<<

COMMENTARY COMMENTARY

BDNF signaling: Harnessing stress to battle mood disorder Pawel Licznerskia and Elizabeth A. Jonasa,1

The link between the onset of major depressive disor- double knockout (DKO) of Gαi1 and Gαi3 results der (MDD) and loss of in the brain is in inhibition of BDNF-induced activation of Akt– of interest to clinicians and basic scientists. MDD is mTORC1 and ERK pathways. shRNA-mediated down- caused by a combination of genetic, environmental, and regulation of Gαi1 and Gαi3 also affects dendritic psychological factors. Trauma, chronic health problems, outgrowth and formation of dendritic spines in the and substance abuse are risks (1), as are grief and other . Specific behavioral studies performed purely emotional/cognitive stresses (2, 3). MDD alters by the Marshall team reveal that shRNA knockdown of the expression of neurotrophins, such as brain-derived Gαi1 and Gαi3 or complete DKO cause depression- neurotrophic factor (BDNF). BDNF is required for neu- like behaviors. Their studies suggest that downstream ronal development, survival, and plasticity (4, 5). Brain BDNF signaling via Gαi1 and Gαi3 is necessary not imaging has shown volumetric changes in limbic regions only for sustaining the well-being of but also in depression attributed either to reduced numbers of for normal antidepressive behaviors (11). So, how and pyramidal neurons or to their reduced cell body does signaling inside the cell specifically size, accompanied by atrophy of pyramidal api- contribute to the regulation of mental functioning? cal and decreases in neurogenesis in dentate Neurotrophins, a unique family of polypeptide gyrus (6). These structural alterations most likely contrib- growth factors that include BDNF, regulate prolifera- ute to features of depression, including cognitive tion, proper differentiation, survival, and death of impairment, helplessness, and anhedonia. Neuronal neuronal and nonneuronal cells. Neurotrophins act dysfunction also affects activation of the hypotha- through downstream signaling cascades following lamic–pituitary–adrenal axis (7). Studies show that struc- receptor activation. Brain neurotrophins regulate early tural and functional neuronal alterations in MDD are prenatal brain development and adult central nervous partially rescued by antidepressant treatment. Antide- system plasticity (4). BDNF is the predominant neuro- pressants, in addition to their effects on neurotransmit- trophin in the brain (12). Synthesized as a precursor ter levels, also increase BDNF and enhance expression proBDNF, it is cleaved to release the mature, active of the receptor for BDNF, tropomyosin-related kinase B form (11). Mature BDNF binds preferentially to the (TrkB), in the hippocampus (8). However, the intracellular TrkB receptor, activating downstream signaling path- mechanisms governing the relationship between BDNF, ways such as mitogen-activated protein kinase (MAPK), structural changes in limbic system cells, and clinical phospholipase Cγ, and phosphatidylinositol-3 kinase manifestations of MDD are still unclear. pathway. These signaling cascades regulate transcrip- In PNAS, Marshall et al. (9) demonstrate that tion and dendritic translation of proteins required for Gαi1 and Gαi3, members of the GαI subclass of het- neuronal survival, differentiation, and learning and mem- erotrimeric G proteins, are essential for BDNF/TrkB ory formation in the hippocampus (13, 14). signaling in hippocampus and are down-regulated by Upon elimination of Gαi1 and Gαi3, Marshall et al. chronic stress. G proteins form membrane-associated also observed decreased dendritic branching and re- heterotrimers consisting of α, β, and γ subunits (10). duced numbers of synaptic spines. Substantial loss of There are four main subclasses: Gs, Gi/o, Gq, and CA1 pyramidal neurons was observed in the Gαi1/ G12/13. GαI belongs to the Gi/o subclass and inhibits Gαi3 DKO mouse, which was not found when Gαi1 adenylyl cyclase. Marshall et al. report that reductions and Gαi3 were depleted in adolescent brain using in Gαi1 and Gαi3 levels affect BDNF-induced TrkB shRNA, although this latter model produced alter- endocytosis and activation of signaling pathways down- ations in synaptic structure and the same behavioral stream of TrkB, resulting in depressive behaviors. deficits as the DKO. It is not clear whether loss of In mouse embryonic fibroblasts, they show that the neurons in the DKO mouse hippocampus was caused

aDepartment of Internal Medicine, Yale University, New Haven, CT 06520 Author contributions: P.L. and E.A.J. wrote the paper. The authors declare no conflict of interest. Published under the PNAS license. See companion article on page E3549. 1To whom correspondence should be addressed. Email: [email protected]. Published online March 28, 2018.

3742–3744 | PNAS | April 10, 2018 | vol. 115 | no. 15 www.pnas.org/cgi/doi/10.1073/pnas.1803645115 Downloaded by guest on October 5, 2021 proBDNF

Presynapc

BDNF pro-pepde BDNF

Stress TrkB p75NTR

P P Postsynapc P-eEF2 MEK 1/2 PI3K Apoptosis ATP Bcl-2 LTD retracon ERK 1/2 AKT

mTOR

Neuronal differenaon, survival and synapc plascity

Fig. 1. BDNF binding to TrkB receptor activates signaling cascades responsible for neuronal survival and synaptic plasticity. ProBDNF binds p75NTR and triggers long-term depression and apoptosis. Upon stress, including high-frequency synaptic stimulation, Bcl-2 is recruited to mitochondria to decrease the production of reactive oxygen species (ROS) and enhance effective ATP production; this supports relevant gene transcription and protein translation. Mild acute stress or high frequency synaptic activity recruit and may require BDNF signaling during learning and memory formation and for antidepressant coping strategies.

by loss of developmental targeting of neurons or by enhanced cell and facilitates hippocampal long-term depression, negatively affect- death during neurogenesis or synaptogenesis. Whether longer ing learning and memory formation (Fig. 1) (4, 23). Perhaps the struc- overexpression of Gαi1/Gαi3 shRNA and/or a more challenging tural changes in the Gαi1- and Gαi3-depleted and the DKO mice, stress paradigm might also induce neuronal death remains to be including loss of pyramidal neurons in the hippocampus, might be answered, but it nevertheless seems clear that negative synaptic partially attributed to the actions of proBDNF; the observed synaptic changes are sufficient to produce the depressive features. deficiencies even in the absence of cell death might be related to Chronic stress substantially lowers BDNF (15). The insult cho- apoptotic signaling in the alone (24, 25). sen by Marshall et al. to cause depression in the animals, chronic Stress also regulates intracellular signaling apart from its mild stress, consists of 3 wk of sequential forced swim, restraint, specific effects on BDNF (Fig. 1). Interestingly, brief stress- water and food deprivation, housing in wet sawdust, light/dark induced release of glucocorticoids, at low doses, supports neuro- cycle reversal, and housing in constant illumination or darkness nal branching and survival and positively influences performance (16, 17). In normal rodents, BDNF mRNA levels increase during in spatial learning and memory tasks (26, 27), implicating benefi- early postnatal days in cortex, hippocampus, and , cor- cial changes in limbic system networks. Under these conditions, relating with the peak of postnatal synaptogenesis (18). BDNF is corticosterone improves mitochondrial membrane potential and also required in adulthood for proper brain functioning, support- oxidation. The glucocorticoid receptor binds to the antiapoptotic ing neuronal network strengthening and plasticity. Although most B-cell lymphoma 2 protein (Bcl-2). This complex translocates into BDNF homozygous knockout mice die in the first 2 postnatal days, mitochondria to reduce the production of damaging reactive ox- some survive for 2–4 wk (19). These surviving mice nevertheless ygen species (ROS) and prevent the opening of the prodeath show severe impairments in growth and in movement coordina- permeability transition pore (28) while enhancing the supply of tion, and have reduced numbers of cranial and spinal sensory ATP required for proper brain function. The regulation of mito- neurons, albeit with no gross structural abnormalities of the hip- chondrial ATP production by low-dose corticosteroids may pocampus (19, 20). They do, however, show impaired hippo- thereby shape the proper response of neurons to external stress. campal long-term potentiation (LTP) and reduced numbers of In contrast, high levels or chronic stress produce the opposite presynaptic vesicles docked at presynaptic active zones (21), suggest- effects on mitochondria, depolarizing them and predisposing to ing dysfunctional synapses. Apart from BDNF binding to the TrkB activation of apoptotic mechanisms (28). receptor, the precleaved form of BDNF, proBDNF, can act as a sig- Neuronal excitability and synaptic plasticity require high naling molecule in the opposite manner to mature BDNF. ProBDNF mitochondrial fidelity (29, 30). Energy-dependent activities include binds specifically to the p75 neurotrophin receptor (p75NTR), a mem- calcium clearance in a timely manner, rapid actin cytoskeletal rear- ber of the tumor necrosis factor superfamily (22). Activation of p75NTR rangements, trafficking of synaptic vesicles, resetting of ion gradi- decreases the complexity of neuronal projections, triggers apoptosis, ents after action potential firing, and new protein phosphorylation

Licznerski and Jonas PNAS | April 10, 2018 | vol. 115 | no. 15 | 3743 Downloaded by guest on October 5, 2021 in response to synaptic stimulation (31). These events, crucial for by exquisitely timed eEF2 dephosphorylation and increased , are regulated by the timing and availability of mi- translation of specific synaptic proteins. In summary (Fig. 1), the tochondrial energy supply. In cultured hippocampal neurons, high-frequency stimulation that produces LTP is a stress to neu- BDNF treatment causes accumulation of mitochondria at presyn- rons and mitochondria that is alleviated by intracellular events that aptic sites (32) where mitochondria contribute to produce synaptic strengthening. Analogously, gene transcription docking and reserve pool galvanization (33–35). Specific phos- and translation of specific proteins in response to synaptic activity phorylation events may relate to mitochondrial ATP production in the limbic system may be necessary for the proper response to during hippocampal synaptic plasticity downstream of NMDA re- stress to prevent depression and enhance positive cognitive cop- ceptor stimulation. For example, phosphorylation of eukaryotic ing strategies. Loss of activation of growth factor-supported sig- elongation factor 2 (eEF2) within the first few minutes after stim- naling pathways contributes to faulty responses to neuronal ulation results in decreased overall protein synthesis (36), followed activity and stress.

1 Otte C, et al. (2016) Major depressive disorder. Nat Rev Dis Primers 2:16065. 2 Kristensen P, Weisæth L, Heir T (2012) Bereavement and mental health after sudden and violent losses: A review. Psychiatry 75:76–97. 3 Giacco D, Laxhman N, Priebe S (November 26, 2017) Prevalence of and risk factors for mental disorders in refugees. Semin Cell Dev Biol, 10.1016/j. semcdb.2017.11.030. 4 Vicario-Abej ´onC, Owens D, McKay R, Segal M (2002) Role of neurotrophins in central formation and stabilization. Nat Rev Neurosci 3:965–974. 5 Smith MA, Makino S, Kvetnansky R, Post RM (1995) Stress and glucocorticoids affect the expression of brain-derived neurotrophic factor and neurotrophin- 3 mRNAs in the hippocampus. J Neurosci 15:1768–1777. 6 Licznerski P, Duman RS (2013) Remodeling of axo-spinous synapses in the pathophysiology and treatment of depression. 251:33–50. 7 Warner-Schmidt JL, Duman RS (2006) Hippocampal neurogenesis: Opposing effects of stress and antidepressant treatment. Hippocampus 16:239–249. 8 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. 9 Marshall J, et al. (2018) Antidepression action of BDNF requires and is mimicked by Gαi1/3 expression in the hippocampus. Proc Natl Acad Sci USA 115:E3549–E3558. 10 Hurowitz EH, et al. (2000) Genomic characterization of the human heterotrimeric G protein alpha, beta, and gamma subunit genes. DNA Res 7:111–120. 11 Autry AE, Monteggia LM (2012) Brain-derived neurotrophic factor and neuropsychiatric disorders. Pharmacol Rev 64:238–258. 12 Pezet S, Malcangio M (2004) Brain-derived neurotrophic factor as a drug target for CNS disorders. Expert Opin Ther Targets 8:391–399. 13 Leal G, Bramham CR, Duarte CB (2017) BDNF and hippocampal synaptic plasticity. Vitam Horm 104:153–195. 14 Leal G, et al. (2017) The RNA-binding protein hnRNP K mediates the effect of BDNF on dendritic mRNA metabolism and regulates synaptic NMDA receptors in hippocampal neurons. eNeuro 4:ENEURO.0268-17.2017. 15 Phillips C (2017) Brain-derived neurotrophic factor, depression, and physical activity: Making the neuroplastic connection. Neural Plast 2017:7260130. 16 Zhou QG, et al. (2011) Hippocampal neuronal nitric oxide synthase mediates the stress-related depressive behaviors of glucocorticoids by downregulating glucocorticoid receptor. J Neurosci 31:7579–7590. 17 Hare BD, Ghosal S, Duman RS (2017) Rapid acting antidepressants in chronic stress models: Molecular and cellular mechanisms. Chronic Stress (Thousand Oaks) 1:10.1177/2470547017697317. 18 McAllister AK, Katz LC, Lo DC (1999) Neurotrophins and synaptic plasticity. Annu Rev Neurosci 22:295–318. 19 Jones KR, Fari~nas I, Backus C, Reichardt LF (1994) Targeted disruption of the BDNF gene perturbs brain and sensory neuron development but not development. Cell 76:989–999. 20 Ernfors P, Lee KF, Jaenisch R (1994) Mice lacking brain-derived neurotrophic factor develop with sensory deficits. Nature 368:147–150. 21 Pozzo-Miller LD, et al. (1999) Impairments in high-frequency transmission, synaptic vesicle docking, and synaptic protein distribution in the hippocampus of BDNF knockout mice. J Neurosci 19:4972–4983. 22 Chao MV (2003) Neurotrophins and their receptors: A convergence point for many signalling pathways. Nat Rev Neurosci 4:299–309. 23 Teng KK, Felice S, Kim T, Hempstead BL (2010) Understanding proneurotrophin actions: Recent advances and challenges. Dev Neurobiol 70:350–359. 24 Li Z, et al. (2010) Caspase-3 activation via mitochondria is required for long-term depression and AMPA receptor internalization. Cell 141:859–871. 25 Jonas EA, et al. (2004) Proapoptotic N-truncated BCL-xL protein activates endogenous mitochondrial channels in living synaptic terminals. Proc Natl Acad Sci USA 101:13590–13595. 26 Quirarte GL, Roozendaal B, McGaugh JL (1997) Glucocorticoid enhancement of memory storage involves noradrenergic activation in the basolateral amygdala. Proc Natl Acad Sci USA 94:14048–14053. 27 Gould E, Woolley CS, McEwen BS (1990) Short-term glucocorticoid manipulations affect neuronal morphology and survival in the adult dentate gyrus. Neuroscience 37:367–375. 28 Du J, et al. (2009) Dynamic regulation of mitochondrial function by glucocorticoids. Proc Natl Acad Sci USA 106:3543–3548. 29 Zhou B, et al. (2016) Facilitation of axon regeneration by enhancing mitochondrial transport and rescuing energy deficits. J Cell Biol 214:103–119. 30 Jonas EA, Porter GA, Alavian KN (2014) Bcl-xL in neuroprotection and plasticity. Front Physiol 5:355. 31 Jonas E (2006) BCL-xL regulates synaptic plasticity. Mol Interv 6:208–222. 32 Su B, Ji YS, Sun XL, Liu XH, Chen ZY (2014) Brain-derived neurotrophic factor (BDNF)-induced mitochondrial motility arrest and presynaptic docking contribute to BDNF-enhanced synaptic transmission. J Biol Chem 289:1213–1226. 33 Verstreken P, et al. (2005) Synaptic mitochondria are critical for mobilization of reserve pool vesicles at Drosophila neuromuscular junctions. Neuron 47:365–378. 34 Li H, et al. (2013) A Bcl-xL-Drp1 complex regulates synaptic vesicle membrane dynamics during endocytosis. Nat Cell Biol 15:773–785. 35 Courchet J, et al. (2013) Terminal axon branching is regulated by the LKB1-NUAK1 kinase pathway via presynaptic mitochondrial capture. Cell 153:1510–1525. 36 Scheetz AJ, Nairn AC, Constantine-Paton M (2000) NMDA receptor-mediated control of protein synthesis at developing synapses. Nat Neurosci 3:211–216.

3744 | www.pnas.org/cgi/doi/10.1073/pnas.1803645115 Licznerski and Jonas Downloaded by guest on October 5, 2021