Skip to main content

Advertisement

Log in

Pituitary Adenylate Cyclase-Activating Polypeptide in the Ventromedial Hypothalamus Is Responsible for Food Intake Behavior by Modulating the Expression of Agouti-Related Peptide in Mice

  • Published:
Molecular Neurobiology Aims and scope Submit manuscript

Abstract

Pituitary adenylate cyclase-activating polypeptide (PACAP) is abundantly expressed in the hypothalamus and contributes to hypothalamic functions, including appetite regulation. Although food intake is suggested to be decreased in PACAP (−/−) mice, the detailed mechanisms are still being discussed. We sought to investigate this link. The food consumption at 8 h after refeeding in the (−/−) mice who had fasted for 2 days was significantly lower than in the PACAP (+/+) mice. The nocturnal and daily food intake of (−/−) mice was significantly lower than those of (+/+) mice, but the diurnal food intake showed a tendency to increase. mRNA expression levels of agouti-related peptide (AgRP) were decreased, but those of proopiomelanocortin (POMC) were increased in the hypothalamus of (−/−) mice 4 h after refeeding. Furthermore, intracerebroventricular administration of a PACAP receptor antagonist, PACAP6–38 (1 nmol/4 μL/mouse), decreased food intake and body weight 1, 2, and 4 h after refeeding, as well as expression levels of AgRP at 4 h after refeeding in (+/+) mice. The selective overexpression of PACAP by the infection of an adeno-associated virus in the ventromedial hypothalamus (VMH) resulted in an increase in food intake and AgRP expression in the nocturnal period in addition to the increased food intake at 8 h after refeeding. These results suggest that food intake behavior in mice is triggered by the increase in PACAP expression in the VMH via modulation of AgRP expression in the hypothalamus, pointing to PACAP inhibition as a potential strategy for the development of anti-obesity drugs.

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.

Institutional subscriptions

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

Similar content being viewed by others

Abbreviations

AgRP:

Agouti-related peptide

BNST:

Bed nucleus of the stria terminalis

CeA:

Central amygdala

CART:

Cocaine- and amphetamine-regulated transcript

NPY:

Neuropeptide Y

SF-1:

Steroidogenic factor 1

PACAP:

Pituitary adenylate cyclase-activating polypeptide

POMC:

Proopiomelanocortin

PVH:

Paraventricular hypothalamus

VMH:

Ventromedial hypothalamus

(−/−):

Knockout

(+/+):

Wild type

References

  1. Miyata A, Arimura A, Dahl RR, Minamino N, Uehara A, Jiang L, Culler MD, Coy DH (1989) Isolation of a novel 38 residue-hypothalamic polypeptide which stimulates adenylate cyclase in pituitary cells. Biochem Biophys Res Commun 164(1):567–574. https://doi.org/10.1016/0006-291X(89)91757-9

    Article  CAS  PubMed  Google Scholar 

  2. Miyata A, Jiang L, Dahl RD, Kitada C, Kubo K, Fujino M, Minamino N, Arimura A (1990) Isolation of a neuropeptide corresponding to the N-terminal 27 residues of the pituitary adenylate cyclase activating polypeptide with 38 residues (PACAP38). Biochem Biophys Res Commun 170(2):643–648. https://doi.org/10.1016/0006-291X(90)92140-U

    Article  CAS  PubMed  Google Scholar 

  3. Vaudry D, Falluel-Morel A, Bourgault S, Basille M, Burel D, Wurtz O, Fournier A, Chow BKC et al (2009) Pituitary adenylate cyclase-activating polypeptide and its receptors: 20 years after the discovery. Pharmacol Rev 61(3):283–357. https://doi.org/10.1124/pr.109.001370

    Article  CAS  PubMed  Google Scholar 

  4. Vaudry D, Gonzalez BJ, Basille M, Yon L, Fournier A, Vaudry H (2000) Pituitary adenylate cyclase-activating polypeptide and its receptors: from structure to functions. Pharmacol Rev 52(2):269–324

    CAS  PubMed  Google Scholar 

  5. Yokai M, Kurihara T, Miyata A (2016) Spinal astrocytic activation contributes to both induction and maintenance of pituitary adenylate cyclase-activating polypeptide type 1 receptor-induced long-lasting mechanical allodynia in mice. Mol Pain 12:1744806916646383. https://doi.org/10.1177/1744806916646383

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Rudecki AP, Gray SL (2016) PACAP in the defense of energy homeostasis. Trends Endocrinol Metab 27(9):620–632. https://doi.org/10.1016/j.tem.2016.04.008

    Article  CAS  PubMed  Google Scholar 

  7. Fulop DB, Humli V, Szepesy J, Ott V, Reglodi D, Gaszner B, Nemeth A, Szirmai A et al (2019) Hearing impairment and associated morphological changes in pituitary adenylate cyclase activating polypeptide (PACAP)-deficient mice. Sci Rep 9(1):14598. https://doi.org/10.1038/s41598-019-50775-z

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Denes V, Geck P, Mester A, Gabriel R (2019) Pituitary adenylate cyclase-activating polypeptide: 30 years in research spotlight and 600 million years in service. J Clin Med 8(9):1488. https://doi.org/10.3390/jcm8091488

    Article  PubMed Central  Google Scholar 

  9. Mounien L, Bizet P, Boutelet I, Gourcerol G, Fournier A, Vaudry H, Jégou S (2006) Pituitary adenylate cyclase-activating polypeptide directly modulates the activity of proopiomelanocortin neurons in the rat arcuate nucleus. Neuroscience 143(1):155–163. https://doi.org/10.1016/j.neuroscience.2006.07.022

    Article  CAS  PubMed  Google Scholar 

  10. Mounien L, Do Rego J-C, Bizet P, Boutelet I, Gourcerol G, Fournier A, Brabet P, Costentin J et al (2008) Pituitary adenylate cyclase-activating polypeptide inhibits food intake in mice through activation of the hypothalamic melanocortin system. Neuropsychopharmacology 34(2):424–435

    Article  PubMed  Google Scholar 

  11. Resch JM, Maunze B, Phillips KA, Choi S (2014) Inhibition of food intake by PACAP in the hypothalamic ventromedial nuclei is mediated by NMDA receptors. Physiol Behav 133:230–235. https://doi.org/10.1016/j.physbeh.2014.05.029

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Resch JM, Boisvert JP, Hourigan AE, Mueller CR, Yi SS, Choi S (2011) Stimulation of the hypothalamic ventromedial nuclei by pituitary adenylate cyclase-activating polypeptide induces hypophagia and thermogenesis. Am J Phys Regul Integr Comp Phys 301(6):R1625–R1634. https://doi.org/10.1152/ajpregu.00334.2011

    Article  CAS  Google Scholar 

  13. Resch JM, Maunze B, Gerhardt AK, Magnuson SK, Phillips KA, Choi S (2013) Intrahypothalamic pituitary adenylate cyclase-activating polypeptide regulates energy balance via site-specific actions on feeding and metabolism. Am J Physiol Endocrinol Metab 305(12):E1452–E1463. https://doi.org/10.1152/ajpendo.00293.2013

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Iemolo A, Ferragud A, Cottone P, Sabino V (2015) Pituitary adenylate cyclase-activating peptide in the central amygdala causes anorexia and body weight loss via the melanocortin and the TrkB systems. Neuropsychopharmacology 40(8):1846–1855. https://doi.org/10.1038/npp.2015.34

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Kocho-Schellenberg M, Lezak KR, Harris OM, Roelke E, Gick N, Choi I, Edwards S, Wasserman E et al (2014) PACAP in the BNST produces anorexia and weight loss in male and female rats. Neuropsychopharmacology 39(7):1614–1623. https://doi.org/10.1038/npp.2014.8

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Nakata M, Kohno D, Shintani N, Nemoto Y, Hashimoto H, Baba A, Yada T (2004) PACAP deficient mice display reduced carbohydrate intake and PACAP activates NPY-containing neurons in the rat hypothalamic arcuate nucleus. Neurosci Lett 370(2–3):252–256. https://doi.org/10.1016/j.neulet.2004.08.034

    Article  CAS  PubMed  Google Scholar 

  17. Tomimoto S, Ojika T, Shintani N, Hashimoto H, K-i H, Ikeda K, Nakata M, Yada T et al (2008) Markedly reduced white adipose tissue and increased insulin sensitivity in Adcyap1-deficient mice. J Pharmacol Sci 107(1):41–48. https://doi.org/10.1254/jphs.FP0072173

    Article  CAS  PubMed  Google Scholar 

  18. Hahn TM, Breininger JF, Baskin DG, Schwartz MW (1998) Coexpression of Agrp and NPY in fasting-activated hypothalamic neurons. Nat Neurosci 1:271–272. https://doi.org/10.1038/1082

    Article  CAS  PubMed  Google Scholar 

  19. Morton GJ, Cummings DE, Baskin DG, Barsh GS, Schwartz MW (2006) Central nervous system control of food intake and body weight. Nature 443:289–295. https://doi.org/10.1038/nature05026

    Article  CAS  PubMed  Google Scholar 

  20. Biebermann H, Kühnen P, Kleinau G, Krude H (2012) The neuroendocrine circuitry controlled by POMC, MSH, and AGRP. In: Joost H-G (ed) Appetite control. Springer, Berlin Heidelberg, pp. 47–75. https://doi.org/10.1007/978-3-642-24716-3_3

    Chapter  Google Scholar 

  21. Hagan MM, Rushing PA, Pritchard LM, Schwartz MW, Strack AM, Ploeg LHTV, Woods SC, Seeley RJ (2000) Long-term orexigenic effects of AgRP-(83—132) involve mechanisms other than melanocortin receptor blockade. Am J Physiol Regul Integr Comp Physiol 279(1):R47–R52. https://doi.org/10.1152/ajpregu.2000.279.1.R47

    Article  CAS  PubMed  Google Scholar 

  22. Luquet S, Perez FA, Hnasko TS, Palmiter RD (2005) NPY/AgRP neurons are essential for feeding in adult mice but can be ablated in neonates. Science 310(5748):683–685. https://doi.org/10.1126/science.1115524

    Article  CAS  PubMed  Google Scholar 

  23. Cowley MA, Smart JL, Rubinstein M, Cerdán MG, Diano S, Horvath TL, Cone RD, Low MJ (2001) Leptin activates anorexigenic POMC neurons through a neural network in the arcuate nucleus. Nature 411(6836):480–484. https://doi.org/10.1038/35078085

    Article  CAS  PubMed  Google Scholar 

  24. Spanswick D, Smith MA, Mirshamsi S, Routh VH, Ashford MLJ (2000) Insulin activates ATP-sensitive K+ channels in hypothalamic neurons of lean, but not obese rats. Nat Neurosci 3(8):757–758. https://doi.org/10.1038/77660

    Article  CAS  PubMed  Google Scholar 

  25. Air EL, Clegg DJ, Seeley RJ, Benoit SC, Woods SC (2002) Insulin and leptin combine additively to reduce food intake and body weight in rats. Endocrinology 143(6):2449–2452. https://doi.org/10.1210/endo.143.6.8948

    Article  CAS  PubMed  Google Scholar 

  26. Cowley MA, Smith RG, Diano S, Tschöp M, Pronchuk N, Grove KL, Strasburger CJ, Bidlingmaier M et al (2003) The distribution and mechanism of action of ghrelin in the CNS demonstrates a novel hypothalamic circuit regulating energy homeostasis. Neuron 37(4):649–661. https://doi.org/10.1016/S0896-6273(03)00063-1

    Article  CAS  PubMed  Google Scholar 

  27. Nakazato M, Murakami N, Date Y, Kojima M, Matsuo H, Kangawa K, Matsukura S (2001) A role for ghrelin in the central regulation of feeding. Nature 409(6817):194–198. https://doi.org/10.1038/35051587

    Article  CAS  PubMed  Google Scholar 

  28. Wren AM, Murphy KG, Seal LJ, Cohen MA, Ghatei MA, Bloom SR, Dhillo WS, Brynes AE et al (2001) Ghrelin enhances appetite and increases food intake in humans. J Clin Endocrinol Metab 86(12):5992–5992. https://doi.org/10.1210/jcem.86.12.8111

    Article  CAS  PubMed  Google Scholar 

  29. K-i N, Cui Z, Li C, Meister J, Cui Y, Fu O, Smith AS, Jain S et al (2016) Gs-coupled GPCR signalling in AgRP neurons triggers sustained increase in food intake. Nat Commun 7:10268. https://doi.org/10.1038/ncomms10268

    Article  CAS  Google Scholar 

  30. Mizuno Y, Kondo K, Terashima Y, Arima H, Murase T, Oiso Y (1998) Anorectic effect of pituitary adenylate cyclase activating polypeptide (PACAP) in rats: lack of evidence for involvement of hypothalamic neuropeptide gene expression. J Neuroendocrinol 10(8):611–616

    Article  CAS  PubMed  Google Scholar 

  31. Krashes MJ, Shah BP, Madara JC, Olson DP, Strochlic DE, Garfield AS, Vong L, Pei H et al (2014) An excitatory paraventricular nucleus to AgRP neuron circuit that drives hunger. Nature 507:238–242. https://doi.org/10.1038/nature12956

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Hashimoto H, Shintani N, Tanaka K, Mori W, Hirose M, Matsuda T, Sakaue M, J-i M et al (2001) Altered psychomotor behaviors in mice lacking pituitary adenylate cyclase-activating polypeptide (PACAP). Proc Natl Acad Sci 98(23):13355–13360. https://doi.org/10.1073/pnas.231094498

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Tong Q, Ye C-P, Jones JE, Elmquist JK, Lowell BB (2008) Synaptic release of GABA by AgRP neurons is required for normal regulation of energy balance. Nat Neurosci 11(9):998–1000

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Tochitani S, Liang F, Watakabe A, Hashikawa T, Yamamori T (2001) The occ1 gene is preferentially expressed in the primary visual cortex in an activity-dependent manner: a pattern of gene expression related to the cytoarchitectonic area in adult macaque neocortex. Eur J Neurosci 13(2):297–307. https://doi.org/10.1046/j.0953-816X.2000.01390.x

    Article  CAS  PubMed  Google Scholar 

  35. An JJ, Gharami K, Liao G-Y, Woo NH, Lau AG, Vanevski F, Torre ER, Jones KR et al (2008) Distinct role of long 3′ UTR BDNF mRNA in spine morphology and synaptic plasticity in hippocampal neurons. Cell 134(1):175–187. https://doi.org/10.1016/j.cell.2008.05.045

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Zolotukhin S, Byrne BJ, Mason E, Zolotukhin I, Potter M, Chesnut K, Summerford C, Samulski RJ et al (1999) Recombinant adeno-associated virus purification using novel methods improves infectious titer and yield. Gene Ther 6:973–985. https://doi.org/10.1038/sj.gt.3300938

    Article  CAS  PubMed  Google Scholar 

  37. Christine Aurnhammer MH, Muether N, Hausl M, Rauschhuber C, Huber I, Nitschko H, Busch U, Sing A et al (2012) Universal real-time PCR for the detection and quantification of adeno-associated virus serotype 2-derived inverted terminal repeat sequences. Hum Gene Ther Methods 23(1):18–28. https://doi.org/10.1089/hgtb.2011.034

    Article  CAS  PubMed  Google Scholar 

  38. Kong D, Tong Q, Ye C, Koda S, Fuller Patrick M, Krashes Michael J, Vong L, Ray Russell S et al (2012) GABAergic RIP-Cre neurons in the arcuate nucleus selectively regulate energy expenditure. Cell 151(3):645–657. https://doi.org/10.1016/j.cell.2012.09.020

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Krashes MJ, Shah BP, Koda S, Lowell BB (2013) Rapid versus delayed stimulation of feeding by the endogenously released AgRP neuron mediators GABA, NPY, and AgRP. Cell Metab 18(4):588–595. https://doi.org/10.1016/j.cmet.2013.09.009

    Article  CAS  PubMed  Google Scholar 

  40. Kirihara Y, Takechi M, Kurosaki K, Kobayashi Y, Kurosawa T (2013) Anesthetic effects of a mixture of medetomidine, midazolam and butorphanol in two strains of mice. Exp Anim 62(3):173–180. https://doi.org/10.1538/expanim.62.173

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Paxinos G, Franklin K (2011) The mouse brain in stereotaxic coordinates, 2nd Edition. Academic Press, San Diego, California. ISBN: 0-12-547637-X.

  42. Kambe Y, Miyata A (2016) Mitochondrial c-Fos may increase the vulnerability of Neuro2a cells to cellular stressors. J Mol Neurosci 59(1):106–112. https://doi.org/10.1007/s12031-015-0710-7

    Article  CAS  PubMed  Google Scholar 

  43. Nakamachi T, Kamata E, Tanigawa A, Konno N, Shioda S, Matsuda K (2018) Distribution of pituitary adenylate cyclase-activating polypeptide 2 in zebrafish brain. Peptides 103:40–47. https://doi.org/10.1016/j.peptides.2018.03.006

    Article  CAS  PubMed  Google Scholar 

  44. Saegusa H, Kurihara T, Zong S, Minowa O, A-a K, Han W, Matsuda Y, Yamanaka H et al (2000) Altered pain responses in mice lacking α1E subunit of the voltage-dependent Ca2+ channel. Proc Natl Acad Sci 97(11):6132–6137. https://doi.org/10.1073/pnas.100124197

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Le H, Ahn BJ, Lee HS, Shin A, Chae S, Lee SY, Shin MW, Lee E-J et al (2017) Disruption of Ninjurin1 leads to repetitive and anxiety-like behaviors in mice. Mol Neurobiol 54(9):7353–7368. https://doi.org/10.1007/s12035-016-0207-6

    Article  CAS  PubMed  Google Scholar 

  46. Krashes MJ, Koda S, Ye C, Rogan SC, Adams AC, Cusher DS, Maratos-Flier E, Roth BL et al (2011) Rapid, reversible activation of AgRP neurons drives feeding behavior in mice. J Clin Invest 121(4):1424–1428. https://doi.org/10.1172/jci46229

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Parker KL (1998) The roles of steroidogenic factor 1 in endocrine development and function. Mol Cell Endocrinol 145 (1):15–20. https://doi.org/10.1016/S0303-7207(98)00164-6

  48. Horvath TL, Diano S (2004) The floating blueprint of hypothalamic feeding circuits. Nat Rev Neurosci 5(8):662–667

    Article  CAS  PubMed  Google Scholar 

  49. Hawke Z, Ivanov TR, Bechtold DA, Dhillon H, Lowell BB, Luckman SM (2009) PACAP neurons in the hypothalamic ventromedial nucleus are targets of central leptin signaling. J Neurosci 29(47):14828–14835. https://doi.org/10.1523/jneurosci.1526-09.2009

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Mustafa T, Jiang SZ, Eiden AM, Weihe E, Thistlethwaite I, Eiden LE (2015) Impact of PACAP and PAC1 receptor deficiency on the neurochemical and behavioral effects of acute and chronic restraint stress in male C57BL/6 mice. Stress 18(4):408–418. https://doi.org/10.3109/10253890.2015.1025044

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Dore R, Iemolo A, Smith KL, Wang X, Cottone P, Sabino V (2013) CRF mediates the anxiogenic and anti-rewarding, but not the anorectic effects of PACAP. Neuropsychopharmacology 38(11):2160–2169. https://doi.org/10.1038/npp.2013.113

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. Bruce AA, Sarah LG, Emma RI, Antonio CB, Antonio JV-P, Nancy MS (2008) Feeding and metabolism in mice lacking pituitary adenylate cyclase-activating polypeptide. Endocrinology 149(4):1571–1580. https://doi.org/10.1210/en.2007-0515

    Article  CAS  Google Scholar 

Download references

Acknowledgments

We would like to thank Ms. Izumi Fujisima and Mr. Tetsuya Kawamura for their technical contribution and all the staff members of the Joint Research Laboratory and the Division of Laboratory Animal Sciences, Kagoshima University for their help with animal care and the use of the facilities. We are also grateful to the Ministry of Agriculture and Rural Development, Vietnam, for the doctoral scholarship to T.T.N.

Funding

This work was supported by a Grant-in-Aid for Scientific Research (C), Japan Society for the Promotion of Science (JSPS) (JSPS KAKENHI Grant No. 17K08310, 17 K08599 and JP19K07121), a Grant-in-Aid for Scientific Research (B) (Grant No. JP17H03989), MEXT KAKENHI, (grant number JP18H05416), and AMED (grant No. JP19dm0107122h0004 and JP19dm0207061h0003).

Author information

Authors and Affiliations

Authors

Contributions

T.T.N carried out the experiments, performed statistical analysis, and drafted the manuscript. YK carried out the experiments, performed behavioral studies, and wrote the manuscript. TK and AM conceived of and participated in the design of the study and wrote the manuscript. TN, NS, and HH participated in the design of the study and reviewed the manuscript. All authors read and approved of the final manuscript.

Corresponding author

Correspondence to Atsuro Miyata.

Ethics declarations

All applicable international, national, and/or institutional guidelines for the care and use of animals were followed. Specifically, all experiments in the present study were approved by the Experimental Animal Research Committee of Kagoshima University (Approval numbers: MD17054 and MD18105) and the Gene Recombination Experiment Safety Committee of Kagoshima University (Approval number: S28006).

Conflict of Interest

The authors declare that they have no conflict of interest.

Additional information

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Electronic Supplementary Material

ESM 1

PACAP (-/-) mice had reduced mesenteric and epididymal fat masses. Fat mass in mesenteric (a) and epididymal tissues (b) under the conditions indicated on the horizontal axis (n = 9 to 11 mice per group). *p<0.05; Student’s t-test (PNG 97 kb)

ESM 2

Successful expression of PACAP after transient transfection of pAAV-CAG::PACAP-IRES-EGFP. (a) HEK293 cells were transiently transfected with pAAV-CAG::IRES-EGFP or pAAV-CAG::PACAP-IRES-EGFP, and mRNA expression levels of PACAP and GAPDH (reference) were quantified by RT-qPCR (n = 2 mice per group). (b) HEK293 cells were transiently transfected with pAAV-CAG::PACAP-IRES-EGFP, and expression of PACAP (red) and GFP (green) was confirmed by immunohistochemistry with each specific antibody. Arrow indicates PACAP immunoreactivity. Scale bars, 10 μm (PNG 601 kb)

ESM 3

PACAP (-/-) mice exhibited enhanced locomotor activity and anti-anxiety-like behaviors compared with their PACAP (+/+) littermates. Distance travelled (a) and time spent in the center region (b) of (+/+) and (-/-) mice in the open-field test. The time spent in the open arms (c) of (+/+) and (-/-) mice in the elevated plus-maze test (n = 5 to 6 mice per group). *p<0.05; Student’s t-test (PNG 104 kb)

ESM 4

List of primer sequences used for genotyping (DOCX 13 kb)

ESM 5

List of primer sequences used for RT-qPCR (DOCX 15 kb)

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Nguyen, T.T., Kambe, Y., Kurihara, T. et al. Pituitary Adenylate Cyclase-Activating Polypeptide in the Ventromedial Hypothalamus Is Responsible for Food Intake Behavior by Modulating the Expression of Agouti-Related Peptide in Mice. Mol Neurobiol 57, 2101–2114 (2020). https://doi.org/10.1007/s12035-019-01864-7

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s12035-019-01864-7

Keywords

Navigation