Open access peer-reviewed chapter

The Role of Brain-Derived Neurotrophic Factor in Psychiatric Disorders

Written By

Sudhiranjan Gupta and Rakeshwar S. Guleria

Submitted: 30 June 2023 Reviewed: 17 July 2023 Published: 21 September 2023

DOI: 10.5772/intechopen.112567

From the Edited Volume

Old Protein, New Medicine - Brain-Derived Neurotrophic Factor

Edited by Oytun Erbaş and İlknur Altuntaş

Chapter metrics overview

55 Chapter Downloads

View Full Metrics

Abstract

Brain derived neurotrophic factor (BDNF) is one of the most extensively studied and widespread growth factors in the brain. BDNF and its receptors are the critical factors having multipotent impact on the central nervous system (CNS). The biological function of BDNF primarily mediated by two receptors, tropomyosin receptor kinase B (TrkB) receptor and p75 neurotrophin receptor. BDNF contributes a pivotal role in neuronal and glial development, modulation and maintaining overall synaptic plasticity of the brain; therefore, widely involved in psychiatric diseases. Current hypotheses indicates that abnormal BDNF level, a vital condition for psychiatric and neurodegeneration diseases are mainly due to the disruption of the BDNF-associated signaling cascades. It is, therefore, crucial to understand how BDNF coordinate the psychiatric diseases in the brain. This review begins with the history of BDNF and its biology in brain homeostasis and focuses on several aspects of BDNF signaling. In addition, the review addresses the impact of BDNF level in diverse neuropsychiatric disorders including major depressive disorder, schizophrenia, bipolar disorder, post-traumatic stress disorder and, possible biological mechanisms of BDNF that may shed new insight for future therapeutic use and drug development.

Keywords

  • BDNF
  • inflammation
  • brain homeostasis
  • brain plasticity
  • psychiatric disorders

1. Introduction

Brain-derived neurotrophic factor (BDNF) is a neurotrophin classified as dimeric polypeptide regulating a wide array of neuronal activities including but not limited to neurogenesis, neuronal growth, differentiation, excitability, and plasticity. BDNF was originally identified by Barde et al. [1] as a factor from cultured embryonic chick which showed survival of sensory neurons. Soon after its discovery, BDNF was recognized and laid a foundation for neuronal plasticity in the adult brain and further observed its’ pivotal role in neuronal activity [2, 3, 4]. Subsequently, BDNF was considered for antidepressant treatments therapy as it was shown that neurotrophins promoted the growth and helped in maturation of neurons [5, 6, 7]. Interestingly, injection of BDNF in the hippocampus elicited antidepressant-like effects in rodents led to advocate a critical role for BDNF in the setting formulating antidepressant drugs [8, 9, 10]. The line of research identified BDNF and its cognate receptor tropomyosin receptor kinase (TrkB, neurotrophic tyrosine kinase receptor, NTRK2) in the hippocampus and cortex suggested antidepressant drug action into neuronal plasticity [11].

BDNF contributed a key role in the development of the nervous system by regulating neuronal development, growth, differentiation, neurogenesis, synaptogenesis, and synaptic plasticity [12, 13, 14]. Moreover, neurodegenerative, and neuropsychiatric diseases appear to be linked with insufficient BDNF level leading to the defects in synaptic plasticity [15, 16]. As a result, strategies to increase the BDNF level in circulation was advocated for therapy in neurological diseases.

This article reviews the current understanding and future directions in BDNF-related research in the central nervous system, with an emphasis on the possible therapeutic application of BDNF in modifying fundamental processes underlying neural disease.

Advertisement

2. BDNF, a neurotrophin family member: synthesis, secretion and function

Nearly three decades earlier discovery of nerve growth factor (NGF) by Rita Levy-Montalcini [17], prompted Yves-Alain Barde searched for a growth factor with similar properties and function like NGF in neurons. The study culminated into a purified protein from pig brain named BDNF [1]. Later, amino acid sequence revealed that BDNF shared a significant homology with NGF along with other members like neurotrophin 3 and neurotrophin 4, together constitute a conserved neurotrophin family [18].

Synthesis and maturation of BDNF is a multistage process, involving formation of several precursor isoforms. BDNF is initially synthesized in the Golgi after cleaving the signal sequence from pre region as a precursor form (pro-BDNF) containing 129 amino acids N-terminal prodomain and a 118 amino acids C-terminal mature domain [19]. The mature domain forms a cysteine knot structure, leading to non-covalent dimerization of the mature domains [20]. When the prodomain is cleaved from intact pro-BDNF, through the actions of proconvertase at a conserved RVRR sequence, the dimeric mature domains are released, and are called mature BDNF, or simply BDNF [21]. Secretion of m-BDNF and pro-BDNF into the extracellular space enables their physiological action (see the diagram, Figure 1).

Figure 1.

Schematic presentation of synthesis and maturation of BDNF. In the intracellular pathway, the pre-pro-BDNF precursor molecule is produced in the endoplasmic reticulum and transported to the Golgi apparatus. During intracellular cleavage, the pre-region is removed, resulting in formation of immature isoform of BDNF called pro-BDNF. Finally, the pro-domain is removed and the mature isoform of BDNF, m-BDNF is produced. The cleavage process is mediated by intracellular proteases, convertases, and furin resulting the release of both pro-BDNF and m-BDNF isoforms into the extracellular space. Here, it is further processed by metalloproteinases 2 and 9 (MMP2 and MMP9), and plasmin.

In neuronal cells, both pro-BDNF and m-BDNF are released following cellular membrane depolarization and maintained a dynamic balance [22, 23, 24]. Both isoforms are important in neuronal function in the brain, but mature-BDNF (m-BDNF) appeared to offer neurogenesis, neuroprotection, synaptic plasticity, and synaptic function in neurons [25, 26]. The m-BDNF is axonally delivered into axon vesical terminals followed by the secretion into axonal cleft [22]. Mechanistically, BDNF requires to bind its’ partner/receptor, Tr, located both pre- and post-synaptic membrane, to complete its function. BDNF is highly conservative and is expressed as a single gene, Bdnf transcript and is dynamically regulated and showed cell-specific neural activity. The human Bdnf gene, a ~ 70 kb, is in the chromosome 11 consisting of 11 exons (I-IX along with Vh and VIIIh) in the 5′ end and 9 promoters in tissues and brain regions [27, 28]. Apart from the above-mentioned BDNF isoforms, the function of BDNF is potentially affected by single nucleotide polymorphism of methionine (Met) to valine (Val) substitution at 66th position of Bdnf gene.

Considering BDNF neuronal function, it is more appreciated as differentiation factor than survival neurotrophin [29, 30]. In addition to synaptic transmission, BDNF elicits long-term potentiation in hippocampus and modulate neuronal circuit function [31]. Moreover, changes in BDNF level in rodent models demonstrated aberrant function in hippocampal regions, including impaired memory, aggression, and hyperphagia [32].

Advertisement

3. BDNF receptors and intracellular signaling

BDNF signals are mediated by TrkB receptor and p75 neurotrophin receptor. BDNF binds with high affinity with TrkB, a tyrosine kinase receptor family, and the p75 neurotrophin receptor (p75 NTR), a member of the tumor necrosis factor (TNF) receptor family and low with p75 receptor. The TrkB is widely expressed in brain including cortex, hippocampus and in spinal cord nuclei [33]. It is noted that the mature BDNF binds to TrkB whereas pro-BDNF binds to p75NTR. The pro-BDNF/p75NTR signaling primarily promoting synaptic elimination by activating c-Jun N-terminal Kinase (JNK) pathway and triggers apoptosis. Other family members of Trk are TrkA which is specific to NGF [34] and TrkC which binds other neurotrophins [35]. This review will focus TrkB and its’ signaling.

Activation of BDNF begins by binding to TrkB, and dimerizing and activating intrinsic kinase cascade before going to autophosphorylation. The BDNF/TrkB complex gets internalized into the neuron and serves as a docking site for diverse signaling platforms, protein phosphorylation and secondary signaling events [36, 37]. Next, the binding of BDNF to TrkB receptor, BDNF/TrkB in complex, leads to phosphorylation and translocation of TrkB into cellular membrane lipid rafts, and activating diverse important intracellular signaling cascades for performing cellular functions that include mitogen-activated protein kinase/extracellular signal-related kinase (MAPK/ERK), guanosine triphosphate hydrolases (GTP-ases) of the Ras homolog (Rho) gene and phospholipase C-γ (PLC-γ), phosphatidylinositol 3-kinase/protein kinase B (PI3K/AKT) pathways [38, 39, 40, 41]. It is evidenced that PI3K/AKT pathway contributed to synaptic plasticity and cell survival or antiapoptotic activity response by modulating N-methyl-D-aspartate receptor (NMDAR) [40, 42]. Furthermore, BDNF-dependent neuroprotection is mediated via NMDAR/Ca2+ synaptic signaling resulting eliminating glutamatergic toxicity and preventing mitochondrial dysfunction and cellular apoptosis [43, 44]. The PLC g-dependent signaling triggers Ca2+-calmodulin-dependent protein kinase (CAMK) and protein kinase C (PKC) to stimulate actin/microtubule synthesis and enhance synaptic plasticity and neuronal fiber growth [40, 45, 46]. The MAPK/Ras signaling regulates neural differentiation [45]. The ERK ½ and cAMP response element-binding protein (CREB) activation are necessary for cytoskeleton protein synthesis for dendritic growth and branching [40, 47]. In summary, the participation of BDNF in several physiological roles in the brain involves different signaling and is pivotal in maintaining a dynamic balance between the stimulus and its’ function. A diagrammatic presentation of BDNF receptor and signaling is shown in Figure 2.

Figure 2.

BDNF signaling cascade. The BDNF is primarily transcribed as a precursor (pro-BDNF) which is later cleaved intra or extracellularly into mBDNF. The pro-BDNF exhibits affinity to sortilin and p75NTR receptors leading to the activation of nuclear factor κB (NF-κB), RhoA and JNK signaling pathways. The functional outcome of theses pathways includes neuronal survival, development, and apoptosis. The mBDNF showed highest affinity towards TrkB receptors. The mBDNF/TrkB complex triggers signaling pathways linked to phosphatidylinositol 3-kinase (PI3K), phospholipase C-γ (PLC-γ) and mitogen activated protein kinase (MAPK) via CREB. The pathways are involved in dendritic growth and branching, synaptic plasticity, and cytoskeleton protein activation.

Advertisement

4. BDNF and brain homeostasis

Homeostasis is a fundamental process and equates to a dynamic balance between interdependent element and the physiological function in the organ of a living system. BDNF plays a significant role in neuronal plasticity in the central and peripheral nervous system [48]. BDNF is expressed throughout the development and adulthood in neurons of the brain and contributing a critical role in many physiological functions. One of the functions is energy homeostasis in the hypothalamus. Energy homeostasis is a complex gets interaction between the brain and peripheral tissues. Neuronal circuitry in the hypothalamus and hindbrain contributes a critical role in orchestrating the peripheral signals associated with energy storage by regulating nutrient intake and energy expenditure. BDNF is synthesized in several regions of hypothalamus including ventromedial hypothalamic nucleus (VMH), the dorsomedial hypothalamic nucleus (DMH), the paraventricular nucleus (PVH) and the lateral hypothalamic area (LH) [49, 50]. In particular, the energy balance is reported to be in the PVH region as evidenced by loss of body weight by injecting BDNF in this region [51]. The report showed that decrease in food intake resulted in increased resting metabolic rate, partly due to upregulation of uncoupling protein 1 (UPC1) in the brown adipose tissue [51]. Hypothalamic injection of BDNF promotes switching white adipose tissue to brown adipose tissue via sympathetic neuron activation and accelerates UCP-1 expression [52, 53]. This is an example of the role of BDNF in increasing energy expenditure by modulating metabolic rate and temperature. The data indicated that BDNF enhanced energy expenditure suggesting an anorexigenic function [52]. Another finding attested the role of BDNF in thermogenic regulation in lateral hypothalamus [54]. On the contrary, deletion of Bdnf gene caused hyperphagia, decreased locomotor activity and impaired thermoregulation [54]. Moreover, it is evident that mutation in the Bdnf gene or its receptor (TrkB) leads to obesity in mice [55, 56]. The Bdnf gene mutation data is corroborated with hyperphagia and impaired cognitive functions in humans [57, 58, 59, 60, 61, 62]. Together, it is suggested that PVH region is critical in energy balance in the brain.

In addition, BDNF plays a key role in energy management in non-neuronal cells. Selective ablation of BDNF in liver cells in mice showed reduction in hyperglycemia and hyperinsulinemia caused by a high fat diet [63]. Compromised BDNF signaling is also linked with obesity and the metabolic syndrome in humans [64]. Furthermore, BDNF administration reduced serum glucose and insulin in obese db/db mice or improvement of glucose tolerance compared to their vehicle treated counterparts [65, 66]. The underlying molecular mechanism may be the interaction of BDNF with glucagon like peptide 1 (GLP1). Gotoh et al. showed that administration of BDNF decreased the portal glucagon level and did not show any effect on insulin [67]. It is also observed that the intraportal administration of GLP-1 increases BDNF levels in the pancreas and reduces glucagon secretion [67]. Recent study also suggested a role on pancreatic-islet-expressed TrkB to promote peripheral insulin secretion [68]. In addition to BDNF and TrkB, the pro-BDNF receptor, p75NTR is suggested to play a role in glucose homeostasis and insulin sensitivity. Conditional knockout of p75NTR showed improvements of glucose and insulin tolerance in adipose and skeletal muscle [68, 69]. Regarding signaling context of BDNF and metabolic homeostasis, it is yet to be defined which receptor mediated action is more appropriate. The rational lies that pro-BDNF exclusively binds to p75NTR and appeared to show an opposite effect to BDNF-TrkB activity [70]. It established that a single nucleotide polymorphism (SNP) in pro domain of BDNF (Val66Met) is linked with neuropsychiatric disorders in humans and seemed to function through p75NTR [71]. The SNP (Val66Met) variant indicated increased appetite in mice via p76NTR [72], along with alteration of anxiety and anorexic-related behavior [73, 74]. The data may suggest a unique control of energy balance in food intake and anxiety. Finally, the downstream signaling between pro-BDNF and mature BDNF are quite distinct and may appeared to reflect different outcome in neuronal cells. TrkB promotes MAPK/ERK, PI3K, and PLCg1, pathways, while p75NTR promotes JNK and Rho pathways [36, 41, 75, 76, 77].

Advertisement

5. BDNF and psychiatric diseases and disorders

We often use the term disorder and diseases in psychiatric illness. There is a subtle difference exists between them however, they are considered as mental illness. The term disease defines an involuntary response of biological, physiological, or pathological consequences of illness and, the underlying cause can be measured. The disorder defines disturbance of normal physical or mental health status and is a collection of signs and symptoms closely associated with specific disease. In general speaking, we can say that all diseases are disorders but not all disorders are diseases.

BDNF is one of the most widely studied neurotrophin signaling molecules in the brain responsible for neurite growth, maturation of synapses during development, and synaptic plasticity. We have discussed BDNF’s biology, receptor alignment for signaling events in the brain. Essentially, BDNF-TrkB signaling, and its intermediate proteins contributed a critical role in different phases of synaptic development and neuroplasticity in the brain [78]. Moreover, BDNF regulates learning and memory process in young and adult humans [79]. Therefore, aberrant expression or imbalance in BDNF level and its cognate TrkB receptor are associated with many psychiatric disorders (diseases) and neurodegenerative diseases. In addition, anomaly of BDNF level and signaling are linked to diverse cardiovascular, metabolic, and inflammatory diseases [80, 81, 82, 83, 84, 85]. This section will discuss the contribution of BDNF in brain illness or psychological diseases (disorders) including major depressive disorder (MDD), schizophrenia (SZ), bipolar disorder (BD) and post-traumatic stress disorder (PTSD).

Advertisement

6. BDNF and MDD

BDNF is well studied molecule in MDD. Eisch et al reported that an increase level of BDNF in the ventral tegmental area (VTA)-nucleus accumbens (NAc) region contributed the onset of depression in rats [86]. A following mechanistic study by the same group using viral-mediated mesolimbic dopamine-specific BDNF knockdown determined the pivotal role of BDNF in depression like behavior [87]. Interestingly, reduced BDNF in cornu ammonis (CA3) and dentate gyrus (DG) of the hippocampus and prefrontal cortex (PFC), resulting in depression-like behavior in mice [88]. Furthermore, targeted deletion of BDNF using NSE-tTA x TetOp-Cre line in the VTA area determined that BDNF in the DG was essential for therapeutic intervention as an antidepressant [89]. Similarly, reduced BDNF protein levels were observed in patients with MDD compared with the healthy control [90, 91]. Taken together, these findings suggest that BDNF acts within the VTA-NAc pathway to induce a depression-like phenotype, whereas in the hippocampus and PFC it produces antidepressant-like effects [92]. It is further observed that TrkB, the receptor for BDNF played a role in MDD. Patient with MDD showed elevated level of TrkB compared to the healthy control [93, 94]. However, it is unclear regarding the role of the partners in MDD and may be the focus of future investigation.

Epigenetic modification like DNA methylation is frequently studied in Bdnf gene and BDNF exon I and IV promoters. A methylation profile in CpG island of exon I of BDNF promoter showed differential pattern of methylation that can distinguish between major depression vs. and healthy controls and suggested to be a good biomarker for MDD [95]. But exon IV did not show any changes. A similar study reported higher methylation of BDNF exon I promoter in patients with MDD [96]. This study further showed reduced methylation pattern with antidepressants treatment [96]. Interestingly, patient with MDD showed poor treatment response when methylation of CpG site −87 of BDNF exon IV promoter was lacking [97].

An association between BDNF Val66Met polymorphism and MDD is extensively studied. Meta analyses revealed that there is no association between Val66Met polymorphism and MDD (depression) [98, 99, 100]. However, few studies have indicated that BDNF Val66Met polymorphism moderated the relationship between stress and depression [100, 101, 102, 103].

Advertisement

7. BDNF and Schizophrenia (SZ)

Schizophrenia is a complex heterogenous disease characterized by multiple symptoms such as hallucinations, social avoidance, withdrawal, paranoia, cognitive deficit, and disorganized thought [104]. The role of BDNF in SZ is well studied because BDNF is involved in neurotransmission. In general, BDNF level is reduced in SZ patients [105, 106] and study has shown further that serum BDNF is positively correlated with antipsychotic drug (clozapine) [107]. This is an interesting finding for a therapeutic purpose. However, recent evidence implicated that BDNF mRNA expression remained unchanged in SZ patients compared to healthy control in postmortem brain samples [108].

Reports are emerging regarding epigenetic mechanism in Bdnf gene and development of SZ [109]. Epigenetic mechanism encompasses DNA methylation, histone modification, chromatin remodeling and DNA methylation is widely studied in SZ [109, 110]. A significant positive correlation was observed in BDNF gene methylation in patients with SZ compared to healthy controls [111]. Another study showed higher methylation level at BDNF promoter compared to controls [112]. Moreover, a differentially methylated CpGs has been identified in SZ patients of postmortem human brains [113]. Moreover the Val66Met SNP on the Bdnf gene has implicated schizophrenia incidence and a recent meta-analysis provided evidence that there was an association between brain volume alterations and variations on the Val66Met SNP in patients of SZ [114, 115, 116]. While studies have shown a positive correlation between reduced level of BDNF and SZ episode, but have not evaluated the role of demographic characteristics such as age, gender, race, and education. Therefore, adequate meta-analysis including demographic factors should be added and warranted further investigation.

Advertisement

8. BDNF and bipolar disorder (BD)

Bipolar disorder is a multifactorial psychiatric disorder characterized by mood fluctuation or instability, depressive, manic episode, and euthymic states [117, 118]. BD makes a distinct category in Diagnostic and Statistical Manual of Mental Disorders, 5th edition into BD I, BD II based on severity of manic episodes [119]. The thirst for potential biomarker in BP is emerging and BDNF is extensively studied in this area. In 2005, Laske et al. first reported reduced BDNF level in the serum of manic and major depressed patients compared to healthy control [120]. Since, then several studies have been conducted in BD and majority of the studies suggested a decline level of peripheral BDNF and considered it as a marker [121, 122, 123, 124, 125], however, BDNF levels were not different in euthymia when compared to controls [126]. Furthermore, at transcription level, BDNF mRNA showed downregulation in postmortem brains of both manic and depressive subjects [127, 128]. Antipsychotic drugs like mood stabilizers are frequently prescribed for manic or depressive disorder but the study did not show any improvement of BDNF level in four weeks treatment [122]. However, another study of sixteen-week follow-up, using extended-release quetiapine showed increase in BDNF levels, but decreases with time in a manic/mixed episode [129].

A common genetic variation in Bdnf gene, the Val66Met, is established as a common platform linked with reduced secretion of BDNF and is associated with many neuropsychiatric disorders and BD is not an exception. Earlier finding suggested an association between BDNF Val66Met polymorphism and BP [130, 131] but recent meta-analyses showed opposite results [132, 133]. Therefore, more data are warranted to determine the role of Val66Met polymorphism in BD.

Epigenetic modulation is well documented in psychiatric disorders and a positive correlation is shown in CpG methylation in BDNF promoter and BD subjects [134, 135, 136]. Alterations in DNA methylation patterns in patients with BD have been extensively investigated for the past years, and possibly recognize a potential biomarker [137, 138, 139]. It may be the case that DNA methylation alters the differences in BDNF level and contributed in part in BD, so, targeting BDNF methylation could be strategy to treat BD.

Advertisement

9. BDNF and post-traumatic stress disorder

Post-traumatic stress disorder (PTSD) is a debilitating psychiatric disorder characterized by hyperarousal, re-experiencing, negative emotions, increased anxiety, and fearful memories following exposure to severe trauma [119]. The role of BDNF in PTSD is emerging. In 2009, a small human study was conducted in University of Pisa, Italy where they recruited 18 drug naïve PTSD patients (12 women and 6 men) with no psychiatric comorbidity and 18 healthy controls in outpatients’ facility. The finding showed reduced level of BDNF in the plasm compared with healthy control [140]. War Veterans have continuously suffered from PTSD and cognitive deficit caused by traumatic brain injury. The possible first combat Veteran study aiming BDNF as a marker in PTSD was investigated in Croatia, 2022. The results revealed a marked reduction in plasma BDNF in Veterans with PTSD and mild cognitive impairment compared with healthy controls [141]. The epigenetic influence in BDNF played a critical role in psychiatric disorders including PTSD, as few studies were conducted to investigate DNA methylation in CpG island and Val66Met polymorphisms. A study was conducted using US military service members deployed in the Middle east for Operation Iraqi Freedom (OIF)/Operation Enduring Freedom (OEF) with PTSD showing a significant association between BDNF Val66 Met genotype and traumatic stress in post deployment [142]. Another study of Vietnam war active service members from South Korea showed an association between higher DNA methylation in BDNF promoter in PTSD subjects suggesting a biomarker of PTSD [143]. Interestingly, another study of Vietnam war Veterans by the Australian or New Zealand Defense Force showed that PTSD was associated with decreased methylation at three BDNF CpG sites [144]. Furthermore, it was observed that BDNF Val66Met was linked with differential Bdnf expression in the peripheral tissues [144]. Another study supported the finding that methylation of CpG island (CpG1, CpG 7 and CpG 18) in BDNF promoter was closely related to PTSD and suggested as a biomarker to PTSD [145].

Although studies have shown a positive correlation between BDNF level and Val66Met polymorphism in PTSD, there were reports that showed the opposite effect. There was a report showing no relationship between BDNF Val66Met and PTSD in victims of urban violence [146]. In addition, two case studies (small sample size) failed to establish the association between Val66Met and PTSD [147, 148]. Moreover, an elevated level of BDNF was observed in patients with PTSD suffering from trauma [149]. A meta-analysis showed that BDNF level is increased in PTSD patients compared to healthy subjects [150]. A discrepancy was noted in OEF/OIF Veteran study. Recently, Wu et al. reported for the first time that a higher serum level of BDNF in chronic combat PTSD Veterans independent of symptom severity [151]. These reports contradict previous findings.

Together it appeared that genetic variants of Bdnf gene and PTSD did not provide any conclusive relationship. The higher and lower value of BDNF were possibly observed due to heterogenous population or low percentage of homozygous Met alleles. More longitudinal and follow-up studies are necessary to make a definitive conclusion.

Advertisement

10. BDNF-miRNAs-psychiatric disorders

The miRNAs are non-coding RNAs, a new class of epigenetic modulators emerging as an attractive molecule for therapeutic intervention. The miRNAs are small 21–23 nucleotides that have the capability to inhibit mRNA and protein resulting in gene regulation [152]. Literature search showed 2844 articles have been published where miRNAs were associated with psychiatric diseases. Interestingly, BDNF-miRNA axis in psychiatric diseases showed 131 reports indicating therapeutic potential of BDNF. Recent studies indicated that several miRNAs target 3′ UTR of Bdnf gene modulated the function associated with psychiatric disorders [153, 154, 155, 156, 157, 158]. In rodent model of anxiety disorder and schizophrenia, miR-124a regulated anxiety like behavior by targeting Bdnf gene [159] and miR-148b is implicated in regulating Bdnf gene in methylazoxymethanol acetate model [160]. In mouse model of PTSD, a set of miRNAs, miR-15a-5p, miR-497a-5p, miR-511-5p and let-7d-5p were shown to be associated with Bdnf and FKBP5, the two key PTSD-linked genes [157]. Moreover, a prolong stress induced rat PTSD model, miR-142-5p is shown to be upregulated in amygdala with a target gene, Npas4 which was reduced [161]. The inhibition of miR-142-5p appeared to reduce the PTSD symptoms by restoring Npas4 and BDNF level suggesting a crucial link between them. In BD condition, a human cohort study was conducted and revealed an association between miR-206 and BDNF polymorphism [162]. Another study showed a panel of miRNAs, miR-7-5p, miR-221-5p and miR-370-5p that are involved in BD II patients by modulating BDNF level [163].

In summary, the data showed promising direction in miRNA-BDNF-axis modulation in psychiatric disorders. However, a strong clinical correlation regarding miRNA-BDNF needs to be established for the development of new diagnostic and therapeutic application to mitigate the cognitive deficit.

11. Conclusion

BDNF is well studied in major psychiatric disorders or diseases. Modern techniques provided us new insights regarding BDNF’s role in psychiatric disease progression and treatment responses. The dysregulation of BDNF/pro-BDNF and its receptors TrkBs resulting in a cascade of neuropathophysiological events leading to the impairment of synaptic plasticity and cognitive deficit. Several lines of evidence support the notion that BDNF is nodal mediator across an array of neuropsychiatric disorders. It is further to make a note that many second-generation antipsychotic drugs showed some promise in providing neuroprotection by enhancing BDNF level, however, a definitive conclusion cannot be made based on few medications. Future investigation including using small molecule compound (mimetics or agonists) for enhancing BDNF synthesis and gene therapy using nanoparticle mediated encapsulation of BDNF, is necessary to extend this efficacy at therapeutic standpoint. Peripheral BDNF level is used as a biomarker in many psychiatric disorders, however, in some cases like MDD, it showed disagreement. This may be due to heterogenous nature and epigenetic modifications that contributed significantly for making a universal conclusion. Nonetheless, it helped to pave the way for better understanding the role of BDNF deep inside human brains. Future studies are warranted to uncover the mechanism of methylation and SNPs of Bdnf gene for better therapeutic treatment.

Acknowledgments

This material is the result of work with resources and the use of facilities at the VISN17 Center of Excellence for Research on Returning War Veterans and the Central Texas Veterans Health Care System (CTVHCS). The authors would like to acknowledge Richard W. Seim, Ph.D., Director of the VISN 17 Center of Excellence for Research on Returning War Veterans, (Texas Waco, USA) for his support. The views expressed herein are those of the authors and do not necessarily reflect the official policy or position of the Department of Veterans Affairs or the United States Government.

Conflict of interest

The authors state that they do not have any conflict of interest.

Funding

The present study was supported by VISN17 Center of Excellence’s internal funds to Biomarkers & Genetics Core.

Notes/thanks/declaration

None.

References

  1. 1. Barde YA, Edgar D, Thoenen H. Purification of a new neurotrophic factor from mammalian brain. The EMBO Journal. 1982;1:549-553. DOI: 10.1002/j.1460-2075. 1982.tb01207. x
  2. 2. Zafra F, Hengerer B, Leibrock J, Thoenen H, Lindholm D. Activity dependent regulation of BDNF and NGF mRNAs in the rat hippocampus is mediated by non-NMDA glutamate receptors. The EMBO Journal. 1990;9:3545-3550. DOI: 10.1002/j.1460-2075. 1990.tb07564. x
  3. 3. Dugich-Djordjevic MM, Tocco G, Lapchak PA, Pasinetti GM, Najm I, Bundry M, et al. Regionally specific and rapid increases in brain-derived neurotrophic factor messenger RNA in the adult rat brain following seizures induced by systemic administration of kainic acid. Neuroscience. 1992;47:303-315. DOI: 10.1016/0306-4522(92)90246-x
  4. 4. Thoenen H. Neurotrophins and neuronal plasticity. Science. 1995;270:593-598. DOI: 10.1126/science.270.5236.593
  5. 5. Lindsay RM, Wiegand SJ, Anthony Altar CA, DiStefano P. Neurotrophic factors: From molecule to man. Trends in Neurosciences. 1994;17:182-190. DOI: 10.1016/0166-2236(94)90099-x
  6. 6. Lindvall O, Kokaia Z, Bengzon J, Elmer E, Kokaia M. Neurotrophins and brain insults. Trends in Neurosciences. 1994;17:490-496. DOI: 10.1016/0166-2236(94)90139-2
  7. 7. Siuciak JA, Anthony Altar CA, Wiegand SJ, Lindsay RM. Antinociceotive effect of brain-derived neurotrophic factor and neurotrophin-3. Brain Research. 1994;633:326-330. DOI: 10.1016/0006-8993(94)91556-3
  8. 8. Shirayama Y, Chen AC, Nakagawa S, Russell DS, Duman RS. Brain-derived neurotrophic factor produces antidepressant effects in behavioral models of depression. The Journal of Neuroscience. 2002;22:3251-3261. DOI: 10.1523/JNEUROSCI.22-08-03251.2002
  9. 9. Siuciak JA, Lewis DR, Wiegand SJ, Lindsay RM. Antidepressant-like effect of brain-derived neurotrophic factor (BDNF). Pharmacology, Biochemistry, and Behavior. 1997;56:131-137. DOI: 10.1016/S0091-3057(96)00169-4
  10. 10. Altar CA. Neurotrophins and depression. Trends in Pharmacological Sciences. 1999;20:59-61. DOI: 10.1016/s0165-6147(99)01309-7
  11. 11. Nibuya M, Morinobu S, Duman RS. Regulation of BDNF and trkB mRNA in rat brain by chronic electroconvulsive seizure and antidepressant drug treatments. The Journal of Neuroscience. 1995;15:7539-7547. DOI: 10.1523/JNEUROSCI.15-11-07539.1995
  12. 12. Tapia-Arancibia L, Aliaga E, Silhol M, Arancibia S. New insights into brain BDNF function in normal aging and Alzheimer disease. Brain Research Reviews. 2008;59:201-220. DOI: 10.1016/j.brainresrev.2008.07.007
  13. 13. Lee J, Duan W, Mattson MP. Evidence that brain-derived neurotrophic factor is required for basal neurogenesis and mediates, in part, the enhancement of neurogenesis by dietary restriction in the hippocampus of adult mice. Journal of Neurochemistry. 2002;82:1367-1375. DOI: 10.1046/j.1471-4159.2002.01085. x
  14. 14. Lu B, Nagappan G, Lu Y. BDNF and synaptic plasticity, cognitive function, and dysfunction. In: Lewin G, Carter B, editors. Neurotrophic Factors. Handbook of Experimental Pharmacology. Berlin/Heidelberg, Germany: Springer; 2014. pp. 223-250. DOI: 10.1007/978-3-642-45106-5_9
  15. 15. Zuccato C, Cattaneo E. Brain-derived neurotrophic factor in neurodegenerative diseases. Nature Reviews. Neurology. 2009;5:311-322. DOI: 10.1038/nrneurol.2009.54
  16. 16. Autry AE, Monteggia LM. Brain-derived neurotrophic factor and neuropsychiatric disorders. Pharmacological Reviews. 2012;64:238-258. DOI: 10.1124/pr.111.005108
  17. 17. Levi-Montalcini R, Hamburger V. Selective growth stimulating effects of mouse sarcoma on the sensory and sympathetic nervous system of chick embryo. The Journal of Experimental Zoology. 1951;116:321-361. DOI: 10.1002/jez.1401160206
  18. 18. Leibrock J, Lottspeich F, Hohn A, Hofer M, Hengerer B, Masiakowski P, et al. Molecular cloning and expression of brain-derived neurotrophic factor. Nature. 1989;341:149-152. DOI: 10.1038/341149a0
  19. 19. Radziejewski C, Robinson RC, DeStefano PS, Taylor JW. Dimeric structure and conformational stability of brain-derived neurotrophic factor and neurotrophin-3. Biochemistry. 1992;31:4431-4436. DOI: 10.1021/bi00133a007
  20. 20. Mowla SJ, Farhadi HF, Pareek S, Atwal JK, Morris SJ, Seidah NG, et al. Biosynthesis and post-translational processing of the precursor to brain-derived neurotrophic factor. The Journal of Biological Chemistry. 2001;276(16):12660-12666. DOI: 10.1074/jbc.M008104200
  21. 21. Foltran RB, Diaz SL. BDNF isoforms: A round trip ticket between neurogenesis and serotonin? Journal of Neurochemistry. 2016;138(2):204-221. DOI: 10.1111/jnc.13658
  22. 22. Dieni S, Matsumoto T, Dekkers M, Rauskolb S, Ionescu MS, Deogracias R, et al. Barde YA BDNF and its pro-peptide are stored in presynaptic dense core vesicles in brain neurons. The Journal of Cell Biology. 2012;196(6):775-788. DOI: 10.1083/jcb.201201038
  23. 23. Yang J, Harte-Hargrove LC, Siao CJ, Marinic T, Clarke R, Ma Q , et al. proBDNF negatively regulates neuronal remodeling, synaptic transmission, and synaptic plasticity in hippocampus. Cell Reports. 2014;7(3):796-806. DOI: 10.1016/j.celrep.2014.03.040
  24. 24. Lessmann V, Brigadski T. Mechanisms, locations, and kinetics of synaptic BDNF secretion: An update. Neuroscience Research. 2009;65:11-22. DOI: 10.1016/j.neures.2009.06.004
  25. 25. Lee J, Seroogy KB, Mattson MP. Dietary restriction enhances neurotrophins expression and neurogenesis in the hippocampus of adult mice. Journal of Neurochemistry. 2002;80:539-547. DOI: 10.1046/j.0022-3042.2001.00747. x
  26. 26. Brigadski T, Leßmann V. The physiology of regulated BDNF release. Cell and Tissue Research. 2020;382(1):15-45. DOI: 10.1007/s00441-020-03253-2
  27. 27. Pruunsild P, Kazantseva A, Aid T, Palm K, Timmusk T. Dissecting the human BDNF locus: Bidirectional transcription, complex splicing, and multiple promoters. Genomics. 2007;90:397-406. DOI: 10.1016/j.ygeno.2007.05.004
  28. 28. Adachi N, Numakawa T, Richards M, Nakajima S, Kunugi H. New insight in expression, transport, and secretion of brain-derived neurotrophic factor: Implications in brain-related diseases. World Journal of Biological Chemistry. 2014;5:409-428. DOI: 10.4331/wjbc. v5.i4.409
  29. 29. Bergami M, Rimondini R, Santi S, Blum R, Gotz M, Canossa M. Deletion of TrkB in adult progenitors alters newborn neuron integration into hippocampal circuits and increases anxiety-like behavior. Proceedings of the National Academy of Sciences of the United States of America. 2008;105(40):15570-15575. DOI: 10.1073/pnas.0803702105
  30. 30. Vilar M, Mira H. Regulation of neurogenesis by neurotrophins during adulthood: Expected and unexpected roles. Frontiers in Neuroscience. 2016;10:26. DOI: 10.3389/fnins.2016.00026
  31. 31. Lu Y, Christian K, Lu B. BDNF: A key regulator for protein-synthesis dependent LTP and long-term memory? Neurobiology of Learning and Memory. 2008;89(3):312-323. DOI: 10.1016/j.nlm.2007.08.018
  32. 32. Lyons WE, Mamounas LA, Ricaurte GA, Coppola V, Reid SW, Bora SH, et al. Brain-derived neurotrophic factor-deficient mice develop aggressiveness and hyperphagia in conjunction with brain serotonergic abnormalities. Proceedings of the National Academy of Sciences of the United States of America. 1999;96:15236-15244. DOI: 10.1073/pnas.96.26.15239
  33. 33. Molteni R, Barnard RJ, Ying Z, Roberts CK, Gomez-Pinilla F. A high-fat, refined sugar diet reduces hippocampal brain-derived neurotrophic factor, neuronal plasticity, and learning. Neuroscience. 2002;112:803-814. DOI: 10.1016/s0306-4522(02)00123-9
  34. 34. Kaplan DR, Hempstead BL, Martin-Zanca D, Chao MV, Parada LF. The trk proto-oncogene product: A signal transducing receptor for nerve growth factor. Science. 1991;252:554-558. DOI: 10.1126/science.1850549
  35. 35. Barbacid M. The trk family of neurotrophin receptors. Journal of Neurobiology. 1994;25:1386-1403. DOI: 10.1126/science.1850549
  36. 36. Yoshii A, Constantine-Paton M. Postsynaptic BDNF-TrkB signaling in synapse maturation, plasticity, and disease. Developmental Neurobiology. 2010;70:304-322. DOI: 10.1002/dneu.20765
  37. 37. Chao M, Hempstead B. p75 and Trk: A two-receptor system. Trends in Neurosciences. 1995;18(7):321-326
  38. 38. Mohammadi A, Amooeian VG, Rashidi E. Dysfunction in brain-derived neurotrophic factor signaling pathway and susceptibility to schizophrenia. Parkinson’s and Alzheimer’s diseases. Current Gene Therapy. 2018;18:45-63. DOI: 10.2174/1566523218666180302163029
  39. 39. Suzuki S, Numakawa T, Shimazu K, Koshimizu H, Hara T, Hatanaka H, et al. BDNF-induced recruitment of TrkB receptor into neuronal lipid rafts: Roles in synaptic modulation. The Journal of Cell Biology. 2004;167(6):1205-1215. DOI: 10.1083/jcb.200404106
  40. 40. Gonzalez A, Moya-Alvarado G, Gonzalez-Billaut C, Bronfman FC. Cellular and molecular mechanisms regulating neuronal growth by brain-derived neurotrophic factor (BDNF). Cytoskeleton (Hoboken). 2016;73(10):612-616. DOI: 10.1002/cm.21312
  41. 41. Huang EJ, Reichardt LF. Trk receptors: Roles in neuronal signal transduction. Annual Review of Biochemistry. 2003;72:609-642. DOI: 10.1146/annurev.biochem.72.121801.161629
  42. 42. Baydyuk M, Xu B. BDNF signaling and survival of striatal neurons. Frontiers in Cellular Neuroscience. 2014;8:254. DOI: 10.3389/fncel.2014.00254
  43. 43. Zuccato C, Cattaneo. Role of brain-derived neurotrophic factor in Huntington disease. Progress in Neurobiology. 2007;81:294-311. DOI: 10.1016/j.pneurobio.2007.01.003
  44. 44. Zhang SJ, Zou M, Lu L, Lau D, Ditzel DAW, Delucinge-Vivier C, et al. Nuclear calcium signaling controls expression of a large gene pool: Identification of a gene program for acquired neuroprotection induced by synaptic activity. PLoS Genetics. 2009;5(8):e1000604. DOI: 10.1371/journal.pgen.1000604
  45. 45. Reichardt LF. Neurotrophin-regulated signalling pathways. Philosophical Transactions of the Royal Society of London. Series B, Biological Sciences. 2006;361(1473):1545-1564. DOI: 10.1098/rstb.2006.1894
  46. 46. Minichiello L. TrkB signalling pathways in LTP and learning. Nature Reviews. Neuroscience. 2009;10(12):850-860. DOI: 10.1038/nrn2738
  47. 47. Kwon M, Fernandez JR, Zegarek GF, Lo SB, Firestein BL. BDNF-promoted increases in proximal dendrites occur via CREB dependent transcriptional regulation of cypin. The Journal of Neuroscience. 2011;31(26):9735-9745. DOI: 10.1523/JNEUROSCI.6785-10.2011
  48. 48. Chao MV. Neurotrophin signaling in health and disease. Clinical Science (London, England). 2006;110:167-173. DOI: 10.1042/CS20050163
  49. 49. Noble EE, Billington CJ, Kotz CM, Wang C. The lighter side of BDNF. American Journal of Physiology. Regulatory, Integrative and Comparative Physiology. 2011;300:R1053-R1069. DOI: 10.1152/ajpregu.00776.2010
  50. 50. Podyma B, Parekh K, Güler AD, Deppmann CD. Metabolic homeostasis via BDNF and its receptors. Trends in Endocrinology and Metabolism. 2021;32:488-499. DOI: 10.1016/j.tem.2021.04.005
  51. 51. Wang C, Bomberg E, Billington C, Levine A, Kotz CM. Brain-derived neurotrophic factor in the hypothalamic paraventricular nucleus increases energy expenditure by elevating metabolic rate. American Journal of Physiology. Regulatory, Integrative and Comparative Physiology. 2007;293:R992-R1002. DOI: 10.1152/ajpregu.00516.2006
  52. 52. Tsuchida A, Nonomura T, Ono-Kishino M, Nakagawa T, Taiji M, Noguchi H. Acute effects of brain-derived neurotrophic factor on energy expenditure in obese diabetic mice. International Journal of Obesity and Related Metabolic Disorders. 2001;25:1286-1293. DOI: 10.1038/sj.ijo.0801678
  53. 53. Cao L, Choi EY, Liu X, Martin A, Wang C, Xu X, et al. White to brown fat phenotypic switch induced by genetic and environmental activation of a hypothalamic-adipocyte axis. Cell Metabolism. 2011;14:324-338. DOI: 10.1016/j.cmet.2011.06.020
  54. 54. You H, Chu P, Guo W, Lu B. A subpopulation of Bdnf-e1-expressing glutamatergic neurons in the lateral hypothalamus critical for thermogenesis control. Molecular Metabolism. 2020;31:109-123. DOI: 10.1016/j.molmet.2019.11.013
  55. 55. An JJ, Liao G-Y, Kinney CE, Sahibzada N, Xu B. Discrete BDNF neurons in the paraventricular hypothalamus control feeding and energy expenditure. Cell Metabolism. 2015;22:175-188. DOI: 10.1016/j.cmet.2015.05.008
  56. 56. Rios M, Fan G, Fekete C, Kelly J, Bates B, Kuehn R, et al. Conditional deletion of brain-derived neurotrophic factor in the postnatal brain leads to obesity and hyperactivity. Molecular Endocrinology. 2001;15:1748-1757. DOI: 10.1210/mend.15.10.0706
  57. 57. Liao GY, An JJ, Gharami K, Waterhouse EG, Vanevski F, Jones KR, et al. Dendritically targeted Bdnf mRNA is essential for energy balance and response to leptin. Nature Medicine. 2012;18:564-571. DOI: 10.1038/nm.2687
  58. 58. Gray J, Yeo GSH, Cox JJ, Morton J, Adlam A-LR, Keogh JM, et al. Hyperphagia, severe obesity, impaired cognitive function, and hyperactivity associated with functional loss of one copy of the brain-derived neurotrophic factor (BDNF) gene. Diabetes. 2006;55:3366-3371. DOI: 10.2337/db06-0550
  59. 59. Mou Z, Hyde TM, Lipska BK, Martinowich K, Wei P, Ong C-J, et al. Human obesity associated with an intronic SNP in the brain-derived neurotrophic factor locus. Cell Reports. 2015;13:1073-1080. DOI: 10.1016/j.celrep.2015.09.065
  60. 60. Han JC, Liu QR, Jones M, Levinn RL, Menzie CM, Jefferson-George KS, et al. Brain-derived neurotrophic factor and obesity in the WAGR syndrome. The New England Journal of Medicine. 2008;359:918-927. DOI: 10.1056/NEJMoa0801119
  61. 61. Xu B, Xie X. Neurotrophic factor control of satiety and body weight. Nature Reviews. Neuroscience. 2016;17:282-292. DOI: 10.1038/nrn.2016.24
  62. 62. Rios M. Neurotrophins and the regulation of energy balance and body weight. Handbook of Experimental Pharmacology. 2014;220:283-307. DOI: 10.1007/978-3-642-45106-5_11
  63. 63. Teillon S, Calderon GA, Rios M. Diminished diet-induced hyperglycemia and dyslipidemia and enhanced expression of PPARalpha and FGF21 in mice with hepatic ablation of brain-derived neurotropic factor. The Journal of Endocrinology. 2010;205:37-47. DOI: 10.1677/JOE-09-0405. doi: 10.1677/JOE-09-0405
  64. 64. Yang AC, Chen TJ, Tsi SJ, Hong C, Kuo CH, Yag CH, et al. BDNF Val66Met polymorphism alters sympathovagal balance in healthy subjects. American Journal of Medical Genetics. Part B, Neuropsychiatric Genetics. 2010;153B:1024-1030. DOI: 10.1002/ajmg.b.31069
  65. 65. Nonomura T, Tsuchida A, Ono-Kishino M, Nakagawa T, Taiji M, Noguchi H. Brain-derived neurotrophic factor regulates energy expenditure through the central nervous system in obese diabetic mice. International Journal of Experimental Diabetes Research. 2001;2:201-209. DOI: 10.1155/edr.2001.201
  66. 66. Nakagawa T, Ogawa Y, Ebihara K, Yamanaka M, Tsuchida A, Taiji M, et al. Anti-obesity and anti-diabetic effects of brain-derived neurotrophic factor in rodent models of leptin resistance. International Journal of Obesity and Related Metabolic Disorders. 2003;27:557-565. DOI: 10.1038/sj.ijo.0802265
  67. 67. Gotoh K, Masaki T, Chiba S, Ando H, Fujiwara K, Shimasaki T, et al. Hypothalamic brain-derived neurotrophic factor regulates glucagon secretion mediated by pancreatic efferent nerves. Journal of Neuroendocrinology. 2013;25:302-311. DOI: 10.1111/jne.12003
  68. 68. Fulgenzi G, Hong Z, Tomassoni-Ardori F, Barella LF, Becker J, Barrick C, et al. Novel metabolic role for BDNF in pancreatic β-cell insulin secretion. Nature Communications. 2020;11:1950. DOI: 10.1038/s41467-020-15833-5
  69. 69. Baeza-Raja B, Li P, Le Moan N, Sachs BD, Schachtrup C, Davalos D, et al. p75 neurotrophin receptor regulates glucose homeostasis and insulin sensitivity. Proceedings of the National Academy of Sciences of the United States of America. 2012;109:5838-5843. DOI: 10.1073/pnas.1103638109
  70. 70. Baeza-Raja B, Sachs BD, Li P, Christian F, Vagena E, Davalos D, et al. p75 neurotrophin receptor regulates energy balance in obesity. Cell Reports. 2016;14:255-268. DOI: 10.1016/j.celrep.2015.12.028
  71. 71. Teng HK, Teng KK, Lee R, Wright S, Tevar S, Almeida RD, et al. ProBDNF induces neuronal apoptosis via activation of a receptor complex of p75NTR and sortilin. The Journal of Neuroscience. 2005;25:5455-5463. DOI: 10.1523/JNEUROSCI.5123-04.2005
  72. 72. Anastasia A, Deinhardt K, Chao MV, Will NE, Irmady K, Lee FS, et al. Val66Met polymorphism of BDNF alters prodomain structure to induce neuronal growth cone retraction. Nature Communications. 2013;4:2490. DOI: 10.1038/ncomms3490
  73. 73. Ieraci A, Barbieri SS, Macchi C, Amadio P, Sandrini L, Magni P, et al. BDNF Val66Met polymorphism alters food intake and hypothalamic BDNF expression in mice. Journal of Cellular Physiology. 2020;235:9667-9675. DOI: 10.1002/jcp.29778
  74. 74. Chen Z-Y, Jing D, Bath KG, Ieraci A, Khan T, Siao CJ, et al. Genetic variant BDNF (Val66Met) polymorphism alters anxiety-related behavior. Science. 2006;314:140-143. DOI: 10.1126/science.1129663
  75. 75. Madra M, Zeltser LM. BDNF-Val66Met variant and adolescent stress interact to promote susceptibility to anorexic behavior in mice. Translational Psychiatry. 2016;6:e776. DOI: 10.1038/tp.2016.35
  76. 76. Lin Z, Tann JY, Goh ETH, Kelly C, Lim KB, Gao JF, et al. Structural basis of death domain signaling in the p75 neurotrophin receptor. eLife. 2015;4:e11692. DOI: 10.7554/eLife.11692
  77. 77. Harrington AW, Kim JY, Yoon SO. Activation of Rac GTPase by p75 is necessary for c-Jun N-terminal kinase-mediated apoptosis. The Journal of Neuroscience. 2002;22:156-166. DOI: 10.1523/JNEUROSCI.22-01-00156.2002
  78. 78. Edelmann E, Lessmann V, Brigadski T. Pre- and postsynaptic twists in BDNF secretion and action in synaptic plasticity. Neuropharmacology. 2014;76(Pt C):610-627. DOI: 10.1016/j.neuropharm.2013.05.043
  79. 79. Boschen KE, Klintsova AY. Neurotrophins in the brain: Interaction with alcohol exposure during development. Vitamins and Hormones. 2017;104:197-242. DOI: 10.1016/bs.vh.2016.10.008
  80. 80. Hang PZ, Zhu H, Li PF, Liu J, Ge FQ , Zhao J, et al. The emerging role of BDNF/TrkB signaling in cardiovascular diseases. Life (Basel). 2021;11(1):70. DOI: 10.3390/life11010070
  81. 81. Kermani P, Hempstead B. BDNF actions in the cardiovascular system: Roles in development, adulthood and response to injury. Frontiers in Physiology. 2019;26(10):455. DOI: 10.3389/fphys.2019.00455
  82. 82. Eyileten C, Kaplon-Cieslicka A, Mirowska-Guzel D, Malek L, Postula M. Antidiabetic effect of brain-derived neurotrophic factor and its association with inflammation in type 2 diabetes mellitus. Journal Diabetes Research. 2017;2017:2823671
  83. 83. Rozanska O, Uruska A, Zozulinska-Ziolkiewicz D. Brain derived neurotrophic factor and diabetes. International Journal of Molecular Sciences. 2020;21(3):841. DOI: 10.3390/ijms21030841
  84. 84. Prakash YS, Martin RJ. Brain-derived neurotrophic factor in the airways. Pharmacology & Therapeutics. 2014;143(1):74-86. DOI: 10.1016/j.pharmthera.2014.02.006
  85. 85. Giacobbo BL, Doorduin J, Klein HC, Dierckx RAJO, Bromberg E. de Vries EFJ brain-derived neurotrophic factor in brain disorders: Focus on neuroinflammation. Molecular Neurobiology. 2019;56(5):3295-3312. DOI: 10.1007/s12035-018-1283-6
  86. 86. Eisch AJ, Bolaños CA, de Wit J, Simonak RD, Pudiak CM, Barrot M, et al. Brain-derived neurotrophic factor in the ventral midbrain-nucleus accumbens pathway: A role in depression. Biological Psychiatry. 2003;54(10):994-1005. DOI: 10.1016/j.biopsych.2003.08.003
  87. 87. Berton O, McClung CA, Dileone RJ, Krishnan V, Renthal W, Russo SJ, et al. Essential role of BDNF in the mesolimbic dopamine pathway in social defeat stress. Science. 2006;311(5762):864-868. DOI: 10.1126/science.1120972
  88. 88. Zhang JC, Wu J, Fujita Y, Yao W, Ren Q , Yang C, et al. Antidepressant effects of TrkB ligands on depression-like behavior and dendritic changes in mice after inflammation. The International Journal of Neuropsychopharmacology. 2015;18(4):pyu077. DOI: 10.1093/ijnp/pyu077
  89. 89. Adachi M, Barrot M, Autry AE, Theobald D, Monteggia LM. Selective loss of brain-derived neurotrophic factor in the dentate gyrus attenuates antidepressant efficacy. Biological Psychiatry. 2008;63(7):642-649. DOI: 10.1016/j.biopsych.2007.09.019
  90. 90. Hsieh MT, Lin CC, Lee CT, Huang TL. Abnormal brain-derived neurotrophic factor exon IX promoter methylation, protein, and mRNA levels in patients with major depressive disorder. Journal of Clinical Medicine. 2019;8:568. DOI: 10.3390/jcm8050568
  91. 91. Dwivedi Y, Rizavi HS, Conley RR, Roberts RC, Tamminga CA, Pandey GN. Altered gene expression of brain-derived neurotrophic factor and receptor tyrosine kinase B in postmortem brain of suicide subjects. Archives of General Psychiatry. 2003;60(8):804-815. DOI: 10.1001/archpsyc.60.8.804
  92. 92. Hashimoto K, Shimizu E, Iyo M. Critical role of brain-derived neurotrophic factor in mood disorders. Brain Research. Brain Research Reviews. 2004;45(2):104-114. DOI: 10.1016/j.brainresrev.2004.02.003
  93. 93. Hung YY, Lin CJ, Huang TL. Higher serum tropomyosin-related kinase B protein level in major depression. Progress in Neuro-Psychopharmacology and Biological Psychiatry. 2010;34:610-612. DOI: 10.1016/j.pnpbp.2010.02.021
  94. 94. Tsai SJ. Down-regulation of the Trk-B signal pathway: The possible pathogenesis of major depression. Medical Hypotheses. 2004;62:215-218. DOI: 10.1016/S0306-9877(03)00299-8
  95. 95. Fuchikami M, Morinobu S, Segawa M, Okamoto Y, Yamawaki S, Ozaki N. DNA methylation profiles of the brain-derived neurotrophic factor (BDNF) gene as a potent diagnostic biomarker in major depression. PLoS One. 2011:6(8):e23881 DOI: 10.1371/journal.pone.0023881
  96. 96. D'Addario C, Dell'Osso B, Galimberti D, Palazzo MC, Benatti B, Di Francesco A. Epigenetic modulation of BDNF gene in patients with major depressive disorder. Biological Psychiatry. 2013;73:e6-e7. DOI: 10.1016/j.biopsych.2012.07.009
  97. 97. Tadic A, Muller-Engling L, Schlicht KF, Kotsiari A, Dreimuller N, Kleimann A. Methylation of the promoter of brain-derived neurotrophic factor exon IV and antidepressant response in major depression. Molecular Psychiatry. 2014;19:281-283. DOI: 10.1038/mp.2013.58
  98. 98. Verhagen M, van der Meij A, van Deurzen PA, Janzing JG, Arias-Vasquez A, Buitelaar JK. Meta-analysis of the BDNF Val66Met polymorphism in major depressive disorder: Effects of gender and ethnicity. Molecular Psychiatry. 2010;15:260-271. DOI: 10.1038/mp.2008.109
  99. 99. Gyekis JP, Yu W, Dong S, Wang H, Qian J, Kota P. No association of genetic variants in BDNF with major depression: A meta- and gene-based analysis. American Journal of Medical Genetics. Part B, Neuropsychiatric Genetics: The Official Publication of the International Society of Psychiatric Genetics. 2013;16(2B):61-70. DOI: 10.1002/ajmg.b.32122
  100. 100. Schroter K, Brum M, Brunkhorst-Kanaan N, Tole F, Ziegler C, Domschke K. Longitudinal multi-level biomarker analysis of BDNF in major depression and bipolar disorder. European Archives of Psychiatry and Clinical Neuroscience. 2020;270:169-181. DOI: 10.1007/s00406-019-01007-y
  101. 101. Hosang GM, Shiles C, Tansey KE, McGuffin P, Uher R. Interaction between stress and the BDNF Val66Met polymorphism in depression: A systematic review and meta-analysis. BMC Medicine. 2014;12:7. DOI: 10.1186/1741-7015-12-7
  102. 102. Januar V, Ancelin ML, Ritchie K, Saffery R, Ryan J. BDNF promoter methylation and genetic variation in late-life depression. Translational Psychiatry. 2015;5:e619. DOI: 10.1038/tp.2015.114
  103. 103. Badamasi IM, Lye MS, Ibrahim N, Stanslas J. Genetic endophenotypes for insomnia of major depressive disorder and treatment-induced insomnia. Journal of Neural Transmission. 2019;126:711-722. DOI: 10.1007/s00702-019-02014-y
  104. 104. Fujimaki K, Takahashi T, Morinobu S. Association of typical versus atypical antipsychotics with symptoms and quality of life in schizophrenia. PLoS One. 2012;7(5):e37087. DOI: 10.1371/journal.pone.0037087
  105. 105. Yang Y, Liu Y, Wang G, Hei G, Wang X, Li RR, et al. Brain-derived neurotrophic factor is associated with cognitive impairments in first-episode and chronic schizophrenia. Psychiatry Research. 2019;273:5281536. DOI: 10.1016/j.psychres.2019.01.051
  106. 106. Favalli G, Li J, Belmonte-De-Abreu P, Wong AH, Daskalakis ZJ. The role of BDNF in the pathophysiology and treatment of schizophrenia. Journal of Psychiatric Research. 2012;46:1-11. DOI: 10.1016/j.jpsychires.2011.09.022
  107. 107. Grillo RW, Ottoni GL, Leke R, Souza DO, Portela LV, Lara DR. Reduced serum BDNF levels in schizophrenic patients on clozapine or typical antipsychotics. Journal of Psychiatric Research. 2007;41:31-35. DOI: 10.1016/j.jpsychires.2006.01.005
  108. 108. Cheah SY, McLeay R, Wockner LF, Lawford BR, Young RM, Morris CP, et al. Expression and methylation of BDNF in the human brain in schizophrenia. The World Journal of Biological Psychiatry: The Official Journal of the World Federation of Societies of Biological Psychiatry. 2007;18(5):392-400. DOI: 10.1080/15622975.2016.1245443
  109. 109. Punzi G, Bharadwaj R, Ursini G. Neuroepigenetics of schizophrenia. Progress in Molecular Biology and Translational Science. 2018;158:195-226. DOI: 10.1016/bs.pmbts.2018.04.010
  110. 110. Ibi D, Gonzalez-Maeso J. Epigenetic signaling in schizophrenia. Cellular Signalling. 2015;27(10):2131-2136. DOI: 10.1016/j.cellsig.2015.06.003
  111. 111. Kordi-Tamandani DM, Sahranavard R, Torkamanzehi A. DNA methylation and expression profiles of the brain-derived neurotrophic factor (BDNF) and dopamine transporter (DAT1) genes in patients with schizophrenia. Molecular Biology Reports. 2012;39:10889-10893. DOI: 10.1007/s11033-012-1986-0
  112. 112. Ikegame T, Bundo M, Murata Y, Kasai K, Kato T, Iwamoto K. DNA methylation of the BDNF gene and its relevance to psychiatric disorders. Journal of Human Genetics. 2013;58:434-438. DOI: 10.1038/jhg.2013.65
  113. 113. Jaffe AE, Gao Y, Deep-Soboslay A, Tao R, Hyde TM, Weinberger DR, et al. Mapping DNA methylation across development, genotype and schizophrenia in the human frontal cortex. Nature Neuroscience. 2016;19(1):40-47. DOI: 10.1038/nn.4181
  114. 114. Ahmed AO, Kramer S, Hofman N, Flynn J, Hansen M, Martin V, et al. A meta-analysis of brain-derived neurotrophic factor effects on brain volume in schizophrenia: Genotype and serum levels. Neuropsychobiology. 2021;80:411-424. DOI: 10.1159/000514126
  115. 115. Zhao X, Huang Y, Chen K, Li D, Han C, Kan Q. The brain-derived neurotrophic factor Val66Met polymorphism is not associated with schizophrenia: An updated meta-analysis of 11,480 schizophrenia cases and 13,490 controls. Psychiatry Research. 2015;225:217-220. DOI: 10.1016/j.psychres.2014.11.015
  116. 116. Notaras M, Hill R, van den Buuse M. A role for the BDNF gene Val66Met polymorphism in schizophrenia? A comprehensive review. Neuroscience & Biobehavioral Reviews. 2015;51:15-30. DOI: 10.1016/j.neubiorev.2014.12.016
  117. 117. Merinkangas KR, Low NC. The epidemiology of mood disorders. Current Psychiatry Reports. 2004;6:411-421. DOI: 10.1007/s11920-004-0004-1
  118. 118. Rihmer Z, Kiss K. Bipolar disorders and suicidal behavior. Bipolar Disorders. 2002;4(Suppl 1):21-25. DOI: 10.1034/j.1399-5618.4. s1.3.x
  119. 119. American Psychiatric Association. Diagnostic and Statistical Manual of Mental Disorders, fifth ed., 2013; DSM-5. Arlington, VA: American Psychiatric Association;
  120. 120. Laske C, Stransky E, Eschweiler GW, Klein R, Wittorf A, Leyhe T, et al. Increased BDNF serum concentration in fibromyalgia with or without depression or antidepressants. Journal of Psychiatric Research. 2007;41(7):600-605. DOI: 10.1016/j.jpsychires.2006.02.007
  121. 121. Fernandes BS, Molendijk ML, |Köhler CA, Soares JC, Leite CM, Machado-Vieira R, et al. Peripheral brain-derived neurotrophic factor (BDNF) as a biomarker in bipolar disorder: A meta-analysis of 52 studies. BMC Medicine. 2015;13:289. DOI: 10.1186/s12916-015-0529-7
  122. 122. Lin CC, Lee CT, Lo YT, Huang TL. Brain-derived neurotrophic factor protein and mRNA levels in patients with bipolar mania - a preliminary study. Biomedical Journal. 2016;39:272-276. DOI: 10.1016/j.bj.2016.08.001
  123. 123. Chiou YJ, Huang TL. Brain-derived neurotrophic factor (BDNF) and bipolar disorder. Psychiatry Research. 2019;274:395-399. DOI: 10.1016/j.psychres.2019.02.051
  124. 124. Aas M, Dieset I, Morch R, Steen NE, Hope S, Reponen EJ. Reduced brain-derived neurotrophic factor is associated with childhood trauma experiences and number of depressive episodes in severe mental disorders. Schizophrenia Research. 2019;205:45-50. DOI: 10.1016/j.schres.2018.08.007
  125. 125. Mora E, Portella MJ, Pinol-Ripoll G, Lopez R, Cuadras D, Forcada I. High BDNF serum levels are associated to good cognitive functioning in bipolar disorder. European Psychiatry. 2019;60:97-107. DOI: 10.1016/j.eurpsy.2019.02.006
  126. 126. Fernandes BS, Gama CS, Cereser KM, Yatham LN, Fries GR, Colpo G. Brain-derived neurotrophic factor as a state-marker of mood episodes in bipolar disorders: A systematic review and meta-regression analysis. Journal of Psychiatric Research. 2011;45:995-1004. DOI: 10.1016/j.jpsychires.2011.03.002
  127. 127. Kim HW, Rapoport SI, Rao JS. Altered expression of apoptotic factors and synaptic markers in postmortem brain from bipolar disorder patients. Neurobiology of Disease. 2010;37:596-603. DOI: 10.1016/j.nbd.2009.11.010
  128. 128. Lin CC, Hung YY, Huang TL. Associations between DNA methylation of promoter exon IX, serum protein and mRNA levels of brain-derived neurotrophic factor in patients with bipolar mania. Neuropsychiatry. 2018;8:224-231
  129. 129. Grande I, Kapczinski F, Stertz L, Colpo GD, Kunz M, Cereser KM. Peripheral brain-derived neurotrophic factor changes along treatment with extended-release quetiapine during acute mood episodes: An open-label trial in drug-free patients with bipolar disorder. Journal of Psychiatric Research. 2012;46:1511-1514. DOI: 10.1016/j.jpsychires.2012.08.017
  130. 130. Neves-Pereira M, Mundo E, Muglia P, King N, Macciardi F, Kennedy JL. The brain-derived neurotrophic factor gene confers susceptibility to bipolar disorder: Evidence from a family-based association study. American Journal of Human Genetics. 2002;71:651-655. DOI: 10.1016/j.jpsychires.2012.08.017
  131. 131. Li M, Chang H, Xiao X. BDNF Val66Met polymorphism and bipolar disorder in European populations: A risk association in case-control, family-based and GWAS studies. Neuroscience and Biobehavioral Reviews. 2016;68:218-233. DOI: 10.1016/j.neubiorev.2016.05.031
  132. 132. Wang Z, Li Z, Gao K, Fang Y. Association between brain-derived neurotrophic factor genetic polymorphism Val66Met and susceptibility to bipolar disorder: A meta-analysis. BMC Psychiatry. 2014;14:366. DOI: 10.1186/s12888-014-0366-9
  133. 133. Harrisberger F, Smieskova R, Schmidt A, Lenz C, Walter A, Wittfeld K, et al. BDNF Val66Met polymorphism and hippocampal volume in neuropsychiatric disorders: A systematic review and meta-analysis. Neuroscience and Biobehavioral Reviews. 2015;55:107-118. DOI: 10.1016/j.neubiorev.2015.04.017
  134. 134. Mill J, Tang T, Kaminsky Z, Khare T, Yazdanpanah S, Bouchard L. Epigenomic profiling reveals DNA-methylation changes associated with major psychosis. American Journal of Human Genetics. 2008;82:696-711. DOI: 10.1016/j.ajhg.2008.01.008
  135. 135. Strauss JS, Khare T, De Luca V, Jeremian R, Kennedy JL, Vincent JB. Quantitative leukocyte BDNF promoter methylation analysis in bipolar disorder. International Journal of Bipolar Disorders. 2013;1:28. DOI: 10.1186/2194-7511-1-28
  136. 136. D'Addario C, Dell'Osso B, Palazzo MC, Benatti B, Lietti L, Cattaneo E. Selective DNA methylation of BDNF promoter in bipolar disorder: Differences among patients with BDI and BDII. Neuropsychopharmacology, The Official Publication of the American College of Neuropsychopharmacology. 2012;37:1647-1655. DOI: 10.1038/npp.2012.10
  137. 137. Stenz L, Zewdie S, Laforge-Escarra T, Prados J, La Harpe R, Dayer A, et al. BDNF promoter I methylation correlates between postmortem human peripheral and brain tissues. Neuroscience Research. 2015;91:1-7. DOI: 10.1016/j.neures.2014.10.003
  138. 138. Carlberg L, Scheibelreiter J, Hassler MR, Schloegelhofer M, Schmoeger M, Ludwig B, et al. Brain-derived neurotrophic factor (BDNF)-epigenetic regulation in unipolar and bipolar affective disorder. Journal of Affective Disorders. 2014;168:399-406. DOI: 10.1016/j.jad.2014.07.022
  139. 139. Rao JS, Keleshian VL, Klein S, Rapoport SI. Epigenetic modifications in frontal cortex from Alzheimer’s disease and bipolar disorder patients. Translational Psychiatry. 2012a;2:e132. DOI: 10.1038/tp.2012.55
  140. 140. Dell’osso L, Carmassi C, Del Debbio A, Dell’osso MC, Bianchi C, da Pozzo E, et al. Brain-derived neurotrophic factor plasma levels in patients suffering from post-traumatic stress disorder. Progress in Neuro-Psychopharmacology & Biological Psychiatry. 2009;33:899-902. DOI: 10.1016/j.pnpbp.2009.04.018
  141. 141. Domitrovic Spudic S, Nikolac Perkovic M, Uzun S, Nedic Erjavec G, Kozumplik O, Svob Strac D, et al. Reduced plasma BDNF concentration and cognitive decline in veterans with PTSD. Psychiatry Research. 2022;316:114772. DOI: 10.1016/j.psychres.2022.114772
  142. 142. Dretsch MN, Williams K, Emmerich T, Crynen G, Ait-Ghezala G, Chaytow H, et al. Brain derived neurotropic factor polymorphisms, traumatic stress, mild traumatic brain injury, and combat exposure contribute to postdeployment traumatic stress. Brain and Behavior: A Cognitive Neuroscience Perspective. 2015;6(1):e00392. DOI: 10.1002/brb3.392
  143. 143. Kim TY, Kim SJ, Chung HG, Choi JH, Kim SH, Kang JI. Epigenetic alterations of the BDNF gene in combat-related post-traumatic stress disorder. Acta Psychiatrica Scandinavica. 2017;135(2):170-179. DOI: 10.1111/acps.12675
  144. 144. Voisey J, Lawford B, Bruenig D, Harvey W, Morris CP, Young RM, et al. PTSD Initiative. Differential BDNF methylation in combat exposed veterans and the association with exercise. Gene. 2019;698:107-112. DOI: 10.1016/j.gene.2019.02.067
  145. 145. Guo JC, Yang YJ, Zheng XA, Jiang XL, Guo M, Wang XD, et al. CpG methylation of brain-derived the neurotrophic factor gene promoter as a potent diagnostic and prognostic biomarker for post-traumatic stress disorder. International Journal of Clinical and Experimental Pathology. 2018;11(10):5101-5109
  146. 146. Valente NL, Vallada H, Cordeiro Q , et al. Candidate-gene approach in posttraumatic stress disorder after urban violence: Association analysis of the genes encoding serotonin transporter, dopamine transporter, and BDNF. Journal of Molecular Neuroscience. 2011;44:59-67. DOI: 10.1007/s12031-011-9513-7
  147. 147. Lee H, Kang R, Lim S, Paik J, Choi M, Lee M. No association between the brain-derived neurotrophic factor gene Val66Met polymorphism and post-traumatic stress disorder. Stress and Health. 2006;22:115-119
  148. 148. Zhang H, Ozbay F, Lappalainen J, Kranzler HR, van Dyck CH, Charney DS, et al. Brain derived neurotrophic factor (BDNF) gene variants and Alzheimer’s disease, affective disorders, posttraumatic stress disorder, schizophrenia, and substance dependence. American Journal of Medical Genetics. 2006;141B:387-393. DOI: 10.1002/ajmg.b.30332
  149. 149. Hauck S, Kapczinski F, Roesler R, de Moura Silveira E Jr, Magalhaes PV, Kruel LR, et al. Serum brain-derived neurotrophic factor in patients with trauma psychopathology. Progress in Neuro-Psychopharmacology & Biological Psychiatry. 2010;34:459-462. DOI: 10.1016/j.pnpbp.2010.01.010
  150. 150. Mojtabavi H, Saghazadeh A, van den Heuvel L, Bucker J, Rezaei N. Peripheral blood levels of brain-derived neurotrophic factor in patients with post-traumatic stress disorder (PTSD): A systematic review and meta-analysis. PLoS One. 2020;15(11):e0241928. DOI: 10.1371/journal.pone.0241928
  151. 151. Wu GWY, Wolkowitz OM, Reus VI, Kang JI, Elnar M, Sarwal R, et al. Serum brain-derived neurotrophic factor remains elevated after long term follow-up of combat veterans with chronic post-traumatic stress disorder. Psychoneuroendocrinology. 2021;134:105360. DOI: 10.1016/j.psyneuen.2021.105360
  152. 152. Bartel DP. MicroRNAs: Genomics, biogenesis, mechanism, and function. Cell. 2004;116:281-297. DOI: 10.1016/s0092-8674(04)00045-5
  153. 153. Varendi K, Kumar A, Härma MA, Andressoo JO. miR-1, miR-10b, miR-155, and miR-191 are novel regulators of BDNF. Cellular and Molecular Life Sciences. 2014;71(22):4443-4456. DOI: 10.1007/s00018-014-1628-x. doi: 10.1007/s00018-014-1628-x
  154. 154. Lau AG, Irier HA, Gu J, Tian D, Ku L, Liu G, et al. Distinct 3'UTRs differentially regulate activity-dependent translation of brain-derived neurotrophic factor (BDNF). Proceedings of the National Academy of Sciences of the United States of America. 2010;107:15945-15950. DOI: 10.1073/pnas.1002929107
  155. 155. Li YJ, Xu M, Gao ZH, Wang YQ , Yue Z, Zhang YX, et al. Alterations of serum levels of BDNF-related miRNAs in patients with depression. PLoS One. 2013;8:e63648. DOI: 10.1371/journal.pone.0063648
  156. 156. Abdolahi S, Zare-Chahoki A, Noorbakhsh F, Gorji A. Review of molecular interplay between neurotrophins and miRNAs in neuropsychological disorders. Molecular Neurobiology. 2022;59(10):6260-6280. DOI: 10.1007/s12035-022-02966-5
  157. 157. Maurel OM, Torrisi SA, Barbagallo C, Purrello M, Salomone S, Drago F, et al. Dysregulation of miR-15a-5p, miR-497a-5p and miR-511-5p is associated with modulation of BDNF and FKBP5 in brain areas of PTSD-related susceptible and resilient mice. International Journal of Molecular Sciences. 2021;22(10):5157. DOI: 10.3390/ijms22105157
  158. 158. Wan Y, Liu Y, Wang X, Wu J, Liu K, Zhou J, et al. Identification of differential MicroRNAs in cerebrospinal fluid and serum of patients with major depressive disorder. PLoS One. 2015;10(3):e0121975. DOI: 10.1371/journal.pone.0121975
  159. 159. Murphy CP, Singewald N. Role of MicroRNAs in anxiety and anxiety-related disorders. Current Topics in Behavioral Neurosciences. 2019;42:185-219. DOI: 10.1007/7854_2019_109
  160. 160. Gunasekaran S, Jacob RS, Omkumar RV. Differential expression of miR-148b, miR-129-2 and miR-296 in animal models of schizophrenia-relevance to NMDA receptor hypofunction. Neuropharmacology. 2022;210:109024. DOI: 10.1016/j.neuropharm.2022.109024
  161. 161. Ji LL, Ye Y, Nie PY, Peng JB, Fu CH, Wang ZY, et al. Dysregulation of miR-142 results in anxiety-like behaviors following single prolonged stress. Behavioural Brain Research. 2019;365:157-163. DOI: 10.1016/j.bbr.2019.03.018
  162. 162. Wang Z, Zhang C, Huang J, Yuan C, Hong W, Chen J, et al. MiRNA-206 and BDNF genes interacted in bipolar I disorder. Journal of Affective Disorders. 2014;162:116-119. DOI: 10.1016/j.jad.2014.03.047
  163. 163. Lee SY, Wang TY, Lu RB, Wang LJ, Chang CH, Chiang YC, et al. Peripheral BDNF correlated with miRNA in BD-II patients. Journal of Psychiatric Research. 2021;136:184-189. DOI: 10.1016/j.jpsychires.2021.02.018

Written By

Sudhiranjan Gupta and Rakeshwar S. Guleria

Submitted: 30 June 2023 Reviewed: 17 July 2023 Published: 21 September 2023