

3,4-Dihydroxyphenylacetic acid

DPPH:

1,1-Diphenyl-2-picrylhydrazyl

DPYL2 (CRMP2):

Dihydropyrimidinase-related protein 2

ERK1/2:

Extracellular regulated protein kinases 1/2

FST:

Forced swim test

GABA:

γ-Aminobutyric acid

GABA-T:

GABA transaminase

GAD:

Glutamate decarboxylase

GCLM:

Glutamate-cysteine ligase regulatory subunit

GDNF:

Glial cell line-derived neurotrophic factor

GE:

Gastrodia elata Blume

GFAP:

Glial fibrillary acidic protein

GSH:

Glutathione

GSHPx:

Glutathione peroxidase

GSHRx:

Glutathione reductase

GSSG:

Glutathione disulfide

H2O2 :

Hydrogen peroxide

HD:

Huntington’s disease

HO-1:

Hemeoxygenase-1

HPA:

Hypothalamic-pituitary-adrenal

Hsp90aa1:

Heat shock protein 90

HVA:

Homovanillic acid

i.p.:

Intraperitoneally

IKK:

IκB kinase

IL:

Interleukin

iNOS:

Inducible nitric oxide synthase

iTRAQ:

Isobaric tags for relative and absolute quantitation

JNK:

c-Jun NH2-terminal kinase

KEAP-1:

Kelch-like ECH-associated protein 1

L-DOPA:

Levodopa

LPS:

Lipopolysaccharide

MAO:

Monoamine oxidase

MAOIs:

Monoamine oxidase inhibitors

MAP2:

Microtubule associated protein 2

MAPK:

Mitogen-activated protein kinase

MDA:

Malondialdehyde

MDD:

Major depressive disorder

MEK:

Mitogen-activated protein kinase kinase

Mobkl3:

Mps one binder kinase activator-like 3

MPP+ :

Methyl-4-phenylpyridinium

NADPH:

Nicotinamide adenine dinucleotide phosphate

NCAM:

Neural cell adhesion molecule

NE:

Norepinephrine

NeuN:

Neuronal neuclei

NF-κB:

Nuclear factor κB

NMDAR:

N-methyl-D-aspartate receptor

NO:

Nitric oxide

NPY:

Neuropeptide Y

NQO1:

NAD(P)H: quinone oxidoreductase

Nrf2:

Nuclear factor E2-related factor 2

NSCs:

Neuronal stem cells

Nxn:

Nucleoredoxin

OFT:

Open field test

OGD/R:

Oxygen-glucose deprivation followed by reperfusion

PACSIN2:

Protein kinase C and casein kinase substrate in neurons protein 2

PC12:

Pheochromocytoma

PD:

Parkinson’s disease

PDI:

Protein disulfide isomerase

PDLIM1/5:

PDZ and LIM domain protein 1/5

PFN1:

Profilin 1

PI3K:

Phosphatidylinositol-3-kinase

PKA:

Protein kinase A

Ppia:

Peptidyl-prolyl cis-trans isomerase A

Prdx6:

Peroxiredoxin 6

Rho A:

Ras homologous member A

ROS:

Reactive oxygen species

RTN1/4:

Reticulon 1/4

Sept2:

Septin-2

sMaf:

Small Maf

SOD:

Superoxide dismutase

SOX2:

Sex-determing region Y-box 2

SPS:

Single prolonged stress

SS(N)RIs:

Selective serotonin (norepinephrine) reuptake inhibitors

SSA:

Succinic semialdehyde

TCAs:

Tricyclic antidepressants

TCM:

Traditional Chinese medicine

TH:

Tyrosine hydroxylase

TLR:

Toll-like receptor

TNF-α:

Tumor necrosis factor-alpha

TPH:

Typtophan hydroxylase

Tpi1:

Triosephosphate isomerase

TrkB:

Tropomyosin-related kinase B

TST:

Tail suspension test

Tuj-1:

Neuron-specific class III beta-tubulin

Uchl1:

Ubiquitin carboxyl-terminal hydrolase isozyme L1

(U)CMS:

(Unpredictable) chronic mild stress

WGE:

Water extract of Gastrodia elata Blume

WHO:

World Health Organization

4-HBA:

4-Hydroxybenzyl alcohol

4-HBAD:

4-Hydroxybenzaldehyde

5-HIAA:

5-Hydroxyindoleacetic acid

5-HT:

Serotonin

5-HTP:

5-Hydroxytryptophan

References

Papers of particular interest, published recently, have been highlighted as: • Of importance •• Of major importance

  1. Mathers CD, Lopez AD, Murray CJL. The burden of disease and mortality by condition: data, methods, and results for 2001. In: Lopez AD, Mathers CD, Ezzati M, Jamison DT, Murray CJL, editors. Global burden of disease and risk factors. Washington (DC); 2006.

  2. Charlson FJ, Ferrari AJ, Flaxman AD, Whiteford HA. The epidemiological modelling of dysthymia: application for the Global Burden of Disease Study 2010. J Affect Disord. 2013;151(1):111–20. https://doi.org/10.1016/j.jad.2013.05.060.

    Article  PubMed  Google Scholar 

  3. Kiyohara C, Yoshimasu K. Molecular epidemiology of major depressive disorder. Environ Health Prev Med. 2009;14(2):71–87. https://doi.org/10.1007/s12199-008-0073-6.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Kessler RC. The costs of depression. Psychiatr Clin North Am. 2012;35(1):1–14. https://doi.org/10.1016/j.psc.2011.11.005.

    Article  PubMed  Google Scholar 

  5. Ueda N, Yoshimura R, Shinkai K, Sakata Y, Nakamura J. Higher plasma 5-hydroxyindoleacetic acid levels are associated with SSRI-induced nausea. Neuropsychobiology. 2003;48(1):31–4. https://doi.org/10.1159/000071826.

    Article  CAS  PubMed  Google Scholar 

  6. Levinstein MR, Samuels BA. Mechanisms underlying the antidepressant response and treatment resistance. Front Behav Neurosci. 2014;8:208. https://doi.org/10.3389/fnbeh.2014.00208.

    Article  PubMed  PubMed Central  Google Scholar 

  7. Simon GE, Savarino J, Operskalski B, Wang PS. Suicide risk during antidepressant treatment. Am J Psychiatry. 2006;163(1):41–7. https://doi.org/10.1176/appi.ajp.163.1.41.

    Article  PubMed  Google Scholar 

  8. Brunoni AR, Teng CT, Correa C, Imamura M, Brasil-Neto JP, Boechat R, et al. Neuromodulation approaches for the treatment of major depression: challenges and recommendations from a working group meeting. Arq Neuropsiquiatr. 2010;68(3):433–51.

    Article  PubMed  Google Scholar 

  9. Nestler EJ. Antidepressant treatments in the 21st century. Biol Psychiatry. 1998;44(7):526–33.

    Article  CAS  PubMed  Google Scholar 

  10. Zarate CA Jr, Singh JB, Carlson PJ, Brutsche NE, Ameli R, Luckenbaugh DA, et al. A randomized trial of an N-methyl-D-aspartate antagonist in treatment-resistant major depression. Arch Gen Psychiatry. 2006;63(8):856–64. https://doi.org/10.1001/archpsyc.63.8.856.

    Article  CAS  PubMed  Google Scholar 

  11. •• Zhan HD, Zhou HY, Sui YP, Du XL, Wang WH, Dai L, et al. The rhizome of Gastrodia elata Blume—an ethnopharmacological review. J Ethnopharmacol. 2016;189:361–85. https://doi.org/10.1016/j.jep.2016.06.057. A very good review that includes the traditional usesages and the modern bioactive fuctions of Gastrodia elata Blume in a comprehensive way.

    Article  CAS  PubMed  Google Scholar 

  12. Chen PJ, Sheen LY. Gastrodiae Rhizoma (tian ma): a review of biological activity and antidepressant mechanisms. J Tradit Complement Med. 2011;1(1):31–40.

  13. Matias M, Silvestre S, Falcao A, Alves G. Gastrodia elata and epilepsy: rationale and therapeutic potential. Phytomedicine. 2016;23(12):1511–26. https://doi.org/10.1016/j.phymed.2016.09.001.

    Article  PubMed  Google Scholar 

  14. Jang JH, Son Y, Kang SS, Bae CS, Kim JC, Kim SH, et al. Neuropharmacological potential of Gastrodia elata Blume and its components. Evid Based Complement Alternat Med. 2015;2015:309261. https://doi.org/10.1155/2015/309261.

    Google Scholar 

  15. Kancheva VD, Kasaikina OT. Bio-antioxidants—a chemical base of their antioxidant activity and beneficial effect on human health. Curr Med Chem. 2013;20(37):4784–805.

    Article  CAS  PubMed  Google Scholar 

  16. Kim BW, Koppula S, Kim JW, Lim HW, Hwang JW, Kim IS, et al. Modulation of LPS-stimulated neuroinflammation in BV-2 microglia by Gastrodia elata: 4-hydroxybenzyl alcohol is the bioactive candidate. J Ethnopharmacol. 2012;139(2):549–57. https://doi.org/10.1016/j.jep.2011.11.048.

    Article  CAS  PubMed  Google Scholar 

  17. Lee JY, Jang YW, Kang HS, Moon H, Sim SS, Kim CJ. Anti-inflammatory action of phenolic compounds from Gastrodia elata root. Arch Pharm Res. 2006;29(10):849–58.

    Article  CAS  PubMed  Google Scholar 

  18. Han YJ, Je JH, Kim SH, Ahn SM, Kim HN, Kim YR, et al. Gastrodia elata shows neuroprotective effects via activation of PI3K signaling against oxidative glutamate toxicity in HT22 cells. Am J Chin Med. 2014;42(4):1007–19. https://doi.org/10.1142/S0192415X14500633.

    Article  PubMed  Google Scholar 

  19. Rosenfeldt F, Wilson M, Lee G, Kure C, Ou R, Braun L, et al. Oxidative stress in surgery in an ageing population: pathophysiology and therapy. Exp Gerontol. 2013;48(1):45–54. https://doi.org/10.1016/j.exger.2012.03.010.

    Article  CAS  PubMed  Google Scholar 

  20. Wigner P, Czarny P, Galecki P, Su KP, Sliwinski T. The molecular aspects of oxidative & nitrosative stress and the tryptophan catabolites pathway (TRYCATs) as potential causes of depression. Psychiatry Res. 2017;262:566–74. https://doi.org/10.1016/j.psychres.2017.09.045.

    Article  CAS  PubMed  Google Scholar 

  21. Jiang G, Hu Y, Liu L, Cai J, Peng C, Li Q. Gastrodin protects against MPP(+)-induced oxidative stress by up regulates heme oxygenase-1 expression through p38 MAPK/Nrf2 pathway in human dopaminergic cells. Neurochem Int. 2014;75:79–88. https://doi.org/10.1016/j.neuint.2014.06.003.

    Article  CAS  PubMed  Google Scholar 

  22. Li Q, Niu C, Zhang X, Dong M. Gastrodin and isorhynchophylline synergistically inhibit MPP(+)-induced oxidative stress in SH-SY5Y cells by targeting ERK1/2 and GSK-3beta pathways: involvement of Nrf2 nuclear translocation. ACS Chem Neurosci. 2017;9:482–93. https://doi.org/10.1021/acschemneuro.7b00247.

    Article  CAS  PubMed  Google Scholar 

  23. Dai JN, Zong Y, Zhong LM, Li YM, Zhang W, Bian LG, et al. Gastrodin inhibits expression of inducible NO synthase, cyclooxygenase-2 and proinflammatory cytokines in cultured LPS-stimulated microglia via MAPK pathways. PLoS One. 2011;6(7):e21891. https://doi.org/10.1371/journal.pone.0021891.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Bie X, Chen Y, Han J, Dai H, Wan H, Zhao T. Effects of gastrodin on amino acids after cerebral ischemia-reperfusion injury in rat striatum. Asia Pac J Clin Nutr. 2007;16(Suppl 1):305–8.

    CAS  PubMed  Google Scholar 

  25. Zeng X, Zhang S, Zhang L, Zhang K, Zheng X. A study of the neuroprotective effect of the phenolic glucoside gastrodin during cerebral ischemia in vivo and in vitro. Planta Med. 2006;72(15):1359–65. https://doi.org/10.1055/s-2006-951709.

    Article  CAS  PubMed  Google Scholar 

  26. Luo L, Kim SW, Lee HK, Kim ID, Lee H, Lee JK. Anti-oxidative effects of 4-hydroxybenzyl alcohol in astrocytes confer protective effects in autocrine and paracrine manners. PLoS One. 2017;12(5):e0177322. https://doi.org/10.1371/journal.pone.0177322.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Yu SS, Zhao J, Lei SP, Lin XM, Wang LL, Zhao Y. 4-hydroxybenzyl alcohol ameliorates cerebral injury in rats by antioxidant action. Neurochem Res. 2011;36(2):339–46. https://doi.org/10.1007/s11064-010-0335-8.

    Article  CAS  PubMed  Google Scholar 

  28. Luo L, Kim SW, Lee HK, Kim ID, Lee H, Lee JK. Anti-Zn2+−toxicity of 4-hydroxybenzyl alcohol in astrocytes and neurons contribute to a robust neuroprotective effects in the postischemic brain. Cell Mol Neurobiol. 2017;38:615–26. https://doi.org/10.1007/s10571-017-0508-y.

    Article  CAS  PubMed  Google Scholar 

  29. Yu S, Zhao J, Wang X, Lei S, Wu X, Chen Y, et al. 4-Hydroxybenzyl alcohol confers neuroprotection through up-regulation of antioxidant protein expression. Neurochem Res. 2013;38(7):1501–16. https://doi.org/10.1007/s11064-013-1052-x.

    Article  CAS  PubMed  Google Scholar 

  30. Yu SS, Zhao J, Zheng WP, Zhao Y. Neuroprotective effect of 4-hydroxybenzyl alcohol against transient focal cerebral ischemia via anti-apoptosis in rats. Brain Res. 2010;1308:167–75. https://doi.org/10.1016/j.brainres.2009.10.037.

    Article  CAS  PubMed  Google Scholar 

  31. Lee HE, Lee YW, Park SJ, Jeon SJ, Kim E, Lee S, et al. 4-Hydroxybenzyl methyl ether improves learning and memory in mice via the activation of dopamine D1 receptor signaling. Neurobiol Learn Mem. 2015;121:30–8. https://doi.org/10.1016/j.nlm.2015.03.004.

    Article  CAS  PubMed  Google Scholar 

  32. Wu LY, Chen WC, Tsai FS, Tsai CC, Wu CR, Lin LW. p-Hydroxybenzyl alcohol, an active phenolic ingredient of Gastrodia elata, reverses the cycloheximide-induced memory deficit by activating the adrenal gland in rats. Am J Chin Med. 2015;43(8):1593–604. https://doi.org/10.1142/S0192415X15500901.

    Article  CAS  PubMed  Google Scholar 

  33. Ju XH, Shi Y, Liu N, Guo DM, Cui X. Determination and pharmacokinetics of gastrodin in human plasma by HPLC coupled with photodiode array detector. J Chromatogr B Anal Technol Biomed Life Sci. 2010;878(22):1982–6. https://doi.org/10.1016/j.jchromb.2010.05.034.

    Article  CAS  Google Scholar 

  34. Jia Y, Li X, Xie H, Shen J, Luo J, Wang J, et al. Analysis and pharmacokinetics studies of gastrodin and p-hydroxybenzyl alcohol in dogs using ultra fast liquid chromatography-tandem mass spectrometry method. J Pharm Biomed Anal. 2014;99:83–8. https://doi.org/10.1016/j.jpba.2014.07.004.

    Article  CAS  PubMed  Google Scholar 

  35. Zhang W, Sheng YX, Zhang JL. Determination and pharmacokinetics of gastrodin and p-hydroxybenzylalcohol after oral administration of Gastrodia elata Bl. extract in rats by high-performance liquid chromatography-electrospray ionization mass spectrometric method. Phytomedicine. 2008;15(10):844–50. https://doi.org/10.1016/j.phymed.2008.02.012.

    Article  CAS  PubMed  Google Scholar 

  36. Lin LC, Chen YF, Tsai TR, Tsai TH. Analysis of brain distribution and biliary excretion of a nutrient supplement, gastrodin, in rat. Anal Chim Acta. 2007;590(2):173–9. https://doi.org/10.1016/j.aca.2007.03.035.

    Article  CAS  PubMed  Google Scholar 

  37. Wang Q, Chen G, Zeng S. Distribution and metabolism of gastrodin in rat brain. J Pharm Biomed Anal. 2008;46(2):399–404. https://doi.org/10.1016/j.jpba.2007.10.017.

    Article  CAS  PubMed  Google Scholar 

  38. • Wu J, Wu B, Tang C, Zhao J. Analytical techniques and pharmacokinetics of Gastrodia elata Blume and its constituents. Molecules. 2017;22(7) https://doi.org/10.3390/molecules22071137. A good review on the pharmokinetic and metabolomic aspects of the active components of Gastrodia elata Blume.

  39. Tai A, Sawano T, Yazama F. Antioxidant properties of ethyl vanillin in vitro and in vivo. Biosci Biotechnol Biochem. 2011;75(12):2346–50. https://doi.org/10.1271/bbb.110524.

    Article  CAS  PubMed  Google Scholar 

  40. Dhanalakshmi C, Janakiraman U, Manivasagam T, Justin Thenmozhi A, Essa MM, Kalandar A, et al. Vanillin attenuated behavioural impairments, neurochemical deficits, oxidative stress and apoptosis against rotenone induced rat model of Parkinson’s disease. Neurochem Res. 2016;41(8):1899–910. https://doi.org/10.1007/s11064-016-1901-5.

    Article  CAS  PubMed  Google Scholar 

  41. Kim IS, Choi DK, Jung HJ. Neuroprotective effects of vanillyl alcohol in Gastrodia elata Blume through suppression of oxidative stress and anti-apoptotic activity in toxin-induced dopaminergic MN9D cells. Molecules. 2011;16(7):5349–61. https://doi.org/10.3390/molecules16075349.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Dhanalakshmi C, Manivasagam T, Nataraj J, Justin Thenmozhi A, Essa MM. Neurosupportive role of vanillin, a natural phenolic compound, on rotenone induced neurotoxicity in SH-SY5Y neuroblastoma cells. Evid Based Complement Alternat Med. 2015;2015:626028. https://doi.org/10.1155/2015/626028.

    Article  PubMed  PubMed Central  Google Scholar 

  43. Kim YH, Park JH. Vanillin and 4-hydroxybenzyl alcohol attenuate cognitive impairment and the reduction of cell proliferation and neuroblast differentiation in the dentate gyrus in a mouse model of scopolamine-induced amnesia. Anat Cell Biol. 2017;50(2):143–51. https://doi.org/10.5115/acb.2017.50.2.143.

    Article  PubMed  PubMed Central  Google Scholar 

  44. Abuhamdah S, Thalji D, Abuirmeileh N, Bahnassi A, Salahat I, Abuirmeileh A. Behavioral and neurochemical alterations induced by vanillin in a mouse model of Alzheimer’s disease. Int J Pharmacol. 2017;13(6):573–82. https://doi.org/10.3923/ijp.2017.573.582.

    Article  Google Scholar 

  45. Nestler EJ, Hyman SE. Animal models of neuropsychiatric disorders. Nat Neurosci. 2010;13(10):1161–9. https://doi.org/10.1038/nn.2647.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Zhou BH, Li XJ, Liu M, Wu Z, Ming Hu X. Antidepressant-like activity of the Gastrodia elata ethanol extract in mice. Fitoterapia. 2006;77(7–8):592–4. https://doi.org/10.1016/j.fitote.2006.06.016.

    Article  PubMed  Google Scholar 

  47. Chen PJ, Hsieh CL, Su KP, Hou YC, Chiang HM, Lin IH, et al. The antidepressant effect of Gastrodia elata Bl. on the forced-swimming test in rats. Am J Chin Med. 2008;36(1):95–106. https://doi.org/10.1142/S0192415X08005618.

    Article  CAS  PubMed  Google Scholar 

  48. Chen PJ, Hsieh CL, Su KP, Hou YC, Chiang HM, Sheen LY. Rhizomes of Gastrodia elata B(L) possess antidepressant-like effect via monoamine modulation in subchronic animal model. Am J Chin Med. 2009;37(6):1113–24. https://doi.org/10.1142/S0192415X09007533.

    Article  PubMed  Google Scholar 

  49. Lin SH, Chen WC, Lu KH, Chen PJ, Hsieh SC, Pan TM, et al. Down-regulation of Slit-Robo pathway mediating neuronal cytoskeletal remodeling processes facilitates the antidepressive-like activity of Gastrodia elata Blume. J Agric Food Chem. 2014;62(43):10493–503. https://doi.org/10.1021/jf503132c.

    Article  CAS  PubMed  Google Scholar 

  50. Chen WC, Lai YS, Lin SH, Lu KH, Lin YE, Panyod S, et al. Anti-depressant effects of Gastrodia elata Blume and its compounds gastrodin and 4-hydroxybenzyl alcohol, via the monoaminergic system and neuronal cytoskeletal remodeling. J Ethnopharmacol. 2016;182:190–9. https://doi.org/10.1016/j.jep.2016.02.001.

    Article  CAS  PubMed  Google Scholar 

  51. Lin YE, Lin SH, Chen WC, Ho CT, Lai YS, Panyod S, et al. Antidepressant-like effects of water extract of Gastrodia elata Blume in rats exposed to unpredictable chronic mild stress via modulation of monoamine regulatory pathways. J Ethnopharmacol. 2016;187:57–65. https://doi.org/10.1016/j.jep.2016.04.032.

    Article  PubMed  Google Scholar 

  52. Zhang R, Peng Z, Wang H, Xue F, Chen Y, Wang Y, et al. Gastrodin ameliorates depressive-like behaviors and up-regulates the expression of BDNF in the hippocampus and hippocampal-derived astrocyte of rats. Neurochem Res. 2014;39(1):172–9. https://doi.org/10.1007/s11064-013-1203-0.

    Article  PubMed  Google Scholar 

  53. Wang H, Zhang R, Qiao Y, Xue F, Nie H, Zhang Z, et al. Gastrodin ameliorates depression-like behaviors and up-regulates proliferation of hippocampal-derived neural stem cells in rats: involvement of its anti-inflammatory action. Behav Brain Res. 2014;266:153–60. https://doi.org/10.1016/j.bbr.2014.02.046.

    Article  CAS  PubMed  Google Scholar 

  54. Lee B, Sur B, Yeom M, Shim I, Lee H, Hahm DH. Gastrodin reversed the traumatic stress-induced depressed-like symptoms in rats. J Nat Med. 2016;70(4):749–59. https://doi.org/10.1007/s11418-016-1010-4.

    Article  CAS  PubMed  Google Scholar 

  55. Cho JH, Park JH, Ahn JH, Lee JC, Hwang IK, Park SM, et al. Vanillin and 4-hydroxybenzyl alcohol promotes cell proliferation and neuroblast differentiation in the dentate gyrus of mice via the increase of brain-derived neurotrophic factor and tropomyosin-related kinase B. Mol Med Rep. 2016;13(4):2949–56. https://doi.org/10.3892/mmr.2016.4915.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  56. Xu J, Xu H, Liu Y, He H, Li G. Vanillin-induced amelioration of depression-like behaviors in rats by modulating monoamine neurotransmitters in the brain. Psychiatry Res. 2015;225(3):509–14. https://doi.org/10.1016/j.psychres.2014.11.056.

    Article  CAS  PubMed  Google Scholar 

  57. Abo-youssef AM. Possible antidepressant effects of vanillin against experimentally induced chronic mild stress in rats. Beni-Suef Univ J Basic Appl Sci. 2016;5(2):6.

    Google Scholar 

  58. Shoeb A, Chowta M, Pallempati G, Rai A, Singh A. Evaluation of antidepressant activity of vanillin in mice. Indian J Pharm. 2013;45(2):141–4. https://doi.org/10.4103/0253-7613.108292.

    Article  Google Scholar 

  59. Porsolt RD, Le Pichon M, Jalfre M. Depression: a new animal model sensitive to antidepressant treatments. Nature. 1977;266(5604):730–2.

    Article  CAS  PubMed  Google Scholar 

  60. Cryan JF, Slattery DA. Animal models of mood disorders: recent developments. Curr Opin Psychiatry. 2007;20(1):1–7. https://doi.org/10.1097/YCO.0b013e3280117733.

    Article  PubMed  Google Scholar 

  61. Schmidt HD, Duman RS. The role of neurotrophic factors in adult hippocampal neurogenesis, antidepressant treatments and animal models of depressive-like behavior. Behav Pharmacol. 2007;18(5–6):391–418. https://doi.org/10.1097/FBP.0b013e3282ee2aa8.

    Article  CAS  PubMed  Google Scholar 

  62. Steru L, Chermat R, Thierry B, Simon P. The tail suspension test: a new method for screening antidepressants in mice. Psychopharmacology. 1985;85(3):367–70.

    Article  CAS  PubMed  Google Scholar 

  63. Thierry B, Steru L, Simon P, Porsolt RD. The tail suspension test: ethical considerations. Psychopharmacology. 1986;90(2):284–5.

    Article  CAS  PubMed  Google Scholar 

  64. Katz RJ, Roth KA, Carroll BJ. Acute and chronic stress effects on open field activity in the rat: implications for a model of depression. Neurosci Biobehav Rev. 1981;5(2):247–51.

    Article  CAS  PubMed  Google Scholar 

  65. Fortunato JJ, Reus GZ, Kirsch TR, Stringari RB, Fries GR, Kapczinski F, et al. Effects of beta-carboline harmine on behavioral and physiological parameters observed in the chronic mild stress model: further evidence of antidepressant properties. Brain Res Bull. 2010;81(4–5):491–6. https://doi.org/10.1016/j.brainresbull.2009.09.008.

    Article  CAS  PubMed  Google Scholar 

  66. Seubert J, Rea AF, Loughead J, Habel U. Mood induction with olfactory stimuli reveals differential affective responses in males and females. Chem Senses. 2009;34(1):77–84. https://doi.org/10.1093/chemse/bjn054.

    Article  PubMed  Google Scholar 

  67. Ha JH, Shin SM, Lee SK, Kim JS, Shin US, Huh K, et al. In vitro effects of hydroxybenzaldehydes from Gastrodia elata and their analogues on GABAergic neurotransmission, and a structure-activity correlation. Planta Med. 2001;67(9):877–80. https://doi.org/10.1055/s-2001-18844.

    Article  CAS  PubMed  Google Scholar 

  68. Shi Y, Dong JW, Tang LN, Kang RX, Shi JG, Zhang JJ. N(6)-(3-methoxyl-4-hydroxybenzyl) adenine riboside induces sedative and hypnotic effects via GAD enzyme activation in mice. Pharmacol Biochem Behav. 2014;126:146–51. https://doi.org/10.1016/j.pbb.2014.09.004.

    Article  CAS  PubMed  Google Scholar 

  69. Shyamala BN, Naidu MM, Sulochanamma G, Srinivas P. Studies on the antioxidant activities of natural vanilla extract and its constituent compounds through in vitro models. J Agric Food Chem. 2007;55(19):7738–43. https://doi.org/10.1021/jf071349+.

    Article  CAS  PubMed  Google Scholar 

  70. Park H, Kim S, Kim S, Song Y, Seung K, Hong D, et al. Antioxidant and anti-inflammatory activities of hydroxybenzyl alcohol releasing biodegradable polyoxalate nanoparticles. Biomacromolecules. 2010;11(8):2103–8. https://doi.org/10.1021/bm100474w.

    Article  CAS  PubMed  Google Scholar 

  71. Lim EJ, Kang HJ, Jung HJ, Park EH. Anti-angiogenic, anti-inflammatory and anti-nociceptive activity of 4-hydroxybenzyl alcohol. J Pharm Pharmacol. 2007;59(9):1235–40. https://doi.org/10.1211/jpp.59.9.0007.

    Article  CAS  PubMed  Google Scholar 

  72. Yan X, Liu DF, Zhang XY, Liu D, Xu SY, Chen GX, et al. Vanillin protects dopaminergic neurons against inflammation-mediated cell death by inhibiting ERK1/2, P38 and the NF-kappaB signaling pathway. Int J Mol Sci. 2017;18(2) https://doi.org/10.3390/ijms18020389.

    Article  PubMed Central  Google Scholar 

  73. Zuo W, Xu F, Zhang K, Zheng L, Zhao J. Proliferation-enhancing effects of gastrodin on RSC96 Schwann cells by regulating ERK1/2 and PI3K signaling pathways. Biomed Pharmacother. 2016;84:747–53. https://doi.org/10.1016/j.biopha.2016.09.106.

    Article  CAS  PubMed  Google Scholar 

  74. Baral S, Pariyar R, Yoon CS, Kim DC, Yun JM, Jang SO, et al. Effects of Gastrodiae rhizoma on proliferation and differentiation of human embryonic neural stem cells. Asian Pac J Trop Med. 2015;8(10):792–7. https://doi.org/10.1016/j.apjtm.2015.09.004.

    Article  PubMed  Google Scholar 

  75. Manavalan A, Ramachandran U, Sundaramurthi H, Mishra M, Sze SK, Hu JM, et al. Gastrodia elata Blume (tianma) mobilizes neuro-protective capacities. Int J Biochem Mol Biol. 2012;3(2):219–41.

  76. Ramachandran U, Manavalan A, Sundaramurthi H, Sze SK, Feng ZW, Hu JM, et al. Tianma modulates proteins with various neuro-regenerative modalities in differentiated human neuronal SH-SY5Y cells. Neurochem Int. 2012;60(8):827–36. https://doi.org/10.1016/j.neuint.2012.03.012.

    Article  CAS  PubMed  Google Scholar 

  77. Sundaramurthi H, Manavalan A, Ramachandran U, Hu JM, Sze SK, Heese K. Phenotyping of tianma-stimulated differentiated rat neuronal b104 cells by quantitative proteomics. Neurosignals. 2012;20(1):48–60. https://doi.org/10.1159/000331492.

    Article  CAS  PubMed  Google Scholar 

  78. Belmaker RH, Agam G. Major depressive disorder. N Engl J Med. 2008;358(1):55–68. https://doi.org/10.1056/NEJMra073096.

    Article  CAS  PubMed  Google Scholar 

  79. Castren E. Is mood chemistry? Nat Rev Neurosci. 2005;6(3):241–6. https://doi.org/10.1038/nrn1629.

    Article  CAS  PubMed  Google Scholar 

  80. Lanni C, Govoni S, Lucchelli A, Boselli C. Depression and antidepressants: molecular and cellular aspects. Cell Mol Life Sci. 2009;66(18):2985–3008. https://doi.org/10.1007/s00018-009-0055-x.

    Article  CAS  PubMed  Google Scholar 

  81. Meyer JH, Wilson AA, Sagrati S, Miler L, Rusjan P, Bloomfield PM, et al. Brain monoamine oxidase A binding in major depressive disorder: relationship to selective serotonin reuptake inhibitor treatment, recovery, and recurrence. Arch Gen Psychiatry. 2009;66(12):1304–12. https://doi.org/10.1001/archgenpsychiatry.2009.156.

    Article  PubMed  Google Scholar 

  82. Macedo A, Leiria E, Filipe A. Pirlindole in the treatment of depression: a meta-analysis. Clin Drug Investig. 2011;31(1):61–71. https://doi.org/10.2165/11586690-000000000-00000.

    Article  CAS  PubMed  Google Scholar 

  83. Vaiva G, Thomas P, Ducrocq F, Fontaine M, Boss V, Devos P, et al. Low posttrauma GABA plasma levels as a predictive factor in the development of acute posttraumatic stress disorder. Biol Psychiatry. 2004;55(3):250–4.

    Article  CAS  PubMed  Google Scholar 

  84. Luscher B, Shen Q, Sahir N. The GABAergic deficit hypothesis of major depressive disorder. Mol Psychiatry. 2011;16(4):383–406. https://doi.org/10.1038/mp.2010.120.

    Article  CAS  PubMed  Google Scholar 

  85. Kucukibrahimoglu E, Saygin MZ, Caliskan M, Kaplan OK, Unsal C, Goren MZ. The change in plasma GABA, glutamine and glutamate levels in fluoxetine- or S-citalopram-treated female patients with major depression. Eur J Clin Pharmacol. 2009;65(6):571–7. https://doi.org/10.1007/s00228-009-0650-7.

    Article  CAS  PubMed  Google Scholar 

  86. Kapczinski F, Frey BN, Zannatto V. Physiopathology of bipolar disorders: what have changed in the last 10 years? Rev Bras Psiquiatr. 2004;26(Suppl 3):17–21.

    Article  PubMed  Google Scholar 

  87. Ahmad A, Rasheed N, Banu N, Palit G. Alterations in monoamine levels and oxidative systems in frontal cortex, striatum, and hippocampus of the rat brain during chronic unpredictable stress. Stress. 2010;13(4):355–64. https://doi.org/10.3109/10253891003667862.

    Article  PubMed  Google Scholar 

  88. Tagliari B, dos Santos TM, Cunha AA, Lima DD, Delwing D, Sitta A, et al. Chronic variable stress induces oxidative stress and decreases butyrylcholinesterase activity in blood of rats. J Neural Transm (Vienna). 2010;117(9):1067–76. https://doi.org/10.1007/s00702-010-0445-0.

    Article  CAS  PubMed  Google Scholar 

  89. Khanzode SD, Dakhale GN, Khanzode SS, Saoji A, Palasodkar R. Oxidative damage and major depression: the potential antioxidant action of selective serotonin re-uptake inhibitors. Redox Rep. 2003;8(6):365–70. https://doi.org/10.1179/135100003225003393.

    Article  CAS  PubMed  Google Scholar 

  90. Raison CL, Miller AH. Role of inflammation in depression: implications for phenomenology, pathophysiology and treatment. Mod Trends Pharmacopsychiatry. 2013;28:33–48. https://doi.org/10.1159/000343966.

    Article  CAS  PubMed  Google Scholar 

  91. Miller AH, Raison CL. The role of inflammation in depression: from evolutionary imperative to modern treatment target. Nat Rev Immunol. 2016;16(1):22–34. https://doi.org/10.1038/nri.2015.5.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  92. Miller AH, Maletic V, Raison CL. Inflammation and its discontents: the role of cytokines in the pathophysiology of major depression. Biol Psychiatry. 2009;65(9):732–41. https://doi.org/10.1016/j.biopsych.2008.11.029.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  93. Logan AC. Omega-3 fatty acids and major depression: a primer for the mental health professional. Lipids Health Dis. 2004;3:25. https://doi.org/10.1186/1476-511X-3-25.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  94. Naert G, Zussy C, Tran Van Ba C, Chevallier N, Tang YP, Maurice T, et al. Involvement of endogenous brain-derived neurotrophic factor in hypothalamic-pituitary-adrenal axis activity. J Neuroendocrinol. 2015;27(11):850–60. https://doi.org/10.1111/jne.12324.

    Article  CAS  PubMed  Google Scholar 

  95. Autry AE, Monteggia LM. Brain-derived neurotrophic factor and neuropsychiatric disorders. Pharmacol Rev. 2012;64(2):238–58. https://doi.org/10.1124/pr.111.005108.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  96. Duman RS, Monteggia LM. A neurotrophic model for stress-related mood disorders. Biol Psychiatry. 2006;59(12):1116–27. https://doi.org/10.1016/j.biopsych.2006.02.013.

    Article  CAS  PubMed  Google Scholar 

  97. Scharfman H, Goodman J, Macleod A, Phani S, Antonelli C, Croll S. Increased neurogenesis and the ectopic granule cells after intrahippocampal BDNF infusion in adult rats. Exp Neurol. 2005;192(2):348–56. https://doi.org/10.1016/j.expneurol.2004.11.016.

    Article  CAS  PubMed  Google Scholar 

  98. Hanson ND, Owens MJ, Nemeroff CB. Depression, antidepressants, and neurogenesis: a critical reappraisal. Neuropsychopharmacology. 2011;36(13):2589–602. https://doi.org/10.1038/npp.2011.220.

    Article  PubMed  PubMed Central  Google Scholar 

  99. Luo L. Actin cytoskeleton regulation in neuronal morphogenesis and structural plasticity. Annu Rev Cell Dev Biol. 2002;18:601–35. https://doi.org/10.1146/annurev.cellbio.18.031802.150501.

    Article  CAS  PubMed  Google Scholar 

  100. Svitkina T, Lin WH, Webb DJ, Yasuda R, Wayman GA, Van Aelst L, et al. Regulation of the postsynaptic cytoskeleton: roles in development, plasticity, and disorders. J Neurosci. 2010;30(45):14937–42. https://doi.org/10.1523/JNEUROSCI.4276-10.2010.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  101. Piubelli C, Gruber S, El Khoury A, Mathe AA, Domenici E, Carboni L. Nortriptyline influences protein pathways involved in carbohydrate metabolism and actin-related processes in a rat gene-environment model of depression. Eur Neuropsychopharmacol. 2011;21(7):545–62. https://doi.org/10.1016/j.euroneuro.2010.11.003.

    Article  CAS  PubMed  Google Scholar 

  102. Piubelli C, Vighini M, Mathe AA, Domenici E, Carboni L. Escitalopram affects cytoskeleton and synaptic plasticity pathways in a rat gene-environment interaction model of depression as revealed by proteomics. Part II: environmental challenge. Int J Neuropsychopharmacol. 2011;14(6):834–55. https://doi.org/10.1017/S1461145710001306.

    Article  PubMed  Google Scholar 

  103. Ladurelle N, Gabriel C, Viggiano A, Mocaer E, Baulieu EE, Bianchi M. Agomelatine (S20098) modulates the expression of cytoskeletal microtubular proteins, synaptic markers and BDNF in the rat hippocampus, amygdala and PFC. Psychopharmacology. 2012;221(3):493–509. https://doi.org/10.1007/s00213-011-2597-5.

    Article  CAS  PubMed  Google Scholar 

  104. Lin YE, Chou ST, Lin SH, Lu KH, Panyod S, Lai YS, et al. Antidepressant-like effects of water extract of Gastrodia elata Blume on neurotrophic regulation in a chronic social defeat stress model. J Ethnopharmacol. 2017;215:132–9. https://doi.org/10.1016/j.jep.2017.12.044.

    Article  PubMed  Google Scholar 

  105. Abelaira HM, Reus GZ, Quevedo J. Animal models as tools to study the pathophysiology of depression. Rev Bras Psiquiatr. 2013;35:S112–S20. https://doi.org/10.1590/1516-4446-2013-1098.

    Article  PubMed  Google Scholar 

  106. Ansari A, Bose S, Yadav MK, Wang JH, Song YK, Ko SG, et al. CST, an herbal formula, exerts anti-obesity effects through brain-gut-adipose tissue axis modulation in high-fat diet fed mice. Molecules. 2016;21(11) https://doi.org/10.3390/molecules21111522.

    Article  PubMed Central  Google Scholar 

  107. Hou Y, Jiang JG. Origin and concept of medicine food homology and its application in modern functional foods. Food Funct. 2013;4(12):1727–41. https://doi.org/10.1039/c3fo60295h.

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgments

This work was supported in part by grants from the Ministry of Science and Technology (NSC 97-2313-B-002-014-MY3 and MOST 104–2320-B-002–036-MY3) and the Council of Agriculture, Executive Yuan (105AS-16.1.1-CI-C2 and 106AS-16.1.1-CI-C2), Taiwan.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Lee-Yan Sheen.

Ethics declarations

Conflict of Interest

Yu-En Lin, Kuan-Hung Lu, and Lee-Yan Sheen declare that they have no conflict of interest.

Human and Animal Rights and Informed Consent

This article does not contain any studies with human or animal subjects performed by any of the authors.

Additional information

This article is part of the Topical Collection on Natural Products: From Chemistry to Pharmacology

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Lin, YE., Lu, KH. & Sheen, LY. Recent Research Progress on the Antidepressant-like Effect and Neuropharmacological Potential of Gastrodia elata Blume. Curr Pharmacol Rep 4, 220–237 (2018). https://doi.org/10.1007/s40495-018-0137-2

Download citation

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s40495-018-0137-2

Keywords

Navigation