Home
JournalsCollections
For Authors For Reviewers For Editorial Board Members
Article Processing Charges Open Access
Ethics Advertising Policy
Editorial Policy Resource Center
Company Information Contact Us Membership Collaborators Partners
Publications > Journals > Gene Expression> Article Full Text
OPEN ACCESS

c-Fos Expression Differentially Acts in the Healthy Brain Compared with Alzheimer’s Disease

  • Parvin Babaei1,2,3,* ,
  • Niloofar Faraji4 and
  • Kimia Eyvani1
Gene Expression   2025

doi: 10.14218/GE.2024.00080

Received:

Revised:

Accepted:

Published online:

 Author information

Citation: Babaei P, Faraji N, Eyvani K. c-Fos Expression Differentially Acts in the Healthy Brain Compared with Alzheimer’s Disease. Gene Expr. Published online: Apr 28, 2025. doi: 10.14218/GE.2024.00080.

Abstract

The proto-oncogene c-Fos is known as a reliable marker of cell activation, which is immediately induced after a new stimulus in specific brain regions, depending on the nature of the stimulus applied. However, the expression of c-Fos is increased in Alzheimer’s disease (AD) and contributes to amyloid β-peptide-induced neurotoxicity. This review attempted to focus on the role of c-Fos in learning and memory in both healthy brain and AD, emphasizing on possible mechanisms. Comparing the available findings, regarding learning and memory, c-Fos expression leads to memory formation through ERK (extracellular signal-regulated kinase)/CREB (cAMP response element-binding protein) and long-term potentiation, while it is down regulated after the repetition and habituation of stimuli. However, its overexpression in neurons and glia of AD, contributes to cognitive deficits and neuronal loss, which represents a defect in its ability to habituate to repeated stimuli. Also, expression pattern in glial is associated with constitutive CREB activation following increasing amyloid beta (Aβ), activation transcription factor (ATF3), and cytochrome c in apoptosis pathways. Thus, two contradictory roles of c-Fos in the healthy brain and AD, reveal more complexity in c-Fos up and down stream signaling pathways, bioavailability, and sensitivity. Future studies focusing on c-Fos modulation, might offer promising strategies to mitigate cognitive decline in AD.

Keywords

c-Fos, Learning, Long term memory, Alzheimer’s disease, Neuron, Astrocytes, Neural plasticity, Apoptosis, Amyloid beta

Introduction

Immediate-early genes (IEGs) constitute the early genomic response to various stimuli, either directly by encoding transcription factors or through their protein products, which modify cell functions.1 More than seventy IEGs have been identified, including the most well-known and well-characterized IEG: c-Fos.2 The c-Fos gene was first introduced as a proto-oncogene responsible for the induction of bone tumors encoded by the Finkel–Biskis–Jinkins murine osteogenic sarcoma virus.3 Basal expression of c-Fos is low, but it rapidly peaks between 30 and 45 m, producing a 380-amino acid protein called “Fos,” which influences target genes.4 Other members of the Fos family consist of four proteins: FOS, FOSB, FOSL1, and FOSL2.1,5

During development, c-Fos is involved in proliferation and differentiation, while in the adult brain, it contributes to neural activity,6 and responses to stress across various regions, including the hippocampus, cerebral cortex, and medial prefrontal cortex,1,5,7 as well as long-term memory and synaptic plasticity,6,8 and activity maturation in the hippocampal–entorhinal under physiological conditions.9,10 Its rapid, sensitive, and synchronized response to various stimuli has made it a marker for neuronal activation.11 Notably, c-Fos expression shows distinct patterns depending on the novelty and intensity of the stimulus, in such a way that acute neuronal activity triggers a strong response.12–14

On the other hand, recently, dysregulation of c-Fos expression has been implicated in numerous neurological disorders, including Alzheimer’s disease (AD).1,15,16 AD is a chronic neurodegenerative disorder characterized by cognitive decline, inflammation, and memory loss, and it still remains as an incurable disorder.17–19 Various combined actions of signaling pathways, including c-Fos, have been reported to be activated during AD.17 Therefore, it is important to investigate these signaling pathways to better understand the etiology and treatment of this disease.

Studies have revealed increased c-Fos expression in the amygdala and hippocampus of AD patients, which correlates with cognitive decline and cellular apoptosis.20,21 The elevation of Fos protein appears to participate in a destructive cycle by promoting amyloid beta (Aβ) accumulation,21 expression of apoptotic genes,22 and neuronal loss.22,23 These findings, together with the detection of c-Fos in inflammatory subtypes of glia, suggest that c-Fos might be involved in neuronal death and the inflammatory response, exacerbating the progression of AD.

The objective of this narrative review was to answer the question of whether c-Fos alterations and responsiveness are the same in the normal and AD brain. Thus, we summarized studies on AD postmortem brains, rodent AD models, and cell cultures. Next, we explore the main data linking mechanisms underlying c-Fos expression in both neurons and astrocytes.

Expression of the c-Fos gene in brain regions

Expression of c-Fos in the brain is induced by physiological synapses and is closely related to the neural activity of the brain in various models of stress and pain,24–26 passive avoidance learning,27 and emotional stress.13 It seems that the pattern of expression occurs within specific brain regions, confirming a mapping pattern of the brain areas involved in response to those stimuli,28 during either physiological or pathological states.29 However, it can be difficult to determine the specificity of expression when a stimulus consists of various components such as stress, emotion, motivation, anxiety, learning, and pain.

Among brain regions, c-Fos expression has been extensively reported in hippocampal regions, including CA1, fimbria, dentate gyrus, hilus, and cerebral cortex,20,30 as well as in the parietal, medial prefrontal, and ventrolateral orbital cortex after noxious stimuli.27 Previously, we showed significantly more Fos-positive neurons in 92 brain regions after a slight electric foot shock, in rats predisposed to emotional stress, compared with the resistant phenotype.13 This extensive pattern of c-Fos expression reflects differences in behavioral typologies and the sensitivity of those brain regions in response to components of stress, anxiety, motivation, fear, emotion, pain, and aversive memory.

The c-Fos gene in different brain cell types

It is obvious that c-Fos is induced in neurons of specific brain regions like the hippocampus, amygdala, and cortex, to connect neural information with brain regions as part of a homeostatic response.24,25 In addition to neurons, it is expressed in the glial cells of adult rat brains.1,31–37 In astrocytes, it is expressed under the influence of proliferation, differentiation, growth, inflammation, repair, damage,1,31 stress,32 cytokines,33 lipopolysaccharide (LPS),34 and infection with adenovirus.35

Glial cells are important resident cells in the brain that are involved in various functions under both physiological and pathological conditions, including the regulation of immune responses,1,38 response to stress,39,40 development of synapses,41 neurotransmitter uptake,42 maintaining the ionic balance in synaptic and extrasynaptic spaces,43 bidirectional communication between neurons and astrocytes,43 and the generation of action potentials via Ca2+ waves.44

c-Fos has also been reported to be induced in oligodendrocytes and microglia in response to stress across different subregions of the medial prefrontal cortex.11 For instance, it has been reported that glutamate activates c-Fos in glial cells via metabotropic glutamate receptor subtype 5,36 in addition to N-methyl-d-aspartate (NMDA),45 α-amino-3-hydroxy-5-methyl-4-isoxazole-propionic acid (AMPA), and kainate receptors.46–48 All these findings indicate that glial c-Fos expression may mediate inflammatory responses and be involved in the mechanisms of neuronal loss in AD.32

Taken together, astrocytes play crucial roles in maintaining brain homeostasis, ranging from proliferation to communication, under both physiological and pathological conditions such as trauma, inflammation, and metabolic disturbances partly via c-Fos signaling. This is in contrast to neurons, whose expression is associated with depolarization and neural plasticity.1

Roles of c-Fos in learning and memory

Immediate early gene expression in neurons, plays an important role in the neuroplastic mechanisms underlying learning and memory,7 particularly for consolidation and memory formation.31,49,50 Among various IEGs, c-Fos has been known to be involved in consolidation, recall,6 and encoding of long term memory (LTM).12,51 Several Neuromolecular Labs have reported the immediate and transient expression of c-Fos in various brain regions following training in behavioral tasks, including associative memory,52 traumatic memory,53 passive avoidance learning,13 Morris water maze,51 socially transmitted food preference,54 and cued fear memory.52 These findings are in line with those experiments that disrupted c-Fos function using antisense oligonucleotides and found long-term spatial memory impairment in the water maze,49 as well as impairments in hippocampus-dependent spatial and associative learning tasks.55 Therefore, c-Fos expression couples extracellular signals to long-term adaptive gene expression changes in novel learning contexts,12 while it downregulates after the prolongation of learned behavior,56 repetition, and habituation.57

Collectively, under physiological conditions, it appears that acute neuronal activity, but not chronic and repetitive stimulation, induces c-Fos expression. This indicates a biphasic regulation of c-Fos, governed by hypersensitivity of the c-Fos promoter following exposure to a stimulus, which is followed by fast adaptation in the hippocampus.6 However, this pattern of expression in acute and chronic stimulation does not mean that the c-Fos neural marker always follows the “all-or-none law,” because c-Fos expression is sensitive to both the frequency and intensity of the stimulus, as was evident in our study in the adult rat brain. We showed that 2.5 mA compared with 0.25 mA electrical foot shock caused more c-Fos expression in brain regions related to noxious stimuli in rats.13,58

Besides its role as a neural activity marker, the pattern of c-Fos is very complex when considering individual differences in control rats.13 Significantly more c-Fos expression was reported in rats predisposed to emotional stress compared with resistant ones.13 Taken together, novelty, as a main component of learning and memory, has a crucial influence on c-Fos expression. Both blocking and activating c-Fos either inhibits or induces memory-associated behaviors, respectively.

Mechanism of c-Fos induction and functions during learning & memory

During learning and memory, various molecular signaling pathways, including the excitatory neurotransmitter glutamate,1 bind to NMDA-R and then activate the expression of c-Fos in neurons through various kinases, that are critical to memory and cognition. These include the calcium-dependent phosphorylation of cAMP response element-binding protein (CREB), extracellular signal-regulated kinase (ERK), Janus kinase 1-2, tyrosine kinase 2, mitogen-activated protein kinase (MAPK), calmodulin kinases (hereinafter referred to as CaMKs), protein kinase A,10,59 and protein kinase C (PKC).60,61 PKC is involved in the early induction phase through phosphorylation of glutamate receptor 1 subunits of AMPA-R, whereas protein kinase A is crucial for the late, protein synthesis-dependent phase through phosphorylated MAPK.62,63 Some of the important mechanisms underlying c-Fos expression during learning and memory are summarized in Table 1.1,6,7,12,20–22,25,31–34,36,42,49,51,55,57,64–86

Table 1

c-Fos-related mechanisms in the normal brain and Alzheimer’s disease

AspectKey findingsSignaling pathways and mechanismsType of specimenRef
c-Fos in memory formationc-Fos is involved in the consolidation, recall, and encoding of long-term memory.Neural activity and Ca through glutamate receptors, stimulate downstream signaling via ERK, CREB, and CaMKIV, and lead in LTM.Rat, mouse6,7,12,49,51,55
c-Fos in synaptic plasticityc-Fos expression increases with novel experiences and is downregulated with habituation.ERK/CREB pathway; AP-1 transcription factor complex formation with c-Jun, and then expresses target genes Arc, BDNF.Rat, mouse6,12,25,57
c-Fos in Alzheimer’s disease (AD)Elevated c-Fos expression in postmortem AD brain tissue and AD models.Aβ42-induced c-Fos activation; FOS/ATF signaling and O-GlcNAcylation of c-Fos reduces CREB/BDNF.Human, mouse, rat2022,51,6668,71,74
c-Fos in neuroinflammationc-Fos is induced in glial cells by LPS, cytokines, and glutamate.MAPK, p38, and CREB/ATF-1 pathways; regulation of inflammatory cytokines.Rat, mouse1,3234,36,64
c-Fos in apoptosisc-Fos promotes apoptosis via AP-1 complex and pro-apoptotic gene activation.ERK/FOS activates BAX, caspase-3, and ATF. ATF3-mediated inhibition of PINK1; BIM translocation to mitochondria leads in apoptosis.Human, rat, mouse22,65,69,7780
c-Fos in oxidative stressOxidative stress upregulates c-Fos via MAPK pathways, contributing to neuronal dysfunction.ROS activation of ERK and JNK; increased transcription of pro-apoptotic genes; dysregulation of Nrf2, MAPK, PI3K/Akt, and Wnt/β-catenin signaling; mitochondrial dysfunction-induces apoptosis.Rat, mouse, human75,76,8186
c-Fos in normal glial cellsc-Fos is expressed in astrocytes, oligodendrocytes, and microglia in response to stress and inflammation.Glutamate-induced activation via mGlu5 receptors; MAPK and PKC pathways.Rat, mouse1,3134,36,42
c-Fos in excitotoxicityc-Fos acts as an excitotoxic marker in AD, contributing to neuronal loss.NMDA receptor activation; calcium influx and ROS production.Rat, mouse70,73
c-Fos expression in glial cells of AD subjectsGlutamate and cytokine stimulation of astrocytes rapidly increases c-Fos expression via calcium and complement factor H and causes neuronal loss.MAPK and PKC pathways in intervertebral disc cells; p38 MAPK/CREB/ATF-1 signaling in glial cells; activation via LPS and cyclic AMP/calcium response elements.Rat, mouse1,3234,36,64

Among the complex protein kinase cascades, phosphorylated ERK activity is primarily important for c-Fos/CREB cycling and LTM formation.87 ERK is phosphorylated by MEK, which is previously activated by Raf and Ca2+ through glutamate NMDA-R.88,89 Therefore, based on training conditions, the frequency and duration of kinase/phosphatase activation will determine ERK activity and the establishment of memory engrams.88–92 Following c-Fos expression, the FOS protein is synthesized and returns to the nucleus, where it acts together with c-Jun by binding and forming the activator protein-1 (AP-1) heterodimer,57,64,93 playing a significant role in neural plasticity.65 Although CREB and ERK are required for initial c-Fos induction, c-Fos is later required to increase CREB expression as well.94 The first c-Fos induction step is required to form LTM, while the second CREB induction is necessary to prolong LTM for at least seven days.88

c-Fos expression in AD

AD accounts for 60–70% of all dementia cases, and approximately 10% of affected individuals experience cognitive decline.95 Pathologically, AD is characterized by the accumulation of amyloid beta (Aβ) plaques and neurofibrillary tangles composed of hyperphosphorylated tau protein. These proteins disrupt neuronal function and contribute to neuroinflammation, which is increasingly recognized as a critical factor in the progression of AD.96 Despite significant research efforts, the etiology and initial molecular signaling events in AD remain unknown, emphasizing the need for further investigation.

According to the literature, c-Fos is involved in age-dependent cognitive decline 12 and various inflammatory processes,20 which are further accompanied by synaptic loss. Elevated c-Fos expression has been reported in postmortem brain tissue from AD patients and is positively associated with cognitive impairment.66,97,98 Moreover, some data show a positive association between AD, Aβ production, and hippocampal hyperactivity,99,100 in line with experiments on facilitating spikes in hippocampal synapses and stimulation of excitatory CA1 neurons in 5xFAD mice.101,102

Amyloid β 1-42 causes a rapid and sustained increase in c-Fos expression in a mouse hippocampal cell line, contributing to neurotoxicity, which was abolished by the administration of c-Fos antisense oligodeoxynucleotides.22 Supporting this, several animal studies reported elevated levels of c-Fos protein in the hippocampus of rat models of Aβ.51,67,68 Interestingly, increased neuronal accumulation of Aβ and c-Fos expression were observed in glutamatergic neurons of the motor cortex in hyperactive double-transgenic mice models of Alzheimer’s disease expressing both amyloid precursor protein and presenilin 1 (APP/PS1),103 as well as in APP swe (Swedish mutation of amyloid precursor protein) and α-synuclein transgenic mice.104–108

Aβ42 elevation may trigger excessive neuronal c-Fos expression,109 contributing to apoptotic signaling.22 In addition, inflammatory markers, such as LPS and interleukins, which are elevated in AD, also induce c-Fos expression in astrocytes.36 Subsequently, activation of ERK/FOS signaling intensifies the inflammatory response and apoptosis,110,111 whereas inhibition of the ERK/FOS pathway reduces levels of inflammation and apoptosis.69,112 In line with this, co-treatment with c-Fos inhibitors and antioxidants showed a positive effect on cognition in neurodegenerative models.113

Consistent with these findings, activation of NMDA-R following rat hyperactivity might cause neuronal loss through Ca2+ and c-Fos signaling.70,114 Meanwhile, in an AD mouse model, c-Fos expression in visual cortical networks correlated with impaired visual experience-dependent memory in a pre-amyloid plaque stage.15

In amyloid neurotoxicity, abnormal c-Fos expression is likely downstream of Aβ elevation and subsequent Ca2+ influx. The expressed c-Fos protein is additionally stabilized by Aβ42-induced O-GlcNAcylation, consequently leading to the activation of pro-apoptotic AP-1.22 Despite all the aforementioned evidence, the relationship between Aβ and c-Fos, is not straightforward. For instance, APP knockout mice displays elevated c-Fos mRNA expression in the prefrontal cortex, while c-Fos gene mutations are rare in hereditary AD and do not directly drive disease phenotypes.24,115

To explain the dichotomy of c-Fos response in normal and AD states, one possibility is that, higher baseline c-Fos levels in hAPP mice may result from a defect in their ability to habituate to repeated everyday experiences, in contrast with normal/control mice, which show lower baseline c-Fos expression.103–108 Collectively, transient expression under healthy conditions appears to be homeostatic, whereas continuous c-Fos expression may disrupt synaptic activity and lead to neuronal death. From a clinical perspective, detecting c-Fos immunoreactivity in AD brains using sensitive imaging techniques may represent an early marker of neurodegeneration.71,114

c-Fos expression in glial cells of AD subjects

In addition to neurons, which show expression of c-Fos in the presence of hyperactivation, neuroinflammation in AD, and glutamate toxicity, c-Fos rapidly increase in astrocytes as well.1,36 Cytokine elevation in AD also induces glial expression of IEGs, which regulate complement factor H and lead to neuronal loss in AD,33 as evidenced by LPS-induced c-Fos expression in astrocytes of the spinal cord in AD.64 Therefore, c-Fos expression in glial cells contributes to neuroinflammation and neuronal loss in AD.

Possible mechanisms of c-Fos in AD

The role of c-Fos in the pathophysiology of AD has not yet been fully understood, and whether c-Fos activation is causative or compensatory requires further investigation. There is no doubt that possible factors initiating AD, including inflammation, oxidative stress, Ca2+ toxicity, glutamate dysregulation, epigenetic alterations, brain metabolic disturbances, and imbalance in neural activity are likely to occur years before symptoms appear, or a diagnosis is made. Therefore, glutamate toxicity,67 elevation in intracellular calcium levels, and reactive oxygen species (ROS) not only independently of c-Fos, initiate neural apoptosis, but also may contribute to synaptic and neural loss through c-Fos.51,70,73 According to evidence, overexpression of c-Fos in the hippocampal neurons may contribute to neurodegeneration, highlighting its potential role in the disease’s progression.72–74

As we reviewed above, in AD which is a chronic condition, neural hyperactivation causes expression without habituation, in contrast to healthy conditions. How can c-Fos differentiate these distinct physiological and pathological situations? One possible explanation might be, the variety of novel upstream extrinsic and intrinsic stimuli in the microenvironment of affected cells in AD, including neurochemical, electrical, metabolic, oxidative stress, calcium, and abnormal protein products of organelles, which cumulatively intensify c-Fos expression. Regrettably, some of these factors contribute to a vicious cycle in AD progression and exacerbate the disease pathology. For example, increased Ca2+ influx into neurons through different pathways,75,116–118 and binding with calmodulin, activates CaMKII, CREB, and MAPK.76,118–120 These regulatory elements bind to the promoter of the c-Fos gene and synthesize Fos protein,75,121 which returns to the nucleus and acts on target genes like Aβ, increasing its expression.21 Then Aβ causes a rapid increase in intracellular hydrogen peroxide in neurons, which may be the signal for c-Fos activation.71 Finally, Fos dimerizes with c-Jun and acts as a transcriptional regulator at the AP-1 binding site of DNA,22 contributing to programmed cell death.65,77

Another explanation for c-Fos elevation in AD is the dysregulation of c-Fos O-GlcNAcylation,23,122–124 which reduces the interaction between OGA and c-Fos,125 resulting in higher transcriptional activity of the c-Fos/c-Jun complex, to downregulate genes including CREB and brain-derived neurotrophic factor (BDNF); two proteins highly involved in memory formation under physiological conditions.126,127 Moreover, c-Fos might act as an epigenetic regulator, modifying chromatin accessibility around a subset of its binding sites across the genome in amyloid neurotoxicity in rats and AD-related hyperactivity.12,67,68,128 For example, ΔFosB binding to c-Fos promoter triggers histone deacetylation,12 and inhibits memory formation (Fig. 1).129 Moreover FOS dimerization with Jun exerts positive modulation, binding with activation transcription factor 3 (ATF3) leads in neural loss.1 It should be noted that the effect of some c-Fos upstream molecules, such as CREB, depends on the level and duration of activation.130 In fact, constitutive CREB activation causes chronic c-Fos expression and leads to memory deficits,131 cognitive decline,132 memory retrieval deficits,131 and neurodegeneration.133 Some of the upstream signaling pathways underlying c-Fos expression in AD have been summarized in Table 1.

Proposed mechanisms by which c-Fos contribute to neuroprotection or neurodegeneration outcome in hippocampus.
Fig. 1  Proposed mechanisms by which c-Fos contribute to neuroprotection or neurodegeneration outcome in hippocampus.

Neural activity in healthy brain releases glutamate which binds to synaptic receptors and activates Ca++ influx and further signaling pathways of CAM/ERK/MAPK/CREB in neurons. CREB elevation leads in memory improvement via expression of c-Fos, and BDNF. Then the FOS protein is returns to the nucleus and acts together with c-Jun by binding and forming the activator protein-1 (AP-1) heterodimer, and leads in long term memory. On the other hand, hyper activation of extra synaptic NMDA receptors elevates ROS and Aβ toxicity, and increase transcription of c-Fos. The expressed c-Fos protein is additionally stabilized by Aβ42-induced O-GlcNAcylation, and the glycosylated c-Fos binds with c-Jun stimulates neuronal cell death by activating the apoptotic factor Bim, ATF3 and cytochrome c, besides, it downregulates CREB and BDNF which leads in memory impairment. AD, Alzheimer’s disease; AMPA, α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptor; AP-1, activator protein-1; ATF, activation transcription factor; Aβ, amyloid beta; BDNF, brain derived neurotrophic factor; BIM, B-cell lymphoma 2 interacting mediator of cell death; CREB, cAMP response element binding; ERK, extracellular signal-regulated kinase; MAPK, mitogen-activated protein kinase; MEK, mitogen-activated protein kinase kinase; NMDA, N-methyl-D-aspartate; PKA, protein kinase A; PKC, protein kinase C; RAF, rapidly accelerated fibrosarcoma kinase.

Collectively, in AD, which is a chronic pathological state, one or more stimuli initiate excitotoxic neuronal activation, which then spreads molecular signaling to glial cells, thereby intensifying neuroinflammation. Below, we explain apoptosis as a mechanism of c-Fos-mediated neuronal death in more detail.

c-Fos and apoptosis in AD

Regulated cell death is a cluster of signaling events involving both gene expression and enzyme activity, resulting in neural death, by various mechanisms including apoptosis, necroptosis, pyroptosis, ferroptosis, and autophagy-dependent cell death in AD.97,134 Sajan et al.97 compared the expression of 14 apoptotic genes between normal and AD human hippocampi and noted upregulation in gene expression for c-Fos and BAK in AD patients, suggesting a role for these genes in the apoptotic cascade of AD. Also, Lee et al.135,136 highlighted increased ATF immunoreactivity within the nuclei of hippocampal pyramidal CA1 neurons in early-stage, and CA2 neurons in late-stage AD, compared to age-matched healthy control brains.

c-Fos expression is linked to apoptosis or hippocampal cell death, which is a significant mechanism of neurodegeneration.69,78,79,137 Fos can increase apoptosis via ATF3, which further inhibits the activity of the PTEN induced kinase 1 (PINK1) promoter and causes cell death.80,138 It is known that glycosylated c-Fos binds with c-Jun and stimulates neuronal cell death by activating the apoptotic factor Bim,22,139 which translocates to the mitochondria to form pores that release cytochrome c and promote cell death,22,140 while inhibition of the ERK/FOS pathway reduces apoptosis.69,78,112 Finally, the finding of synaptic degeneration preceding neuronal loss and memory impairment in AD patients, may be related to the apoptotic roles of c-Fos via ATF3 or complement factor H (Fig. 1).33,141–143

Oxidative stress and c-Fos expression in AD

Exposure of neurons to multiple stresses and divergent cytotoxic mechanisms including elevation in ROS levels, synaptic dysfunction, excitotoxicity, ER stress, inflammation, and mitochondrial dysfunction results in neuronal cell death.142,144,145 Oxidative stress plays a central role in AD pathogenesis by disrupting cellular homeostasis and triggering multiple signaling cascades that contribute to neurodegeneration.144

In AD, oxidative stress is primarily driven by mitochondrial dysfunction, Aβ aggregation, and neuroinflammation, which together activate various redox-sensitive signaling pathways, exacerbating neuronal damage via activation of the nuclear factor erythroid 2-related factor 2 (NRF2) signaling pathway.146NRF2 is a key transcription factor that regulates the antioxidant response by inducing the expression of detoxifying enzymes, such as heme oxygenase-1, superoxide dismutase, and glutathione peroxidase.147,148

Increased oxidative stress results in the hyperactivation of JNK and ERK, which in turn upregulate c-Fos expression.81,82 Activation of c-Fos not only leads to programmed cell death,83 but also disrupts mitochondrial function, which amplifies oxidative damage and neuroinflammation through increased inflammatory cytokines.1,84–86,149–151

Collectively, ROS-induced c-Fos contributes to AD progression not only through the production of Aβ at early stages, but also through apoptosis and neuronal death at later stages. Then, neuronal death reduces brain volume and leads to hyperactivation of neurons, which exacerbates the production of Aβ.

Conclusions

In this narrative review, we compiled evidence from cell cultures, animal, and human studies supporting the relevance of c-Fos for learning and memory in neurons and glia. Although, the brains of AD patients, differ in terms of neural network synchrony and epigenetic regulation, which alter the response to stimuli at the levels of c-Fos expression compared with normal conditions. c-Fos contributes to AD progression not only through the production of Aβ at early stages, but also through apoptosis and neuronal death at later stages, through prolonged CREB activation and increasing ATF3, Bim, and cytochrome c. The relationship between c-Fos expression and AD suggests it as a biomarker for disease progression or a possible target for therapeutic intervention. Thus, tracing of c-Fos alterations in both neurons and glia might offer a useful and reliable understanding of physiological or pathological responses to specific stimuli.

Declarations

Acknowledgement

The corresponding author would like to express their deep gratitude to Professor K.V. Anokhin from Anokhin Institute of Normal Physiology, Academy of Medical Sciences, Moscow, Russia for his thoughtful ideas and rich knowledge on c-Fos.

Funding

There was no funding for this review.

Conflict of interest

The authors have declared that no competing interests exist.

Authors’ contributions

Conception, manuscript writing, reference update (PB), table, figure drawing, drafting of the manuscript (NF), data searching (KE), data bank searching, writing, revision (PB, NF). All authors have made a significant contribution to this study and have approved the final manuscript.

References

  1. Cruz-Mendoza F, Jauregui-Huerta F, Aguilar-Delgadillo A, García-Estrada J, Luquin S. Immediate Early Gene c-fos in the Brain: Focus on Glial Cells. Brain Sci 2022;12(6):687 View Article PubMed/NCBI
  2. Healy S, Khan P, Davie JR. Immediate early response genes and cell transformation. Pharmacol Ther 2013;137(1):64-77 View Article PubMed/NCBI
  3. Morgan JI, Cohen DR, Hempstead JL, Curran T. Mapping patterns of c-fos expression in the central nervous system after seizure. Science 1987;237(4811):192-197 View Article PubMed/NCBI
  4. Groves A, Kihara Y, Jonnalagadda D, Rivera R, Kennedy G, Mayford M, et al. A Functionally Defined In Vivo Astrocyte Population Identified by c-Fos Activation in a Mouse Model of Multiple Sclerosis Modulated by S1P Signaling: Immediate-Early Astrocytes (ieAstrocytes). eNeuro 2018;5(5):e0239-18.2018 View Article PubMed/NCBI
  5. Minatohara K, Akiyoshi M, Okuno H. Role of Immediate-Early Genes in Synaptic Plasticity and Neuronal Ensembles Underlying the Memory Trace. Front Mol Neurosci 2015;8:78 View Article PubMed/NCBI
  6. Christensen DZ, Thomsen MS, Mikkelsen JD. Reduced basal and novelty-induced levels of activity-regulated cytoskeleton associated protein (Arc) and c-Fos mRNA in the cerebral cortex and hippocampus of APPswe/PS1ΔE9 transgenic mice. Neurochem Int 2013;63(1):54-60 View Article PubMed/NCBI
  7. Carter SD, Mifsud KR, Reul JMHM. Acute Stress Enhances Epigenetic Modifications But Does Not Affect the Constitutive Binding of pCREB to Immediate-Early Gene Promoters in the Rat Hippocampus. Front Mol Neurosci 2017;10:416 View Article PubMed/NCBI
  8. Xu C, Huang H, Zhang M, Zhang P, Li Z, Liu X, et al. Methyltransferase-Like 3 Rescues the Amyloid-beta protein-Induced Reduction of Activity-Regulated Cytoskeleton Associated Protein Expression via YTHDF1-Dependent N6-Methyladenosine Modification. Front Aging Neurosci 2022;14:890134 View Article PubMed/NCBI
  9. Hagenston AM, Bading H, Bas-Orth C. Functional Consequences of Calcium-Dependent Synapse-to-Nucleus Communication: Focus on Transcription-Dependent Metabolic Plasticity. Cold Spring Harb Perspect Biol 2020;12(4):a035287 View Article PubMed/NCBI
  10. Pompeiano M, Colonnese MT. cFOS as a biomarker of activity maturation in the hippocampal formation. Front Neurosci 2023;17:929461 View Article PubMed/NCBI
  11. Aguilar-Delgadillo A, Cruz-Mendoza F, Luquin-de Andais Teh S, Ruvalcaba-Delgadillo Y, Jáuregui-Huerta F. Stress-induced c-fos expression in the medial prefrontal cortex differentially affects the main residing cell phenotypes. Heliyon 2024;10(20):e39325 View Article PubMed/NCBI
  12. Corbett BF, You JC, Zhang X, Pyfer MS, Tosi U, Iascone DM, et al. ΔFosB Regulates Gene Expression and Cognitive Dysfunction in a Mouse Model of Alzheimer’s Disease. Cell Rep 2017;20(2):344-355 View Article PubMed/NCBI
  13. Babai P, Anokhin KV, Dolgov N, Sudakov KV. Characteristics of c-fos gene expression in the brains of rats with different investigative and defensive behaviors. Neurosci Behav Physiol 2001;31(6):583-588 View Article PubMed/NCBI
  14. de Bartolomeis A, Buonaguro EF, Latte G, Rossi R, Marmo F, Iasevoli F, et al. Immediate-Early Genes Modulation by Antipsychotics: Translational Implications for a Putative Gateway to Drug-Induced Long-Term Brain Changes. Front Behav Neurosci 2017;11:240 View Article PubMed/NCBI
  15. L’esperance OJ, McGhee J, Davidson G, Niraula S, Smith AS, Sosunov AA, et al. Functional connectivity favors aberrant visual network c-Fos expression accompanied by cortical synapse loss in a mouse model of Alzheimer’s disease. J Alzheimers Dis 2024;101(1):111-131 View Article PubMed/NCBI
  16. Stefaniuk M, Pawłowska M, Barański M, Nowicka K, Zieliński Z, Bijoch Ł, et al. Global brain c-Fos profiling reveals major functional brain networks rearrangements after alcohol reexposure. Neurobiol Dis 2023;178:106006 View Article PubMed/NCBI
  17. Shah AJ, Mir PA, Adnan M, Patel M, Maqbool M, Mir RH, et al. Synthetic and Natural Bioactive Molecules in Balancing the Crosstalk among Common Signaling Pathways in Alzheimer’s Disease: Understanding the Neurotoxic Mechanisms for Therapeutic Intervention. ACS Omega 2023;8(43):39964-39983 View Article PubMed/NCBI
  18. Scheltens P, Blennow K, Breteler MM, de Strooper B, Frisoni GB, Salloway S, et al. Alzheimer’s disease. Lancet 2016;388(10043):505-517 View Article PubMed/NCBI
  19. Safiri S, Ghaffari Jolfayi A, Fazlollahi A, Morsali S, Sarkesh A, Daei Sorkhabi A, et al. Alzheimer’s disease: a comprehensive review of epidemiology, risk factors, symptoms diagnosis, management, caregiving, advanced treatments and associated challenges. Front Med (Lausanne) 2024;11:1474043 View Article PubMed/NCBI
  20. Xu C, Zhang M, Zu L, Zhang P, Sun L, Liu X, et al. Repressor element-1 silencing transcription factor regulates glutamate receptors and immediate early genes to affect synaptic plasticity. Aging (Albany NY) 2021;13(11):15569-15579 View Article PubMed/NCBI
  21. MacGibbon GA, Lawlor PA, Walton M, Sirimanne E, Faull RL, Synek B, et al. Expression of Fos, Jun, and Krox family proteins in Alzheimer’s disease. Exp Neurol 1997;147(2):316-332 View Article PubMed/NCBI
  22. Choi H, Kim C, Song H, Cha MY, Cho HJ, Son SM, et al. Amyloid β-induced elevation of O-GlcNAcylated c-Fos promotes neuronal cell death. Aging Cell 2019;18(1):e12872 View Article PubMed/NCBI
  23. Förster S, Welleford AS, Triplett JC, Sultana R, Schmitz B, Butterfield DA. Increased O-GlcNAc levels correlate with decreased O-GlcNAcase levels in Alzheimer disease brain. Biochim Biophys Acta 2014;1842(9):1333-1339 View Article PubMed/NCBI
  24. Hendrickx A, Pierrot N, Tasiaux B, Schakman O, Kienlen-Campard P, De Smet C, et al. Epigenetic regulations of immediate early genes expression involved in memory formation by the amyloid precursor protein of Alzheimer disease. PLoS One 2014;9(6):e99467 View Article PubMed/NCBI
  25. Chung L. A Brief Introduction to the Transduction of Neural Activity into Fos Signal. Dev Reprod 2015;19(2):61-67 View Article PubMed/NCBI
  26. Miao B, Yao H, Chen P, Song XJ. Differential Activation of pERK1/2 and c-Fos Following Injury to Different Regions of Primary Sensory Neuron. Life (Basel) 2022;12(5):752 View Article PubMed/NCBI
  27. Chowdhury A, Caroni P. Time units for learning involving maintenance of system-wide cFos expression in neuronal assemblies. Nat Commun 2018;9(1):4122 View Article PubMed/NCBI
  28. Pinto M, Lima D, Tavares I. Correlation of noxious evoked c-fos expression in areas of the somatosensory system during chronic pain: involvement of spino-medullary and intra-medullary connections. Neurosci Lett 2006;409(2):100-105 View Article PubMed/NCBI
  29. Zhang L, Cao C, Luo C, Ruan H, Xu C, Wang Y, et al. Comparison of chronic restraint stress-and lipopolysaccharide-induced mouse models of depression: Behavior, c-Fos expression, and microglial and astrocytic activation. J Neurorestoratology 2024;12(3):100130 View Article PubMed/NCBI
  30. Zhang P, Hirsch EC, Damier P, Duyckaerts C, Javoy-Agid F. c-fos protein-like immunoreactivity: distribution in the human brain and over-expression in the hippocampus of patients with Alzheimer’s disease. Neuroscience 1992;46(1):9-21 View Article PubMed/NCBI
  31. Xu C, Li Q, Efimova O, Jiang X, Petrova M, Vinarskaya AK, et al. Identification of Immediate Early Genes in the Nervous System of Snail Helix lucorum. eNeuro 2019;6(3):e0416-18.2019 View Article PubMed/NCBI
  32. Fan F, Li L, Liu W, Yang M, Ma X, Sun H. Astrocytes and neurons in locus coeruleus mediate restraint water immersion stress-induced gastric mucosal damage through the ERK1/2 signaling pathway. Neurosci Lett 2018;675:95-102 View Article PubMed/NCBI
  33. Suh HW, Choi SS, Lee JK, Lee HK, Han EJ, Lee J. Regulation of c-fos and c-jun gene expression by lipopolysaccharide and cytokines in primary cultured astrocytes: effect of PKA and PKC pathways. Arch Pharm Res 2004;27(4):396-401 View Article PubMed/NCBI
  34. Simi A, Edling Y, Ingelman-Sundberg M, Tindberg N. Activation of c-fos by lipopolysaccharide in glial cells via p38 mitogen-activated protein kinase-dependent activation of serum or cyclic AMP/calcium response element. J Neurochem 2005;92(4):915-924 View Article PubMed/NCBI
  35. Flores EN, Duggan A, Madathany T, Hogan AK, Márquez FG, Kumar G, et al. A non-canonical pathway from cochlea to brain signals tissue-damaging noise. Curr Biol 2015;25(5):606-612 View Article PubMed/NCBI
  36. Edling Y, Ingelman-Sundberg M, Simi A. Glutamate activates c-fos in glial cells via a novel mechanism involving the glutamate receptor subtype mGlu5 and the transcriptional repressor DREAM. Glia 2007;55(3):328-340 View Article PubMed/NCBI
  37. Dragunow M, Currie RW, Robertson HA, Faull RL. Heat shock induces c-fos protein-like immunoreactivity in glial cells in adult rat brain. Exp Neurol 1989;106(1):105-109 View Article PubMed/NCBI
  38. Chan A, Hummel V, Weilbach FX, Kieseier BC, Gold R. Phagocytosis of apoptotic inflammatory cells downregulates microglial chemoattractive function and migration of encephalitogenic T cells. J Neurosci Res 2006;84(6):1217-1224 View Article PubMed/NCBI
  39. Jauregui-Huerta F, Ruvalcaba-Delgadillo Y, Gonzalez-Castañeda R, Garcia-Estrada J, Gonzalez-Perez O, Luquin S. Responses of glial cells to stress and glucocorticoids. Curr Immunol Rev 2010;6(3):195-204 View Article PubMed/NCBI
  40. Kim W, Chung C. Brain-wide cellular mapping of acute stress-induced activation in male and female mice. FASEB J 2021;35(12):e22041 View Article PubMed/NCBI
  41. Sominsky L, De Luca S, Spencer SJ. Microglia: Key players in neurodevelopment and neuronal plasticity. Int J Biochem Cell Biol 2018;94:56-60 View Article PubMed/NCBI
  42. Aguilar-Delgadillo A, Cruz-Mendoza F, Luquin S, Ruvalcaba-Delgadillo Y, Jáuregui-Huerta F. Stress-induced c-fos expression in the medial prefrontal cortex of male rats differentially involves the main glial cell phenotypes. Heliyon 2024;10(20):e39325 View Article PubMed/NCBI
  43. Wilson CS, Mongin AA. The signaling role for chloride in the bidirectional communication between neurons and astrocytes. Neurosci Lett 2019;689:33-44 View Article PubMed/NCBI
  44. Adamsky A, Kol A, Kreisel T, Doron A, Ozeri-Engelhard N, Melcer T, et al. Astrocytic Activation Generates De Novo Neuronal Potentiation and Memory Enhancement. Cell 2018;174(1):59-71.e14 View Article PubMed/NCBI
  45. Li JL, Xiong KH, Dong YL, Fujiyama F, Kaneko T, Mizuno N. Vesicular glutamate transporters, VGluT1 and VGluT2, in the trigeminal ganglion neurons of the rat, with special reference to coexpression. J Comp Neurol 2003;463(2):212-220 View Article PubMed/NCBI
  46. Graziano A, Liu XB, Murray KD, Jones EG. Vesicular glutamate transporters define two sets of glutamatergic afferents to the somatosensory thalamus and two thalamocortical projections in the mouse. J Comp Neurol 2008;507(2):1258-1276 View Article PubMed/NCBI
  47. Birey F, Kokkosis AG, Aguirre A. Oligodendroglia-lineage cells in brain plasticity, homeostasis and psychiatric disorders. Curr Opin Neurobiol 2017;47:93-103 View Article PubMed/NCBI
  48. Liu G, Shen C, Qiu A. Amyloid-β accumulation in relation to functional connectivity in aging: A longitudinal study. Neuroimage 2023;275:120146 View Article PubMed/NCBI
  49. Guzowski JF. Insights into immediate-early gene function in hippocampal memory consolidation using antisense oligonucleotide and fluorescent imaging approaches. Hippocampus 2002;12(1):86-104 View Article PubMed/NCBI
  50. Hafner AS, Donlin-Asp PG, Leitch B, Herzog E, Schuman EM. Local protein synthesis is a ubiquitous feature of neuronal pre- and postsynaptic compartments. Science 2019;364(6441):eaau3644 View Article PubMed/NCBI
  51. Köylü A, Altunkaynak BZ, Delibaş B. Effects of tacrolimus on c-fos in hippocampus and memory performances in streptozotocin model of Alzheimer’s disease of rats. Turk J Med Sci 2021;51(4):2159-2166 View Article PubMed/NCBI
  52. Ivashkina OI, Gruzdeva AM, Roshchina MA, Toropova KA, Anokhin KV. Imaging of C-fos Activity in Neurons of the Mouse Parietal Association Cortex during Acquisition and Retrieval of Associative Fear Memory. Int J Mol Sci 2021;22(15):8244 View Article PubMed/NCBI
  53. Zamorina TA, Ivashkina OI, Toropova KA, Anokhin KV. Inhibition of Protein Synthesis Attenuates Formation of Traumatic Memory and Normalizes Fear-Induced c-Fos Expression in a Mouse Model of Posttraumatic Stress Disorder. Int J Mol Sci 2024;25(12):6544 View Article PubMed/NCBI
  54. Pettit NL, Yap EL, Greenberg ME, Harvey CD. Fos ensembles encode and shape stable spatial maps in the hippocampus. Nature 2022;609(7926):327-334 View Article PubMed/NCBI
  55. Fleischmann A, Hvalby O, Jensen V, Strekalova T, Zacher C, Layer LE, et al. Impaired long-term memory and NR2A-type NMDA receptor-dependent synaptic plasticity in mice lacking c-Fos in the CNS. J Neurosci 2003;23(27):9116-9122 View Article PubMed/NCBI
  56. Anokhin KV, Rose SP. Learning-induced Increase of Immediate Early Gene Messenger RNA in the Chick Forebrain. Eur J Neurosci 1991;3(2):162-167 View Article PubMed/NCBI
  57. Saha RN, Dudek SM. Splitting hares and tortoises: a classification of neuronal immediate early gene transcription based on poised RNA polymerase II. Neuroscience 2013;247:175-181 View Article PubMed/NCBI
  58. Babaei P, Dolgov ON, Sudakov KV, Anokhin KV. Stress-induced expression c-fos in the rat brain: A comparison of averaging and typological analysis. Neurosci Res Commun 2000;27(2):95-102 View Article PubMed/NCBI
  59. Besnard A, Bouveyron N, Kappes V, Pascoli V, Pagès C, Heck N, et al. Alterations of molecular and behavioral responses to cocaine by selective inhibition of Elk-1 phosphorylation. J Neurosci 2011;31(40):14296-14307 View Article PubMed/NCBI
  60. Takigami S, Sunada H, Lukowiak K, Kuzirian AM, Alkon DL, Sakakibara M. Protein kinase C mediates memory consolidation of taste avoidance conditioning in Lymnaea stagnalis. Neurobiol Learn Mem 2014;111:9-18 View Article PubMed/NCBI
  61. Zhong S, Zhang S, Fan X, Wu Q, Yan L, Dong J, et al. A single-cell RNA-seq survey of the developmental landscape of the human prefrontal cortex. Nature 2018;555(7697):524-528 View Article PubMed/NCBI
  62. Wirsching HG, Galanis E, Weller M. Glioblastoma. Handb Clin Neurol 2016;134:381-397 View Article PubMed/NCBI
  63. Boehm J, Kang MG, Johnson RC, Esteban J, Huganir RL, Malinow R. Synaptic incorporation of AMPA receptors during LTP is controlled by a PKC phosphorylation site on GluR1. Neuron 2006;51(2):213-225 View Article PubMed/NCBI
  64. Yokoyama K, Hiyama A, Arai F, Nukaga T, Sakai D, Mochida J. C-Fos regulation by the MAPK and PKC pathways in intervertebral disc cells. PLoS One 2013;8(9):e73210 View Article PubMed/NCBI
  65. Ameyar M, Wisniewska M, Weitzman JB. A role for AP-1 in apoptosis: the case for and against. Biochimie 2003;85(8):747-752 View Article PubMed/NCBI
  66. Marcus DL, Strafaci JA, Miller DC, Masia S, Thomas CG, Rosman J, et al. Quantitative neuronal c-fos and c-jun expression in Alzheimer’s disease. Neurobiol Aging 1998;19(5):393-400 View Article PubMed/NCBI
  67. Faraji N, Badrikoohi M, Babaei P. Effect of BRD4 Inhibitor on Cognitive Deficit and c-Fos /BDNF level in rats with Alzheimer’s disease. Neurosci Behav Physiol 2023;53(4):678-687 View Article PubMed/NCBI
  68. Eyvani K, Letafatkar N, Babaei P. AMPA Receptors Endocytosis Inhibition Attenuates Cognition Deficit Via c-Fos/BDNF Signaling in Amyloid β Neurotoxicity. Exp Aging Res 2024;51(3):303-315 View Article PubMed/NCBI
  69. Xu X, Jiang R, Gong P, Liu Q, Chen Y, Hou S, et al. Up-regulation of FOS-like antigen 1 contributes to neuronal apoptosis in the cortex of rat following traumatic brain injury. Metab Brain Dis 2018;33(1):115-125 View Article PubMed/NCBI
  70. Ghosh A, Giese KP. Calcium/calmodulin-dependent kinase II and Alzheimer’s disease. Mol Brain 2015;8(1):78 View Article PubMed/NCBI
  71. Gillardon F, Skutella T, Uhlmann E, Holsboer F, Zimmermann M, Behl C. Activation of c-Fos contributes to amyloid beta-peptide-induced neurotoxicity. Brain Res 1996;706(1):169-172 View Article PubMed/NCBI
  72. Lacor PN, Buniel MC, Furlow PW, Clemente AS, Velasco PT, Wood M, et al. Abeta oligomer-induced aberrations in synapse composition, shape, and density provide a molecular basis for loss of connectivity in Alzheimer’s disease. J Neurosci 2007;27(4):796-807 View Article PubMed/NCBI
  73. Mohanan AG, Gunasekaran S, Jacob RS, Omkumar RV. Role of Ca(2+)/Calmodulin-Dependent Protein Kinase Type II in Mediating Function and Dysfunction at Glutamatergic Synapses. Front Mol Neurosci 2022;15:855752 View Article PubMed/NCBI
  74. Lu W, Mi R, Tang H, Liu S, Fan M, Wang L. Over-expression of c-fos mRNA in the hippocampal neurons in Alzheimer’s disease. Chin Med J (Engl) 1998;111(1):35-37 View Article PubMed/NCBI
  75. Hsieh HL, Wang HH, Wu CY, Yang CM. Reactive Oxygen Species-Dependent c-Fos/Activator Protein 1 Induction Upregulates Heme Oxygenase-1 Expression by Bradykinin in Brain Astrocytes. Antioxid Redox Signal 2010;13(12):1829-1844 View Article PubMed/NCBI
  76. Roux PP, Blenis J. ERK and p38 MAPK-activated protein kinases: a family of protein kinases with diverse biological functions. Microbiol Mol Biol Rev 2004;68(2):320-344 View Article PubMed/NCBI
  77. Shaulian E, Karin M. AP-1 in cell proliferation and survival. Oncogene 2001;20(19):2390-2400 View Article PubMed/NCBI
  78. Markopoulou S, Kontargiris E, Batsi C, Tzavaras T, Trougakos I, Boothman DA, et al. Vanadium-induced apoptosis of HaCaT cells is mediated by c-fos and involves nuclear accumulation of clusterin. FEBS J 2009;276(14):3784-3799 View Article PubMed/NCBI
  79. Bejjani F, Evanno E, Zibara K, Piechaczyk M, Jariel-Encontre I. The AP-1 transcriptional complex: Local switch or remote command?. Biochim Biophys Acta Rev Cancer 2019;1872(1):11-23 View Article PubMed/NCBI
  80. Yuan Z, Gong S, Luo J, Zheng Z, Song B, Ma S, et al. Opposing roles for ATF2 and c-Fos in c-Jun-mediated neuronal apoptosis. Mol Cell Biol 2009;29(9):2431-2442 View Article PubMed/NCBI
  81. Espinosa-Diez C, Miguel V, Mennerich D, Kietzmann T, Sánchez-Pérez P, Cadenas S, et al. Antioxidant responses and cellular adjustments to oxidative stress. Redox Biol 2015;6:183-197 View Article PubMed/NCBI
  82. Cui J, Holmes EH, Liu PK. Oxidative damage to the c-fos gene and reduction of its transcription after focal cerebral ischemia. J Neurochem 1999;73(3):1164-1174 View Article PubMed/NCBI
  83. Weinreb O, Mandel S, Youdim MBH. Concentration-Dependent Gene and Protein Expressions of Neuroprotective and Neurotoxic Activities of Antioxidants, Including Nutrients. Nutrigenomics. CRC Press; 2005:123-140 View Article PubMed/NCBI
  84. Li N, Oquendo E, Capaldi RA, Robinson JP, He YD, Hamadeh HK, et al. A systematic assessment of mitochondrial function identified novel signatures for drug-induced mitochondrial disruption in cells. Toxicol Sci 2014;142(1):261-273 View Article PubMed/NCBI
  85. Guo C, Sun L, Chen X, Zhang D. Oxidative stress, mitochondrial damage and neurodegenerative diseases. Neural Regen Res 2013;8(21):2003-2014 View Article PubMed/NCBI
  86. Hong Y, Boiti A, Vallone D, Foulkes NS. Reactive Oxygen Species Signaling and Oxidative Stress: Transcriptional Regulation and Evolution. Antioxidants (Basel) 2024;13(3):312 View Article PubMed/NCBI
  87. Wang H, Peng RY. Basic roles of key molecules connected with NMDAR signaling pathway on regulating learning and memory and synaptic plasticity. Mil Med Res 2016;3(1):26 View Article PubMed/NCBI
  88. Miyashita T, Kikuchi E, Horiuchi J, Saitoe M. Long-Term Memory Engram Cells Are Established by c-Fos/CREB Transcriptional Cycling. Cell Rep 2018;25(10):2716-2728.e3 View Article PubMed/NCBI
  89. Johansen JP, Cain CK, Ostroff LE, LeDoux JE. Molecular mechanisms of fear learning and memory. Cell 2011;147(3):509-524 View Article PubMed/NCBI
  90. Anokhin KV, Ryabinin AE, Sudakov KV. Expression of the c-fos gene in the mouse brain during the acquisition of defensive behavior habits. Neurosci Behav Physiol 2001;31(2):139-143 View Article PubMed/NCBI
  91. Miyashita T, Kubik S, Lewandowski G, Guzowski JF. Networks of neurons, networks of genes: an integrated view of memory consolidation. Neurobiol Learn Mem 2008;89(3):269-284 View Article PubMed/NCBI
  92. Rylski M, Kaczmarek L. Ap-1 targets in the brain. Front Biosci 2004;9:8-23 View Article PubMed/NCBI
  93. Ahn JH, Shin MC, Park JH, Kim IH, Lee JC, Yan BC, et al. Increased immunoreactivity of c-Fos in the spinal cord of the aged mouse and dog. Mol Med Rep 2015;11(2):1043-1048 View Article PubMed/NCBI
  94. Sanyal S, Sandstrom DJ, Hoeffer CA, Ramaswami M. AP-1 functions upstream of CREB to control synaptic plasticity in Drosophila. Nature 2002;416(6883):870-874 View Article PubMed/NCBI
  95. Kim J, Kaang BK. Cyclic AMP response element-binding protein (CREB) transcription factor in astrocytic synaptic communication. Front Synaptic Neurosci 2022;14:1059918 View Article PubMed/NCBI
  96. Iqbal K, Liu F, Gong CX, Grundke-Iqbal I. Tau in Alzheimer disease and related tauopathies. Curr Alzheimer Res 2010;7(8):656-664 View Article PubMed/NCBI
  97. Sajan FD, Martiniuk F, Marcus DL, Frey WH, Hite R, Bordayo EZ, et al. Apoptotic gene expression in Alzheimer’s disease hippocampal tissue. Am J Alzheimers Dis Other Demen 2007;22(4):319-328 View Article PubMed/NCBI
  98. Rao JS, Harry GJ, Rapoport SI, Kim HW. Increased excitotoxicity and neuroinflammatory markers in postmortem frontal cortex from bipolar disorder patients. Mol Psychiatry 2010;15(4):384-392 View Article PubMed/NCBI
  99. Cirrito JR, Kang JE, Lee J, Stewart FR, Verges DK, Silverio LM, et al. Endocytosis is required for synaptic activity-dependent release of amyloid-beta in vivo. Neuron 2008;58(1):42-51 View Article PubMed/NCBI
  100. Marsh J, Alifragis P. Synaptic dysfunction in Alzheimer’s disease: the effects of amyloid beta on synaptic vesicle dynamics as a novel target for therapeutic intervention. Neural Regen Res 2018;13(4):616-623 View Article PubMed/NCBI
  101. Dolev I, Fogel H, Milshtein H, Berdichevsky Y, Lipstein N, Brose N, et al. Spike bursts increase amyloid-β 40/42 ratio by inducing a presenilin-1 conformational change. Nat Neurosci 2013;16(5):587-595 View Article PubMed/NCBI
  102. Frere S, Slutsky I. Alzheimer’s Disease: From Firing Instability to Homeostasis Network Collapse. Neuron 2018;97(1):32-58 View Article PubMed/NCBI
  103. Takahashi RH, Nagao T, Gouras GK. Plaque formation and the intraneuronal accumulation of β-amyloid in Alzheimer’s disease. Pathol Int 2017;67(4):185-193 View Article PubMed/NCBI
  104. Eichenbaum H. The role of the hippocampus in navigation is memory. J Neurophysiol 2017;117(4):1785-1796 View Article PubMed/NCBI
  105. Lelos MJ, Good MA. c-Fos expression reveals aberrant neural network activity during cued fear conditioning in APPswe transgenic mice. Neurobiol Learn Mem 2012;98(1):1-11 View Article PubMed/NCBI
  106. Chao OY, de Souza Silva MA, Yang YM, Huston JP. The medial prefrontal cortex - hippocampus circuit that integrates information of object, place and time to construct episodic memory in rodents: Behavioral, anatomical and neurochemical properties. Neurosci Biobehav Rev 2020;113:373-407 View Article PubMed/NCBI
  107. Schell H, Boden C, Chagas AM, Kahle PJ. Impaired c-Fos and polo-like kinase 2 induction in the limbic system of fear-conditioned α-synuclein transgenic mice. PLoS One 2012;7(11):e50245 View Article PubMed/NCBI
  108. Tan Y, Liu Q, Li Z, Yang S, Cui L. Pyroptosis-triggered pathogenesis: New insights on antiphospholipid syndrome. Front Immunol 2023;14:1155222 View Article PubMed/NCBI
  109. Fu W, Ruangkittisakul A, MacTavish D, Shi JY, Ballanyi K, Jhamandas JH. Amyloid β (Aβ) peptide directly activates amylin-3 receptor subtype by triggering multiple intracellular signaling pathways. J Biol Chem 2012;287(22):18820-18830 View Article PubMed/NCBI
  110. Curson JEB, Liu L, Luo L, Muusse TW, Lucas RM, Gunther KS, et al. TLR4 phosphorylation at tyrosine 672 activates the ERK/c-FOS signaling module for LPS-induced cytokine responses in macrophages. Eur J Immunol 2023;53(7):e2250056 View Article PubMed/NCBI
  111. Tan J, Li W, Teng Z, Wang G, Li Y, Zhang Y. Senkyunolide H inhibits activation of microglia and attenuates lipopolysaccharide-mediated neuroinflammation and oxidative stress in BV2 microglia cells via regulating ERK and NF-κB pathway. Kaohsiung J Med Sci 2022;38(4):378-384 View Article PubMed/NCBI
  112. Fang C, Guo JW, Wang YJ, Li XQ, Zhang H, Cui J, et al. Diterbutyl phthalate attenuates osteoarthritis in ACLT mice via suppressing ERK/c-fos/NFATc1 pathway, and subsequently inhibiting subchondral osteoclast fusion. Acta Pharmacol Sin 2022;43(5):1299-1310 View Article PubMed/NCBI
  113. Wang C, Cui X, Dong Z, Liu Y, Xia P, Wang X, et al. Attenuated memory impairment and neuroinflammation in Alzheimer’s disease by aucubin via the inhibition of ERK-FOS axis. Int Immunopharmacol 2024;126:111312 View Article PubMed/NCBI
  114. Lara Aparicio SY, Laureani Fierro ÁJ, Aranda Abreu GE, Toledo Cárdenas R, García Hernández LI, Coria Ávila GA, et al. Current Opinion on the Use of c-Fos in Neuroscience. NeuroSci 2022;3(4):687-702 View Article PubMed/NCBI
  115. Morris SW, St Clair DM. Eliminating c-fos as a candidate gene for early-onset familial Alzheimer’s disease. Neurology 1994;44(9):1762-1764 View Article PubMed/NCBI
  116. Ijomone OM, Iroegbu JD, Aschner M, Bornhorst J. Impact of environmental toxicants on p38- and ERK-MAPK signaling pathways in the central nervous system. Neurotoxicology 2021;86:166-171 View Article PubMed/NCBI
  117. Zybura AS, Baucum AJ, Rush AM, Cummins TR, Hudmon A. CaMKII enhances voltage-gated sodium channel Nav1.6 activity and neuronal excitability. J Biol Chem 2020;295(33):11845-11865 View Article PubMed/NCBI
  118. Hudson AE. Genetic Reporters of Neuronal Activity: c-Fos and G-CaMP6. Methods Enzymol 2018;603:197-220 View Article PubMed/NCBI
  119. Yao WD, Wu CF. Distinct roles of CaMKII and PKA in regulation of firing patterns and K(+) currents in Drosophila neurons. J Neurophysiol 2001;85(4):1384-1394 View Article PubMed/NCBI
  120. Hayakawa K, Esposito E, Wang X, Terasaki Y, Liu Y, Xing C, et al. Transfer of mitochondria from astrocytes to neurons after stroke. Nature 2016;535(7613):551-555 View Article PubMed/NCBI
  121. Vámosi G, Baudendistel N, von der Lieth CW, Szalóki N, Mocsár G, Müller G, et al. Conformation of the c-Fos/c-Jun complex in vivo: a combined FRET, FCCS, and MD-modeling study. Biophys J 2008;94(7):2859-2868 View Article PubMed/NCBI
  122. Liu Y, Li X, Yu Y, Shi J, Liang Z, Run X, et al. Developmental regulation of protein O-GlcNAcylation, O-GlcNAc transferase, and O-GlcNAcase in mammalian brain. PLoS One 2012;7(8):e43724 View Article PubMed/NCBI
  123. Wang P, Lazarus BD, Forsythe ME, Love DC, Krause MW, Hanover JA. O-GlcNAc cycling mutants modulate proteotoxicity in Caenorhabditis elegans models of human neurodegenerative diseases. Proc Natl Acad Sci U S A 2012;109(43):17669-17674 View Article PubMed/NCBI
  124. Zhu Y, Shan X, Yuzwa SA, Vocadlo DJ. The emerging link between O-GlcNAc and Alzheimer disease. J Biol Chem 2014;289(50):34472-34481 View Article PubMed/NCBI
  125. Tai HC, Khidekel N, Ficarro SB, Peters EC, Hsieh-Wilson LC. Parallel identification of O-GlcNAc-modified proteins from cell lysates. J Am Chem Soc 2004;126(34):10500-10501 View Article PubMed/NCBI
  126. Amidfar M, de Oliveira J, Kucharska E, Budni J, Kim YK. The role of CREB and BDNF in neurobiology and treatment of Alzheimer’s disease. Life Sci 2020;257:118020 View Article PubMed/NCBI
  127. Nikseresht Z, Ahangar N, Badrikoohi M, Babaei P. Synergistic enhancing-memory effect of D-serine and RU360, a mitochondrial calcium uniporter blocker in rat model of Alzheimer’s disease. Behav Brain Res 2021;409:113307 View Article PubMed/NCBI
  128. You JC, Muralidharan K, Park JW, Petrof I, Pyfer MS, Corbett BF, et al. Epigenetic suppression of hippocampal calbindin-D28k by ΔFosB drives seizure-related cognitive deficits. Nat Med 2017;23(11):1377-1383 View Article PubMed/NCBI
  129. Badrikoohi M, Esmaeili-Bandboni A, Babaei P. Simultaneous administration of bromodomain and histone deacetylase I inhibitors alleviates cognition deficit in Alzheimer’s model of rats. Brain Res Bull 2022;179:49-56 View Article PubMed/NCBI
  130. Deisseroth K, Bito H, Tsien RW. Signaling from synapse to nucleus: postsynaptic CREB phosphorylation during multiple forms of hippocampal synaptic plasticity. Neuron 1996;16(1):89-101 View Article PubMed/NCBI
  131. Viosca J, Malleret G, Bourtchouladze R, Benito E, Vronskava S, Kandel ER, et al. Chronic enhancement of CREB activity in the hippocampus interferes with the retrieval of spatial information. Learn Mem 2009;16(3):198-209 View Article PubMed/NCBI
  132. Ramos BP, Birnbaum SG, Lindenmayer I, Newton SS, Duman RS, Arnsten AF. Dysregulation of protein kinase a signaling in the aged prefrontal cortex: new strategy for treating age-related cognitive decline. Neuron 2003;40(4):835-845 View Article PubMed/NCBI
  133. Lopez de Armentia M, Jancic D, Olivares R, Alarcon JM, Kandel ER, Barco A. cAMP response element-binding protein-mediated gene expression increases the intrinsic excitability of CA1 pyramidal neurons. J Neurosci 2007;27(50):13909-13918 View Article PubMed/NCBI
  134. Umeda T, Tomiyama T, Sakama N, Tanaka S, Lambert MP, Klein WL, et al. Intraneuronal amyloid β oligomers cause cell death via endoplasmic reticulum stress, endosomal/lysosomal leakage, and mitochondrial dysfunction in vivo. J Neurosci Res 2011;89(7):1031-1042 View Article PubMed/NCBI
  135. Lee SH, Kim KR, Ryu SY, Son S, Hong HS, Mook-Jung I, et al. Impaired short-term plasticity in mossy fiber synapses caused by mitochondrial dysfunction of dentate granule cells is the earliest synaptic deficit in a mouse model of Alzheimer’s disease. J Neurosci 2012;32(17):5953-5963 View Article PubMed/NCBI
  136. Lee JH, Cheon YH, Woo RS, Song DY, Moon C, Baik TK. Evidence of early involvement of apoptosis inducing factor-induced neuronal death in Alzheimer brain. Anat Cell Biol 2012;45(1):26-37 View Article PubMed/NCBI
  137. Chen X, Shen J, Wang Y, Chen X, Yu S, Shi H, et al. Up-regulation of c-Fos associated with neuronal apoptosis following intracerebral hemorrhage. Cell Mol Neurobiol 2015;35(3):363-376 View Article PubMed/NCBI
  138. Dosek A, Ohno H, Acs Z, Taylor AW, Radak Z. High altitude and oxidative stress. Respir Physiol Neurobiol 2007;158(2-3):128-131 View Article PubMed/NCBI
  139. Smeyne RJ, Vendrell M, Hayward M, Baker SJ, Miao GG, Schilling K, et al. Continuous c-fos expression precedes programmed cell death in vivo. Nature 1993;363(6425):166-169 View Article PubMed/NCBI
  140. Vaid A. Nilotinib as first-line therapy for chronic myeloid leukemia. Indian J Cancer 2011;48(4):438-445 View Article PubMed/NCBI
  141. Arendt T, Brückner MK, Morawski M, Jäger C, Gertz HJ. Early neurone loss in Alzheimer’s disease: cortical or subcortical?. Acta Neuropathol Commun 2015;3:10 View Article PubMed/NCBI
  142. Chi H, Chang HY, Sang TK. Neuronal Cell Death Mechanisms in Major Neurodegenerative Diseases. Int J Mol Sci 2018;19(10):3082 View Article PubMed/NCBI
  143. Goel P, Chakrabarti S, Goel K, Bhutani K, Chopra T, Bali S. Neuronal cell death mechanisms in Alzheimer’s disease: An insight. Front Mol Neurosci 2022;15:937133 View Article PubMed/NCBI
  144. Currais A, Fischer W, Maher P, Schubert D. Intraneuronal protein aggregation as a trigger for inflammation and neurodegeneration in the aging brain. FASEB J 2017;31(1):5-10 View Article PubMed/NCBI
  145. Seward ME, Swanson E, Norambuena A, Reimann A, Cochran JN, Li R, et al. Amyloid-β signals through tau to drive ectopic neuronal cell cycle re-entry in Alzheimer’s disease. J Cell Sci 2013;126(Pt 5):1278-1286 View Article PubMed/NCBI
  146. Butterfield DA, Halliwell B. Oxidative stress, dysfunctional glucose metabolism and Alzheimer disease. Nat Rev Neurosci 2019;20(3):148-160 View Article PubMed/NCBI
  147. Ma Q. Role of nrf2 in oxidative stress and toxicity. Annu Rev Pharmacol Toxicol 2013;53:401-426 View Article PubMed/NCBI
  148. Li N, Alam J, Venkatesan MI, Eiguren-Fernandez A, Schmitz D, Di Stefano E, et al. Nrf2 is a key transcription factor that regulates antioxidant defense in macrophages and epithelial cells: protecting against the proinflammatory and oxidizing effects of diesel exhaust chemicals. J Immunol 2004;173(5):3467-3481 View Article PubMed/NCBI
  149. Novoa C, Salazar P, Cisternas P, Gherardelli C, Vera-Salazar R, Zolezzi JM, et al. Inflammation context in Alzheimer’s disease, a relationship intricate to define. Biol Res 2022;55(1):39 View Article PubMed/NCBI
  150. Zhou L, Xue C, Chen Z, Jiang W, He S, Zhang X. c-Fos is a mechanosensor that regulates inflammatory responses and lung barrier dysfunction during ventilator-induced acute lung injury. BMC Pulm Med 2022;22(1):9 View Article PubMed/NCBI
  151. Kinney JW, Bemiller SM, Murtishaw AS, Leisgang AM, Salazar AM, Lamb BT. Inflammation as a central mechanism in Alzheimer’s disease. Alzheimers Dement (N Y) 2018;4:575-590 View Article PubMed/NCBI

About this Article

Cite this article
Babaei P, Faraji N, Eyvani K. c-Fos Expression Differentially Acts in the Healthy Brain Compared with Alzheimer’s Disease. Gene Expr. Published online: Apr 28, 2025. doi: 10.14218/GE.2024.00080.
Copy Export to RIS Export to EndNote
Article History
Received Revised Accepted Published
December 31, 2024 March 22, 2025 March 26, 2025 April 28, 2025
DOI http://dx.doi.org/10.14218/GE.2024.00080