The Pleiotropic Face of CREB Family Transcription Factors
Md. Arifur Rahman Chowdhury, Jungeun An
Abstract
cAMP responsive element-binding protein (CREB) is one of the most intensively studied phosphorylation-dependent transcription factors that provide evolutionarily conserved mechanisms of differential gene expression in vertebrates and invertebrates. Many cellular protein kinases that function downstream of distinct cell surface receptors are responsible for the activation of CREB. Upon functional dimerization of the activated CREB to cis-acting cAMP responsive elements within the promoters of target genes, it facilitates signal-dependent gene expression. From the discovery of CREB, which is ubiquitously expressed, it has been proven to be involved in a variety of cellular processes that include cell proliferation, adaptation, survival, differentiation, and physiology, through the control of target gene expression. In this review, we highlight the essential roles of CREB proteins in the nervous system, the immune system, cancer development, hepatic physiology, and cardiovascular function and further discuss a wide range of CREB-associated diseases and molecular mechanisms underlying the pathogenesis of these diseases.
INTRODUCTION
The 43-kDa ubiquitous protein CREB belongs to the CREB/activating transcription factor (ATF) family, and has a conserved basic region/leucine zipper (bZIP) domain (Hai and Hartman, 2001; Sassone-Corsi, 1995). For transcriptional initiation, activation of CREB is primarily modulated by phosphorylation at the kinase-inducible domain (KID) in response to a variety of extracellular stimuli and mitogen stress signals (Fig. 1) (Montminy et al., 2004). Secondly, the active dimer of the CREB/ATF family binds to the conserved cis-acting cAMP responsive elements (CREs) 5′-TGACGT(C/G)A-3′ or 5′-CGTCA-3′ (Hai and Hartman, 2001; Impey et al., 2004), while the Drosophila CREB, dCREBB binds to the consensus sequence 5′-GTGACGT(A/C)(A/G)-3′ that is usually present in viral and cellular promoters, or within the promoters of target genes (Mayr and Montminy, 2001). Lastly, the CREB association with CREs promotes the recruitment of transcriptional machinery, such as CREB-binding protein (CBP), p300, and CREB-regulated transcription coactivators (CRTCs), to regulate transcription (Montminy et al., 1990).

CRTCs contain RNA-binding motifs, enhance transcriptional activity, also promote alternative splicing (pre-mRNA splicing) (Iourgenko et al., 2003). In particular, CRTCs are considered the key coactivators of CREB-regulated gluconeogenesis, lipid metabolism, synaptic plasticity, and mitochondrial biogenesis. Upon dephosphorylation, this signal-dependent co-activator translocate into the nucleus to interact with CREB and modulates transcription (Wang et al., 2021). As of today, CRTC1, one of the three identified CRTCs, which is abundant in the brain, controls neuronal plasticity and overall memory formation (Parra-Damas et al., 2017). CRTC1 has also been shown to modulate energy balance (Altarejos et al., 2008). In addition, CRTC2 in the liver activates the gluconeogenic program (Han et al., 2017), whereas CRTC3 correlates to weight gain as it controls lipid metabolism and energy balance (Conkright et al., 2003).
Many pioneering experimental studies have demonstrated the link between cAMP, protein kinase A (PKA), the bZIP domain, and CREB (Ferraris et al., 2002). Earl Sutherland (Nobel laureate, 1971) was the first to discover the mechanism of action of the hormone epinephrine, and demonstrate that the activation of phosphorylase by epinephrine was triggered by cAMP, which is known as the second messenger. Functionally, the hormone-stimulated production of cAMP in the cell was actually triggered by an enzyme called adenylyl cyclase (Rall et al., 1956; Sutherland, 1972). In 1987, influential findings in PC12 cells demonstrated that CREB functions as a nuclear protein that binds to the CRE found in the somatostatin gene promoter (Quinn and Granner, 1990). So far CREB has been reported to be phosphorylated by up to 300 different types of stimuli. However, the pioneering studies on reversible phosphorylation (switching between different cellular processes to activate or regulate them) by Edmond Henri Fischer and Ed Krebs (Nobel laureate, 1992) identified PKA as being a primary element associated with cAMP action.
A pleiotropic signaling molecule CREB serves as a transcription factor, a neuron-associated molecule, a metabolic factor, and a factor involved in cell cycle and proliferation (Wen et al., 2010). A result of CREB activation is the transcription initiation of several distinct genes, including c-Fos, BDNF, several neuropeptide genes, as well as genes related to the mammalian circadian clock (PER1 and PER2) (Purves et al., 2008). Cellular processes associated with CREB engagement include cell cycle progression, body’s defense mechanism, and DNA repair (Sands and Palmer, 2008). Several other enzyme modifications are also carried out by CREB, such as glycosylation, ubiquitination, phosphorylation, and SUMOylation, contributing to the maintenance of physiological homeostasis (Lamarre-Vincent and Hsieh-Wilson, 2003). In fact, CREB is now recognized as one of the leading factors in neural development, synaptic plasticity in synapses, survival of neurons, neurotransmission, and overall memory (Belgacem and Borodinsky, 2017). Therefore, CREB signaling dysregulation is involved in a range of neuropathological and neurodegenerative diseases, including schizophrenia, Alzheimer’s disease (AD), Huntington’s disease (HD), and Parkinson’s disease (PD) (Amidfar et al., 2020). Contemporary studies also indicated that CREB plays a key role in integrating various physiological functions in non-neuronal cells. For example, CREB regulates glucose metabolism, lipid metabolism, energy balance, and fatty acid oxidation in hepatic cells, and also cardiac contraction, cardiac remodeling, and angiogenesis in cardiovascular cells (Yin et al., 2021). In addition, CREB reduces the inflammation-induced injury by suppressing oxidative stress in kidney cells, whereas it is well recognized for its regulation over pro-inflammatory and anti-inflammatory responses in immune cells (Portilla et al., 2002; Tran et al., 2011). Thus CREB are also involved in non-neuronal diseases like cancer, metabolic disorder (type 2 diabetes mellitus [T2DM]), hepatic disorder and lung fibrosis, and cerebral ischemic injury (Huang et al., 2015). Throughout our review, we first address the functional role of CREB in the nervous system, as well as non-neuronal tissues. In addition, we thoroughly discuss CREB activation and the evidence for and against it in different disease progression or remission scenarios.
CREB FUNCTIONS IN THE NERVOUS SYSTEM
Neuronal function
A growing body of evidence has demonstrated that CREB regulates neuroprotection by upregulating neurotrophins and anti-apoptotic genes, and detoxifying reactive oxygen species in neurons. CREB modulates immediate–early genes, for instance c-Fos, leptin, IAPs, BDNF and nNOS, insulin-like growth factor 1 (IGF-1), pituitary adenylate cyclase-activating polypeptide, and B cell lymphoma-2 (Bcl-2). They have all found to have an impact on neuronal viability and neural development (Tabuchi et al., 2002). BDNF is an established target gene for CREB that contributes profoundly to the development of neuronal circuits and the activity-dependent survival of existing neurons. BDNF contributes to sympathetic neuronal survival via CREB-induced expression of the anti-apoptotic Bcl-2 (Riccio et al., 1999). Another study demonstrates that CREB-regulated anti-apoptotic Bcl-2 family member, myeloid cell leukemia sequence 1, is required for cortical neurogenesis and neuronal survival after DNA damage (Arbour et al., 2008). Nerve growth factor binding to its cognate receptor has been shown to result in the activation of RSK2 kinase, thereby phosphorylating CREB at Ser-133, which is required for the Bcl-2 expression (Xing et al., 1996). BDNF knockout animal models showed either progressive atrophy of neurons or the degeneration of specific neurons, leading to the severe impairment of synaptic transmission and long-term potentiation (Bianchi et al., 1996). Several studies confirm that low BDNF levels are the probable cause for neuronal dysfunction in AD, as disease progression induces symptoms of synapse loss and cognitive decline (Amidfar et al., 2020). In experimental studies, increasing BDNF levels can reduce the risk of memory deficits and cognitive dysfunction in AD patients, as well as reduce the amount of amyloid beta peptide (Aβ) aggregation in the brain (Hampel et al., 2021). Clinical studies show that the overexpression of BDNF and its receptor tyrosine kinase-coupled receptor (TrkB) in the hippocampus can reverse memory impairment. Several studies also suggest that both BDNF and TrkB are target genes of CREB that may contribute to memory formation and neuroprotection (Amidfar et al., 2020). Overall, the evidence suggests that CREB improves memory and acts as a neuroprotective modulator through a variety of underlying mechanisms.
Neural cell adhesion molecules (NCAMs) have been shown to promote neurite outgrowth through at least two mechanisms: (1) triggering the activation of FGFR, and (2) interacting with Fyn and focal adhesion kinase (FAK) to create intracellular signaling complexes (Fig. 2) (Ditlevsen et al., 2008). The intracellular interaction partners Fyn and FAK phosphorylate NCAM and transiently activate CREB and some of CREB upstream regulators, including ERK1/2, when they interact with NCAM (Fig. 2). It has been shown that CREB promotes the activation of target genes involved in axonal outgrowth, survival, and synaptic transmission in neuronal cells. In a genome-wide screen, miR132 was identified as a neuronal CREB target whose expression is highly induced by neurotrophins, leading to neurite outgrowth (Vo et al., 2005).

This claim was also supported by experiments involving avoidance conditioning and spatial escape learning in rodents. Numerous in vivo and in vitro genetic manipulation research indicated that CREB is essential for neuronal survival and plasticity (Josselyn and Nguyen, 2005). In the mice model, hypomorphic CREB mice exhibited deficits in both spatial memories, and hippocampal long-term potentiation (LTP) (Bianchi et al., 1996). Meanwhile, targeted deletion or targeted disruption of the α and δ isoforms of CREB in mice is profoundly deficient in LTM (Blendy et al., 1996). Even an experiment on the disruption of CREB in the dorsal hippocampus demonstrated impairment in water maze performance (Pittenger et al., 2002). In CREB null mice, there are aberrant axonal projections in the anterior commissure and corpus callosum, and more interestingly, homozygous null mutation in all CREB isoforms resulted in perinatal death (Rudolph et al., 1998). An interesting characteristic of CREB is that it exhibits LTM formation through interacting with its coactivators, such as CBP and cAMP responsive element modulator (CREM), which regulate the expression of genes that are essential for memory consolidation and neuronal survival (Cameron and Glover, 2015; Sekeres et al., 2010). During neural development, mice lacking both CREM and CREB1 in brain exhibit significant neuronal apoptosis (Mantamadiotis et al., 2002). Additionally, in vitro expression of the CREB family members’ dominant-negative form impairs the dendritic development of cortical neurons (Pignataro et al., 2015). Numerous studies indicate that other members of the CREB family are also involved in embryonic development, neuroplasticity, and spatial memory. In experiments on deficient mice for both CREB and ATF1 (ATF1−/−CREB1−/− or ATF1+/−CREB1−/−), the mutant embryos experience developmental arrest prior to implantation and exhibit a fatal phenotype by embryonic day 9.5 as a result of widespread apoptosis (Bleckmann et al., 2002). The overall findings suggest that CREB family-mediated signals are also crucial for maintaining cell viability in initial embryonic development. Nevertheless, the necessity of CREB1 in hippocampal plasticity is not absolute, but beyond dispute, and several studies have suggested the loss of CREB1 could be compensated by other molecular components (Kogan et al., 1997).
Neurodegenerative diseases
The evidence clearly suggests that CREB is a neuroprotectant. Since CREB-mediated neuroprotection requires the expression of CREB target genes, like the pro-survival gene Bcl-2, CREB dysfunction is associated with multiple neuronal diseases (Meller et al., 2005). In particular, the CREB pathway participates in the pathogenesis of several neurodegenerative diseases, such as HD, RTS, AD, and CLS (Table 1) (Hallam and Bourtchouladze, 2006; Harum et al., 2001; Saura and Valero, 2011). In addition to neurodegenerative diseases and polygenic disorders, CREB is considered one of many genes that contribute to improper mood regulation, or other polygenic diseases. Therefore, many CREB-regulated genes are implicated in the pathogenesis of mood disorders, depression, and psychomotor retardation (Dragunow, 2004).
HD is the most prevalent hereditary neurodegenerative condition, an autosomal dominant disease characterized by irreversible motor impairments, cognitive decline, and psychiatric difficulties, which progresses to dementia and mortality 15-20 years after onset (Eggert et al., 2022). In HD, mutations resulting in expanded CAG repeats are responsible for long segments of polyglutamine (polyQ) in the HD protein Huntingtin (Htt) (IT15 gene encodes the Htt; normal repeats range [6 to 35]; repeats of 36 or more are synonymous with HD) (Kay et al., 2016). Mutant Htt protein and expanded polyQ tracts form nuclear aggregates that contain CREB-binding protein CBP (Table 1) (McCampbell et al., 2000). However, the CBP correlation with HD or Htt toxicity has been controversial. Surprisingly, experimental evidence demonstrates that transfection with mutant Htt induced endogenous CBP to be depleted from Neuro2a cells’ nucleus (Jiang et al., 2003). Also, it has been recommended that mutant Htt expression may exert an impact on histone acetyltransferases (HATs) like CBP to decrease histone acetylation levels, which is associated with polyQ toxicity (Steffan et al., 2001). Numerous studies have demonstrated that, in addition to CBP, altered CREB function also plays a role in the HD pathogenesis (Landles and Bates, 2004). In mice lacking both CREB1 and CREM in the postnatal forebrain HD-like progressive neurodegeneration was observed in the dorsolateral striatum and hippocampal region (Mantamadiotis et al., 2002). Consistent with this, some of CRE-regulated genes have been downregulated in the HD patients. Furthermore, transgenic A-CREB, which expresses a dominant negative form of CREB, showed increased 3-nitropropionic acid-induced striatal lesion size and motor impairment, and also robustly deteriorated motor dysfunction observed in genetic mouse model of HD. In summary, these observations demonstrate that loss of CREB activity and the resulting disruption of CREB-mediated gene expression contributes to the pathogenesis of HD (Landles and Bates, 2004).
RTS is a rare congenital condition that is marked by mental and physical impairment, mood instability, behavioral stereotypes, and abnormalities of the thumbs, big toes, and face (Hallam and Bourtchouladze, 2006). Many chromosomal breakpoints observed in patients with RTS have been shown to be associated with the CBP locus (Table 1) (Petrij et al., 1995). Biochemical and genetic studies using RTS-associated HAT domain mutants uncovered that these mutations led to loss of in vitro acetyltransferase activity and also reduced CREB-mediated transcription (Kalkhoven et al., 2003).
Accumulation of Aβ and mutations in two presenilin (PS) genes are considered as the familial cause of AD pathogenesis (Jeong, 2017). Preceding genetic investigations have demonstrated that PSs play indispensable roles in synaptic function, memory, and neuronal survival (Zhang et al., 2009). Amyloid beta 42 (Aβ42) peptides, which are produced through sequential proteolysis of the APP (amyloid precursor protein) by β-secretase and γ-secretase, and are major insoluble components of neuritic plaques found in patients of AD, lowered the PKA activity, as well as the levels of phosphorylated CREB (Table 1) (Vitolo et al., 2002). Even the most hazardous Aβ species, Aβ oligomers, also block CREB phosphorylation in primary neurons through a mechanism necessitating NMDA receptor degradation (Ma et al., 2007). It is evident that the expression of CREB-target genes and CBP is reduced in the absence of wild-type PSs, which implies a direct pathophysiological correlation of AD. Calcium influx and CREB phosphorylation mediate the effect of neuronal activity on PS1 expression, whereas the CREB with a mutated Ser-133 residue (S133A) lowers expression levels of PS1 (Mitsuda et al., 2001). CREB also regulates the transcription of PEN-2, that is a critical component of the γ-secretase complex (Wang et al., 2006). In conclusion, these findings show that PS/γ-secretase and CREB signaling may be regulated in a reciprocal manner during activity-dependent synaptic function.
Postmortem examinations have shown that the cortices of CLS patients contain reduced concentrations of CREB, compared with patients treated with anti-depressants. A growing number of studies has shown that decreased levels of total or phosphorylated CREB are detected in the hippocampus of elderly mice or rats (Kudo et al., 2005). Interestingly, overexpression of the CREB gene in the dentate gyrus (all sensory modalities merge and play a critical role in learning and memory) exhibits anti-depressive behavior that is quite similar to that observed when anti-depressant drugs are used (Blendy, 2006). More recent researches have revealed that CREB activity modulates the behavioral phenotypes of mice in response to emotional stimuli (Barrot et al., 2002). In addition, prolonged anti-depressant treatment increases CREB expression in the hippocampus, indicating the role of CREB in the pathogenic process and therapy of depression (Gass and Riva, 2007). CREB activity is associated with upregulation of neural circuit excitability that improves motor performance after stroke. Therefore, motor recovery following a stroke is improved by raising CREB levels, whereas stroke recovery is inhibited by limiting CREB signaling (Caracciolo et al., 2018).
CREB FUNCTIONS IN THE IMMUNE SYSTEM
Pro-inflammatory signaling
Bacterial lipopolysaccharide (LPS) can initiate pro-inflammatory signaling through the activation of TLRs (Fig. 3, left). Subsequently, the activation of the NF-κB family members via the adaptor molecules, such as MyD88, IRAK4, and TRAF6, are required for the production of pro-inflammatory cytokines. The nuclear factor-κB (NF-κB) family transcription factors serve as evolutionarily conserved regulators of the innate immune responses. Five members of the NF-κB family are NF-κB1, NF-κB2, RelA, RelB, and c-Rel, and have a conserved Rel homology domain (RHD) in the amino-terminal region (Ghosh et al., 1998). Functionally, they combine to create homo- and heterodimeric complexes that are transcriptionally active, whereas the RHD contains sequences necessary for dimerization, DNA binding, interacting with IκBs, and nuclear translocation. The formation of the active RelA/p50 complex requires proteasomal degradation of phosphorylated IκB that is also induced by LPS (Fig. 3). The CREB coactivator CBP needs to directly interact with the RelA Ser-276 for optimal RelA/p50 activity, whereas acetylation of CBP further increases RelA activity. Interestingly, since RelA competes with phospho-CREB for CBP, the NF-κB activity can be inhibited by increased CREB activation or enhanced by overexpression of CBP (Fig. 3) (Ollivier et al., 1996).
Anti-inflammatory signaling
In macrophages, several TLR signals (TLRs 2, 3, 4, 7, and 9) induce the expression of IL-10, which is a potent anti-inflammatory cytokine that limits overactivation of inflammatory signaling, and minimize undesirable tissue damage (Saraiva and O’Garra, 2010). Upon TLR ligation by LPS, NF-κB/MAPKs-dependent signaling cascades produce anti-inflammatory cytokine IL-10 together with pro-inflammatory cytokines, such as TNF-α, IL-1, and IL-6. MSK1 and MSK2 are required for the phosphorylation of CREB and AP-1, their binding to the promotor of IL-10, and transcriptional activation (Fig. 3) (Saraiva and O’Garra, 2010). Interferon-γ (IFN-γ), which is predominantly generated by natural killer cells and T cells, suppresses the production of IL-10 through two different mechanisms. First, IFN-γ inhibits the TLR-mediated activation of PI3K-AKT and the resulting derepression of GSK3β interferes with CREB activation. Second, IFN-γ also directly downregulates MAPK pathway (Fig. 3) (Hu et al., 2006).
CREB FUNCTIONS IN CANCER DEVELOPMENT
The CREB transcription factor drives expression of numerous target genes that are involved in proliferation, self-renewal, differentiation, and apoptosis (Steven et al., 2020). However, there is no concrete proof that mutated CREB protein is directly associated with cancer development. Rather, the mutation of diverse upstream regulators appear to mediate the constitutive activation of CREB target genes, such as early growth response protein 1, cyclins A1 & D1, and Bcl-2, contributing to tumorigenesis (Sakamoto and Frank, 2009).
The discovery of a chromosomal t (12; 22) (q13; q12) translocation that resulted in the production of a fusion protein EWS-ATF1 in soft tissue clear cell sarcomas provided the first evidence of CREB-associated cancer (Schaefer et al., 2004). As of now, CREB is linked to a wide range of cancer types, including hematopoietic and solid tumors, acute myeloid leukemia, prostate and lung cancers, as well as gastric, melanoma, pancreatic, and breast carcinomas, since CREB has been shown to act either as a direct mediator or as a proto-oncogene (Table 2) (Sakamoto and Frank, 2009; Shankar et al., 2005). In most of the case hyperphosphorylated and overexpressed CREB are identified in both nonhematologic and hematological cancers. In fact, elevated CREB expression and activation are linked to cancer initiation and progression, increased chemo-resistance (cisplatin resistance), and lower survival rate of cancer patients (Steven et al., 2020; Zhang et al., 2017). In the most common subtype of lung cancer, CREB was overexpressed, and inhibited ferroptosis to enhance the rapid growth of cancer (Xiao et al., 2010).
Consistent with the fact that elevated CREB expression correlates with tumorigenesis, a large number of previous studies have shown that downregulation of CREB is closely related to suppression of tumorigenesis in many different cells. The knockdown of CREB downregulates anti-apoptotic Bcl-2 and IAP family members, such as Bcl-2, Bcl-xL, Mcl-1, XIAP and survivin, validating the connection between CREB and these prosurvival oncogenes (Xiang et al., 2006). Ro-31-8220 (a synthetic S6 kinases inhibitor) mediated CREB inactivation arrests the cell cycle at the G2-M phase, and also mediates the inhibitory action to anti-apoptotic factors Bcl-2 and Bcl-xL, signifying that CREB could be a potential therapeutic target in non-small cell lung carcinoma (NSCLC) (Xiang et al., 2006). Interestingly, the treatment of lung adenocarcinoma cells with AMPA (α-amino-3-hydroxy-5-methyl-4-isoxazolepropionate) antagonists reduced phosphorylation of CREB, suppressed the expression of cyclin D1, upregulated the tumor suppressor proteins p21 and p53, and decreased the number of metastatic cells (Stepulak et al., 2007). Furthermore, a dominant-negative form of CREB and siRNA-mediated knockdown of CREB suppressed the proliferation and induced apoptosis of NSCLC (Xiao et al., 2010). In a same manner, decoy oligonucleotides and RNA interference both suppress CREB-mediated gene transcription thereby negatively impacting tumor growth, increased apoptosis, become sensitive to antiproliferative signals and inhibition of anchorage-independent proliferation (Sakamoto and Frank, 2009; Xie et al., 2015). The metastatic potential of tumor cells is also inhibited by the overexpression of dominant negative form of CREB (KCREB), which loses its ability to bind to CRE elements, but can form a nonfunctional heterodimer with wild-type CREB (Linnerth-Petrik et al., 2012).
Apart from CREB, there are a number of regulatory loops involved in migration, invasion, and metastasis formation, including CBP, CREM, and CRTCs (Iourgenko et al., 2003). In mutagen analysis of CRTCs, the expression of dominant-negative mutant specifically inhibits the oncogenic transcriptional program of CREB (Ostojić et al., 2021). Other than the functional component, processes like dimerization, CRE-dependent regulation of CREB target gene expression, posttranslational modifications (PTMs), circumstantial excessive phosphorylation, and the ratio of repressor or ICER are also involved in cancer progression and overall pathogenesis (Voropaev et al., 2019). The qualitative control of CREB, through different combinations of dimerization and PTMs, such as phosphorylation, ubiquitination, methylation, glycosylation, and SUMOylation, seems more important than the quantitative regulation of CREB expression levels during tumorigenesis. Number of experimental models indicate that PTM affects the overall stability and activation. For example, multiple site phosphorylation of CREB at Ser129 and Ser133 enhances transcription activity, whereas the phosphorylation of Ser111 and Ser121 totally abolishes the CREB-dependent gene expression (Sapio et al., 2020). Furthermore, previous studies demonstrate that hyperphosphorylation of CREB is correlated with its ubiquitination and increased proteasomal degradation (Steven et al., 2020).
CREB activity can be also regulated by several miRNAs at the transcriptional level. In certain conditions, miRNAs may function as either tumor suppressors or oncogenes. In a contextual manner, CREB either regulates miRNAs or miRNAs regulate CREB expression in different types of cancer proliferative signaling (Pigazzi et al., 2009). A recent investigation in acute myeloid leukemia revealed that the 3′-UTR of CREB contains a miR-34b regulatory element, providing a negative feedback regulation of CREB activity (Pigazzi et al., 2009). Extensive studies demonstrate that miR-200b and miR-203 have been shown to target CREB, suggesting their tumor-suppressing mechanism (Noguchi et al., 2016). Consistent with this, low miR-200b expression coupled with high levels of CREB expression can serve as a significant factor of prognosis in astrocytoma (Zhang et al., 2014).
Unexpectedly, high CREB expression also possess benefit in some cancer types, such as clear cell renal cell carcinoma, breast cancer, and esophageal squamous cell carcinoma. For example, overexpression of CREB in breast cancer (HER-2/neu-positive or basal-like or luminal-type A) collectively improves the survival of patient as well as recurrence-free survival (Steven et al., 2020). In order to get deeper knowledge of the fundamental processes of CREB regulation and function, it is essential to further examine CREB as "friend or foe" due to its dual and opposing roles that confound various cancer entities. Nevertheless, it seems clear that CREB is regarded as a promising biomarker and an ideal therapeutic target gene for a wide range of cancers due to its essential role in the development, maintenance, and proliferation of many different types of cancer (Table 2) (Sakamoto and Frank, 2009).
CREB FUNCTIONS IN HEPATIC PHYSIOLOGY AND DISEASES
CREB plays a vital role in liver to respond to various metabolic demands responsible for normal physiological functions of major body organs (Table 3) (Wang et al., 2015a). Glucose homeostasis is controlled by two antagonistic hormones glucagon and insulin. During fasting time glucagon enhances the CREB transcriptional activity, resulting in the expression of gluconeogenic genes, such as pyruvate carboxylase (PC), glucose-6-phosphatase (G6Pase), and phosphoenolpyruvate carboxykinase 1 (PEPCK1), and an increase in overall glucose output (Oh et al., 2022; Zhang et al., 2016). However, feeding conditions enhance the secretion of insulin and sequentially activates AKT and SIK2. Both kinases phosphorylate CBP/P300 and CRTC2 in an inhibitory manner for forming active complex with CREB, leading to inhibition of gluconeogenic program and decreased glucose output (Oh et al., 2013).
Communally, CREB in the liver participates in the control of lipogenesis and lipolysis, in addition to glucose homeostasis, specifically gluconeogenesis, through PEPCK and G6Pase (Table 3) (Han et al., 2016; Rowe and Arany, 2014). Studies have demonstrated that peroxisome proliferator-activated receptor-γ (PPARγ) coactivator-1α (PGC-1α), which is activated by CREB activation, not only upregulates gluconeogenesis, but also enhances lipolysis via the oxidation of long-chain fatty acids (Huang et al., 2017; Lin et al., 2005). Since a nuclear hormone receptor PPAR-γ is one of key lipogenic mediators, CREB negatively regulates the lipid synthesis during fasting conditions through inhibition of PPAR-γ expression (Herzig et al., 2003).
It has been shown that dysregulation of the CREB-dependent gluconeogenic gene pathway can result in metabolic diseases (Han et al., 2020). In pathological conditions like obesity and T2DM, insulin fails to regulate hepatic metabolism, resulting in excess glucose and fat production, as well as hepatic insulin resistance (Petersen et al., 2017). Remarkably, genetic ablation or downregulation of the CREB gene resulted in profoundly fasting hypoglycemia and also reduced mRNA expression of gluconeogenic genes such as PC, G6Pase, and PEPCK (Herzig et al., 2001).
Studies have demonstrated that PGC-1α, which is associated with the pathogenesis of T2DM, induced expression of a number of transcription factors that drive the expression of key gluconeogenic genes (Liang and Ward, 2006).
There are several forms of liver disorders that can be caused by infections, hereditary abnormalities, obesity, and alcohol abuse (Li et al., 2019). The most common chronic liver illness, nonalcoholic fatty liver disease (NAFLD), which includes simple hepatic steatosis, nonalcoholic steatohepatitis, liver fibrosis, and liver cirrhosis, is characterized by abnormal accumulation of lipids, involving hepatic injury and inflammation (Awaad et al., 2020).
Additionally, a fatty liver symptom combined with increased expression of PPARγ, a key regulator of lipogenesis, were observed in CREB-deficient animals (Herzig et al., 2003). Based on the fact that during fasting CREB activation upregulates gluconeogenesis through the induction of PGC-1α, but suppresses lipogenesis by inhibition of PPARγ, the discovery and development of selective CREB antagonists may provide an effective treatment for diabetic patients by improving glucose control and/or insulin sensitivity (Herzig et al., 2001).
According to the proposed model, NAFLD is caused by lipid peroxidation-mediated liver injury owing to a “two-hit” pathogenesis. Early in the disease, the first hit involves excessive accumulation of hepatic triglyceride along with insulin resistance, while the second hit includes pro-inflammatory cytokines, mitochondrial dysfunction, and oxidative stress, leading to hepatic fibrosis and cirrhosis (Fang et al., 2018). When high fat diet was given to rats to drive the development of NAFLD, higher levels of both cAMP and CREB in the liver tissue were significantly detected in these NAFLD rats, compared to control animals (Awaad et al., 2020).
Additionally, CREB controls the expression of the 3-hydroxy-3-methylglutaryl coenzyme A (HMG−CoA) synthase gene which contains a consensus CRE and two binding sites for sterol regulatory element-binding proteins (SREBPs) in its promotor (Dooley et al., 1998). The HMG-CoA synthase, which converts acetoacetyl-CoA and acetyl-CoA into HMG-CoA, is the rate-limiting enzyme for cholesterol synthesis. The SREBPs collaborate with CREB to drive gene expression of HMG-CoA synthase, in response to low levels of cellular cholesterol (Dooley et al., 1999).
Liver fibrosis is caused by the extensive accumulation of extracellular matrix, and can lead to liver cirrhosis, portal hypertension, and even multi-organ dysfunction (Liu et al., 2017). Numerous researches conducted over the past several decades has advanced our understanding of the close correlation between hepatic fibrosis and CREB-dependent gene expression (Li et al., 2019). Investigations demonstrate that the upregulated activation of CREB1 antagonizes the development of liver fibrosis through the downregulation of transforming growth factor-β1 (TGF-β1) signaling pathway, which is thought to serve as a key fibrogenic driver (Li et al., 2019). TGF-β1-induced phospho-Smad2 and phosphor-ERK1/2 expression was significantly suppressed by CREB1 overexpression (Deng et al., 2016). As a consequence, acetylation and/or extended phosphorylation of CREB-1 inhibit TGF-β1-mediated fibrogenesis in hepatic stellate cells via Smad2-dependent and independent pathways (Deng et al., 2016).
ROLES OF CREB IN CARDIOVASCULAR FUNCTION AND REMODELING
Numerous findings show that CREB is involved in both the positive and negative aspects of cardiovascular remodeling (Table 3). Other transcription factors that are activated at the same time as CREB by particular extracellular stimuli may influence whether CREB is beneficial or harmful to cardiovascular remodeling. Several important functions of CREB in the cardiovascular system have been identified. First, CREB activity is required for normal contractile response to extracellular stimuli, gene expression of voltage-gated K+ channel Kv4.3 in the heart, and IGF-1-mediated suppression of apoptosis in cardiac myocytes (Schulte et al., 2012). Second, CREB in the endothelial cells appears to mediate the expression of angiogenesis-related genes and Cox-2, a key inflammatory response gene, suggesting an essential function for CREB in vascular remodeling (Scoditti et al., 2010). Lastly, angiotensin II- and thrombin-induced hypertrophy of vascular smooth muscle cells (VSMC) requires the CREB activity, emphasizing the importance of CREB function in VSMC proliferation/survival (Truong et al., 2021).
CONCLUSION
It is clear that CREB acts as a phosphorylation-dependent transcription factor that is associated with a wide range of cellular processes that include cell proliferation, survival, differentiation, and physiology. These pleiotropic effects of CREB are mediated by distinct target gene expression in response to diverse physiological stimuli. A lot of evidence supports that several regulatory modes of CREB family transcription factors contribute to selective target gene expression, and thus the pleiotropic roles of CREB proteins in neuronal and non-neuronal cells. Despite more than three decades of intensive research, there are still a few crucial unanswered questions with regard to the molecular mechanisms underlying the activation of transcription by CREB. Therefore, better understanding of CREB-mediated transcriptional regulation should be essential for the development of therapeutics for CREB-related diseases.
Article information
Articles from Mol. Cells are provided here courtesy of Mol. Cells
References
- Abel, T., Bhatt, R., Maniatis, T. (1992). A Drosophila CREB/ATF transcriptional activator binds to both fat body- and liver-specific regulatory elements. Genes Dev.. 6, 466-480.
- Altarejos, J.Y., Goebel, N., Conkright, M.D., Inoue, H., Xie, J., Arias, C.M., Sawchenko, P.E., Montminy, M. (2008). The Creb1 coactivator Crtc1 is required for energy balance and fertility. Nat. Med.. 14, 1112-1117.
- Amidfar, M., de Oliveira, J., Kucharska, E., Budni, J., Kim, Y.K. (2020). The role of CREB and BDNF in neurobiology and treatment of Alzheimer's disease. Life Sci.. 257, 118020.
- Arany, I., Megyesi, J.K., Reusch, J.E.B., Safirstein, R.L. (2005). CREB mediates ERK-induced survival of mouse renal tubular cells after oxidant stress. Kidney Int.. 68, 1573-1582.
- Arany, Z., Novikov, M., Chin, S., Ma, Y., Rosenzweig, A., Spiegelman, B.M. (2006). Transverse aortic constriction leads to accelerated heart failure in mice lacking PPAR-γ coactivator 1α. Proc. Natl. Acad. Sci. U. S. A.. 103, 10086-10091.
- Arbour, N., Vanderluit, J.L., Le Grand, J.N., Jahani-Asl, A., Ruzhynsky, V.A., Cheung, E.C.C., Kelly, M.A., MacKenzie, A.E., Park, D.S., Opferman, J.T. (2008). Mcl-1 is a key regulator of apoptosis during CNS development and after DNA damage. J. Neurosci.. 28, 6068-6078.
- Asakawa, M., Takano, H., Nagai, T., Uozumi, H., Hasegawa, H., Kubota, N., Saito, T., Masuda, Y., Kadowaki, T., Komuro, I. (2002). Peroxisome proliferator-activated receptor γ plays a critical role in inhibition of cardiac hypertrophy in vitro and in vivo. Circulation. 105, 1240-1246.
- Awaad, A.K., Kamel, M.A., Mohamed, M.M., Helmy, M.H., Youssef, M.I., Zaki, E.I., Essawy, M.M., Hegazy, M.G.A. (2020). The role of hepatic transcription factor cAMP response element-binding protein (CREB) during the development of experimental nonalcoholic fatty liver: a biochemical and histomorphometric study. Egypt. Liver J.. 10, 36.
- Barrot, M., Olivier, J.D.A., Perrotti, L.I., DiLeone, R.J., Berton, O., Eisch, A.J., Impey, S., Storm, D.R., Neve, R.L., Yin, J.C. (2002). CREB activity in the nucleus accumbens shell controls gating of behavioral responses to emotional stimuli. Proc. Natl. Acad. Sci. U. S. A.. 99, 11435-11440.
- Belgacem, Y.H., Borodinsky, L.N. (2017). CREB at the crossroads of activity-dependent regulation of nervous system development and function. Adv. Exp. Med. Biol.. 1015, 19-39.
- Bianchi, L.M., Conover, J.C., Fritzsch, B., DeChiara, T., Lindsay, R.M., Yancopoulos, G.D. (1996). Degeneration of vestibular neurons in late embryogenesis of both heterozygous and homozygous BDNF null mutant mice. Development. 122, 1965-1973.
- Bleckmann, S.C., Blendy, J.A., Rudolph, D., Monaghan, A.P., Schmid, W., Schütz, G. (2002). Activating transcription factor 1 and CREB are important for cell survival during early mouse development. Mol. Cell. Biol.. 22, 1919-1925.
- Blendy, J.A. (2006). The role of CREB in depression and antidepressant treatment. Biol. Psychiatry. 59, 1144-1150.
- Blendy, J.A., Kaestner, K.H., Weinbauer, G.F., Nieschlag, E., Schütz, G. (1996). Severe impairment of spermatogenesis in mice lacking the CREM gene. Nature. 380, 162-165.
- Cameron, H.A., Glover, L.R. (2015). Adult neurogenesis: beyond learning and memory. Annu. Rev. Psychol.. 66, 53-81.
- Caracciolo, L., Marosi, M., Mazzitelli, J., Latifi, S., Sano, Y., Galvan, L., Kawaguchi, R., Holley, S., Levine, M.S., Coppola, G. (2018). CREB controls cortical circuit plasticity and functional recovery after stroke. Nat. Commun.. 9, 2250.
- Castellucci, V.F., Kandel, E.R., Schwartz, J.H., Wilson, F.D., Nairn, A.C., Greengard, P. (1980). Intracellular injection of the catalytic subunit of cyclic AMP-dependent protein kinase simulates facilitation of transmitter release underlying behavioral sensitization in Aplysia. Proc. Natl. Acad. Sci. U. S. A.. 77, 7492-7496.
- Cesare, D.D., Jacquot, S., Hanauer, A., Sassone-Corsi, P. (1998). Rsk-2 activity is necessary for epidermal growth factor-induced phosphorylation of CREB protein and transcription of c-fos gene. Proc. Natl. Acad. Sci. U. S. A.. 95, 12202-12207.
- Chan, E.C., Dusting, G.J., Guo, N., Peshavariya, H.M., Taylor, C.J., Dilley, R., Narumiya, S., Jiang, F. (2010). Prostacyclin receptor suppresses cardiac fibrosis: role of CREB phosphorylation. J. Mol. Cell. Cardiol.. 49, 176-185.
- Cho, E.C., Mitton, B., Sakamoto, K.M. (2011). CREB and leukemogenesis. Crit. Rev. Oncog.. 16, 37-46.
- Conkright, M.D., Canettieri, G., Screaton, R., Guzman, E., Miraglia, L., Hogenesch, J.B., Montminy, M. (2003). TORCs: transducers of regulated CREB activity. Mol. Cell. 12, 413-423.
- Deng, X., Deng, L., Wang, P., Cheng, C., Xu, K. (2016). Post-translational modification of CREB-1 decreases collagen I expression by inhibiting the TGF-β1 signaling pathway in rat hepatic stellate cells. Mol. Med. Rep.. 14, 5751-5759.
- Ditlevsen, D.K., Povlsen, G.K., Berezin, V., Bock, E. (2008). NCAM-induced intracellular signaling revisited. J. Neurosci. Res.. 86, 727-743.
- Dooley, K.A., Bennett, M.K., Osborne, T.F. (1999). A critical role for cAMP response element-binding protein (CREB) as a Co-activator in sterol-regulated transcription of 3-hydroxy-3-methylglutaryl coenzyme A synthase promoter. J. Biol. Chem.. 274, 5285-5291.
- Dooley, K.A., Millinder, S., Osborne, T.F. (1998). Sterol regulation of 3-hydroxy-3-methylglutaryl-coenzyme A synthase gene through a direct interaction between sterol regulatory element binding protein and the trimeric CCAAT-binding factor/nuclear factor Y. J. Biol. Chem.. 273, 1349-1356.
- Dragunow, M. (2004). CREB and neurodegeneration. Front. Biosci.. 9, 100-103.
- Eggert, S., Kins, S., Endres, K., Brigadski, T. (2022). Brothers in arms: proBDNF/BDNF and sAPPα/Aβ-signaling and their common interplay with ADAM10, TrkB, p75NTR, sortilin, and sorLA in the progression of Alzheimer's disease. Biol. Chem.. 403, 43-71.
- Fang, Y.L., Chen, H., Wang, C.L., Liang, L. (2018). Pathogenesis of non-alcoholic fatty liver disease in children and adolescence: from "two hit theory" to "multiple hit model". World J. Gastroenterol.. 24, 2974-2983.
- Fentzke, R.C., Korcarz, C.E., Lang, R.M., Lin, H., Leiden, J.M. (1998). Dilated cardiomyopathy in transgenic mice expressing a dominant-negative CREB transcription factor in the heart. J. Clin. Invest.. 101, 2415-2426.
- Ferraris, J.D., Persaud, P., Williams, C.K., Chen, Y., Burg, M.B. (2002). cAMP-independent role of PKA in tonicity-induced transactivation of tonicity-responsive enhancer/ osmotic response element-binding protein. Proc. Natl. Acad. Sci. U. S. A.. 99, 16800-16805.
- Fisher, M.L., LeMalefant, R.M., Zhou, L., Huang, G., Turner, J.R. (2017). Distinct roles of CREB within the ventral and dorsal hippocampus in mediating nicotine withdrawal phenotypes. Neuropsychopharmacology. 42, 1599-1609.
- Friedrich, M., Heimer, N., Stoehr, C., Steven, A., Wach, S., Taubert, H., Hartmann, A., Seliger, B. (2020). CREB1 is affected by the microRNAs miR-22-3p, miR-26a-5p, miR-27a-3p, and miR-221-3p and correlates with adverse clinicopathological features in renal cell carcinoma. Sci. Rep.. 10, 6499.
- Garnier, A., Fortin, D., Deloménie, C., Momken, I., Veksler, V., Ventura-Clapier, R. (2003). Depressed mitochondrial transcription factors and oxidative capacity in rat failing cardiac and skeletal muscles. J. Physiol.. 551, 491-501.
- Gass, P., Riva, M.A. (2007). CREB, neurogenesis and depression. Bioessays. 29, 957-961.
- Ghosh, S., May, M.J., Kopp, E.B. (1998). NF-κB and Rel proteins: evolutionarily conserved mediators of immune responses. Annu. Rev. Immunol.. 16, 225-260.
- Gonzalez, G.A., Montminy, M.R. (1989). Cyclic AMP stimulates somatostatin gene transcription by phosphorylation of CREB at serine 133. Cell. 59, 675-680.
- Hai, T., Hartman, M.G. (2001). The molecular biology and nomenclature of the activating transcription factor/cAMP responsive element binding family of transcription factors: activating transcription factor proteins and homeostasis. Gene. 273, 1-11.
- Hallam, T.M., Bourtchouladze, R. (2006). Rubinstein-Taybi syndrome: molecular findings and therapeutic approaches to improve cognitive dysfunction. Cell. Mol. Life Sci.. 63, 1725-1735.
- Hampel, H., Hardy, J., Blennow, K., Chen, C., Perry, G., Kim, S.H., Villemagne, V.L., Aisen, P., Vendruscolo, M., Iwatsubo, T. (2021). The amyloid-β pathway in Alzheimer's disease. Mol. Psychiatry. 26, 5481-5503.
- Han, H.S., Choi, B.H., Kim, J.S., Kang, G., Koo, S.H. (2017). Hepatic Crtc2 controls whole body energy metabolism via a miR-34a-Fgf21 axis. Nat. Commun.. 8, 1878.
- Han, H.S., Kang, G., Kim, J.S., Choi, B.H., Koo, S.H. (2016). Regulation of glucose metabolism from a liver-centric perspective. Exp. Mol. Med.. 48, e218.
- Han, H.S., Kwon, Y., Koo, S.H. (2020). Role of CRTC2 in metabolic homeostasis: key regulator of whole-body energy metabolism?. Diabetes Metab. J.. 44, 498-508.
- Harum, K.H., Alemi, L., Johnston, M.V. (2001). Cognitive impairment in Coffin-Lowry syndrome correlates with reduced RSK2 activation. Neurology. 56, 207-214.
- Herzig, S., Hedrick, S., Morantte, I., Koo, S.H., Galimi, F., Montminy, M. (2003). CREB controls hepatic lipid metabolism through nuclear hormone receptor PPAR-γ. Nature. 426, 190-193.
- Herzig, S., Long, F., Jhala, U.S., Hedrick, S., Quinn, R., Bauer, A., Rudolph, D., Schutz, G., Yoon, C., Puigserver, P. (2001). CREB regulates hepatic gluconeogenesis through the coactivator PGC-1. Nature. 413, 179-183.
- Hu, X., Paik, P.K., Chen, J., Yarilina, A., Kockeritz, L., Lu, T.T., Woodgett, J.R., Ivashkiv, L.B. (2006). IFN-γ suppresses IL-10 production and synergizes with TLR2 by regulating GSK3 and CREB/AP-1 proteins. Immunity. 24, 563-574.
- Huang, T.Y., Zheng, D., Houmard, J.A., Brault, J.J., Hickner, R.C., Cortright, R.N. (2017). Overexpression of PGC-1α increases peroxisomal activity and mitochondrial fatty acid oxidation in human primary myotubes. Am. J. Physiol. Endocrinol. Metab.. 312, E253-E263.
- Huang, W., Liu, X., Cao, J., Meng, F., Li, M., Chen, B., Zhang, J. (2015). miR-134 regulates ischemia/reperfusion injury-induced neuronal cell death by regulating CREB signaling. J. Mol. Neurosci.. 55, 821-829.
- Hughes-Fulford, M., Sugano, E., Schopper, T., Li, C.F., Boonyaratanakornkit, J.B., Cogoli, A. (2005). Early immune response and regulation of IL-2 receptor subunits. Cell. Signal.. 17, 1111-1124.
- Impey, S., McCorkle, S.R., Cha-Molstad, H., Dwyer, J.M., Yochum, G.S., Boss, J.M., McWeeney, S., Dunn, J.J., Mandel, G., Goodman, R.H. (2004). Defining the CREB regulon: a genome-wide analysis of transcription factor regulatory regions. Cell. 119, 1041-1054.
- Iourgenko, V., Zhang, W., Mickanin, C., Daly, I., Jiang, C., Hexham, J.M., Orth, A.P., Miraglia, L., Meltzer, J., Garza, D. (2003). Identification of a family of cAMP response element-binding protein coactivators by genome-scale functional analysis in mammalian cells. Proc. Natl. Acad. Sci. U. S. A.. 100, 12147-12152.
- Jeong, S. (2017). Molecular and cellular basis of neurodegeneration in Alzheimer's disease. Mol. Cells. 40, 613-620.
- Jiang, H., Nucifora, F.C.J., Ross, C.A., DeFranco, D.B. (2003). Cell death triggered by polyglutamine-expanded huntingtin in a neuronal cell line is associated with degradation of CREB-binding protein. Hum. Mol. Genet.. 12, 1-12.
- Josselyn, S.A., Nguyen, P.V. (2005). CREB, synapses and memory disorders: past progress and future challenges. Curr. Drug Targets CNS Neurol. Disord.. 4, 481-497.
- Kaang, B.K., Kandel, E.R., Grant, S.G. (1993). Activation of cAMP-responsive genes by stimuli that produce long-term facilitation in Aplysia sensory neurons. Neuron. 10, 427-435.
- Kalkhoven, E., Roelfsema, J.H., Teunissen, H., den Boer, A., Ariyurek, Y., Zantema, A., Breuning, M.H., Hennekam, R.C.M., Peters, D.J.M. (2003). Loss of CBP acetyltransferase activity by PHD finger mutations in Rubinstein-Taybi syndrome. Hum. Mol. Genet.. 12, 441-450.
- Kay, C., Collins, J.A., Miedzybrodzka, Z., Madore, S.J., Gordon, E.S., Gerry, N., Davidson, M., Slama, R.A., Hayden, M.R. (2016). Huntington disease reduced penetrance alleles occur at high frequency in the general population. Neurology. 87, 282-288.
- Kida, S., Josselyn, S.A., Peña, , de Ortiz, S., Kogan, J.H., Chevere, I., Masushige, S., Silva, A.J. (2002). CREB required for the stability of new and reactivated fear memories. Nat. Neurosci.. 5, 348-355.
- Kogan, J.H., Frankland, P.W., Blendy, J.A., Coblentz, J., Marowitz, Z., Schütz, G., Silva, A.J. (1997). Spaced training induces normal long-term memory in CREB mutant mice. Curr. Biol.. 7, 1-11.
- Kudo, K., Wati, H., Qiao, C., Arita, J., Kanba, S. (2005). Age-related disturbance of memory and CREB phosphorylation in CA1 area of hippocampus of rats. Brain Res.. 1054, 30-37.
- Lamarre-Vincent, N., Hsieh-Wilson, L.C. (2003). Dynamic glycosylation of the transcription factor CREB: a potential role in gene regulation. J. Am. Chem. Soc.. 125, 6612-6613.
- Landles, C., Bates, G.P. (2004). Huntingtin and the molecular pathogenesis of Huntington's disease. Fourth in molecular medicine review series. EMBO Rep.. 5, 958-963.
- Li, G., Jiang, Q., Xu, K. (2019). CREB family: a significant role in liver fibrosis. Biochimie. 163, 94-100.
- Liang, H., Ward, W.F. (2006). PGC-1α: a key regulator of energy metabolism. Adv. Physiol. Educ.. 30, 145-151.
- Lin, J., Handschin, C., Spiegelman, B.M. (2005). Metabolic control through the PGC-1 family of transcription coactivators. Cell Metab.. 1, 361-370.
- Linnerth-Petrik, N.M., Santry, L.A., Yu, D.L., Wootton, S.K. (2012). Adeno-associated virus vector mediated expression of an oncogenic retroviral envelope protein induces lung adenocarcinomas in immunocompetent mice. PLoS One. 7, e51400.
- Liu, J., Liu, B., Yuan, P., Cheng, L., Sun, H., Gui, J., Pan, Y., Huang, D., Chen, H., Jiang, L. (2021). Role of PKA/CREB/BDNF signaling in PM2.5-induced neurodevelopmental damage to the hippocampal neurons of rats. Ecotoxicol. Environ. Saf.. 214, 112005.
- Liu, Y., Xu, H., Geng, Y., Xu, D., Zhang, L., Yang, Y., Wei, Z., Zhang, B., Li, S., Gao, X. (2017). Dibutyryl-cAMP attenuates pulmonary fibrosis by blocking myofibroblast differentiation via PKA/CREB/CBP signaling in rats with silicosis. Respir. Res.. 18, 38.
- Loerch, P.M., Lu, T., Dakin, K.A., Vann, J.M., Isaacs, A., Geula, C., Wang, J., Pan, Y., Gabuzda, D.H., Li, C. (2008). Evolution of the aging brain transcriptome and synaptic regulation. PLoS One. 3, e3329.
- Ma, Y.Y., Chu, N.N., Guo, C.Y., Han, J.S., Cui, C.L. (2007). NR2B-containing NMDA receptor is required for morphine-but not stress-induced reinstatement. Exp. Neurol.. 203, 309-319.
- Mantamadiotis, T., Lemberger, T., Bleckmann, S.C., Kern, H., Kretz, O., Martin Villalba, A., Tronche, F., Kellendonk, C., Gau, D., Kapfhammer, J. (2002). Disruption of CREB function in brain leads to neurodegeneration. Nat. Genet.. 31, 47-54.
- Mayo, L.D., Kessler, K.M., Pincheira, R., Warren, R.S., Donner, D.B. (2001). Vascular endothelial cell growth factor activates CRE-binding protein by signaling through the KDR receptor tyrosine kinase. J. Biol. Chem.. 276, 25184-25189.
- Mayr, B., Montminy, M. (2001). Transcriptional regulation by the phosphorylation-dependent factor CREB. Nat. Rev. Mol. Cell Biol.. 2, 599-609.
- McCampbell, A., Taylor, J.P., Taye, A.A., Robitschek, J., Li, M., Walcott, J., Merry, D., Chai, Y., Paulson, H., Sobue, G. (2000). CREB-binding protein sequestration by expanded polyglutamine. Hum. Mol. Genet.. 9, 2197-2202.
- Mehrhof, F.B., Müller, F.U., Bergmann, M.W., Li, P., Wang, Y., Schmitz, W., Dietz, R., Von Harsdorf, R. (2001). In cardiomyocyte hypoxia, insulin-like growth factor-I-induced antiapoptotic signaling requires phosphatidylinositol-3-OH-kinase-dependent and mitogen-activated protein kinase-dependent activation of the transcription factor cAMP response element-binding protein. Circulation. 104, 2088-2094.
- Meller, R., Minami, M., Cameron, J.A., Impey, S., Chen, D., Lan, J.Q., Henshall, D.C., Simon, R.P. (2005). CREB-mediated Bcl-2 protein expression after ischemic preconditioning. J. Cereb. Blood Flow Metab.. 25, 234-246.
- Mitsuda, N., Ohkubo, N., Tamatani, M., Lee, Y.D., Taniguchi, M., Namikawa, K., Kiyama, H., Yamaguchi, A., Sato, N., Sakata, K. (2001). Activated cAMP-response element-binding protein regulates neuronal expression of presenilin-1. J. Biol. Chem.. 276, 9688-9698.
- Montminy, M., Koo, S.H., Zhang, X. (2004). The CREB family: key regulators of hepatic metabolism. Ann. Endocrinol. (Paris). 65, 73-75.
- Montminy, M.R., Gonzalez, G.A., Yamamoto, K.K. (1990). Regulation of cAMP-inducible genes by CREB. Trends Neurosci.. 13, 184-188.
- Morris, K.A., Gold, P.E. (2012). Age-related impairments in memory and in CREB and pCREB expression in hippocampus and amygdala following inhibitory avoidance training. Mech. Ageing Dev.. 133, 291-299.
- Noguchi, S., Kumazaki, M., Mori, T., Baba, K., Okuda, M., Mizuno, T., Akao, Y. (2016). Analysis of microRNA-203 function in CREB/MITF/RAB27a pathway: comparison between canine and human melanoma cells. Vet. Comp. Oncol.. 14, 384-394.
- Oh, G.S., Kim, S.R., Lee, E.S., Yoon, J., Shin, M.K., Ryu, H.K., Kim, D.S., Kim, S.W. (2022). Regulation of hepatic gluconeogenesis by nuclear receptor coactivator 6. Mol. Cells. 45, 180-192.
- Oh, K.J., Han, H.S., Kim, M.J., Koo, S.H. (2013). CREB and FoxO1: two transcription factors for the regulation of hepatic gluconeogenesis. BMB Rep.. 46, 567-574.
- Ollivier, V., Parry, G.C., Cobb, R.R., de Prost, D., Mackman, N. (1996). Elevated cyclic AMP inhibits NF-κB-mediated transcription in human monocytic cells and endothelial cells. J. Biol. Chem.. 271, 20828-20835.
- Ostojić, J., Yoon, Y.S., Sonntag, T., Nguyen, B., Vaughan, J.M., Shokhirev, M., Montminy, M. (2021). Transcriptional co-activator regulates melanocyte differentiation and oncogenesis by integrating cAMP and MAPK/ERK pathways. Cell Rep.. 35, 109136.
- Parra-Damas, A., Rubió-Ferrarons, L., Shen, J., Saura, C.A. (2017). CRTC1 mediates preferential transcription at neuronal activity-regulated CRE/TATA promoters. Sci. Rep.. 7, 18004.
- Petersen, M.C., Vatner, D.F., Shulman, G.I. (2017). Regulation of hepatic glucose metabolism in health and disease. Nat. Rev. Endocrinol.. 13, 572-587.
- Petrij, F., Giles, R.H., Dauwerse, H.G., Saris, J.J., Hennekam, R.C., Masuno, M., Tommerup, N., van Ommen, G.J., Goodman, R.H., Peters, D.J. (1995). Rubinstein-Taybi syndrome caused by mutations in the transcriptional co-activator CBP. Nature. 376, 348-351.
- Pigazzi, M., Manara, E., Baron, E., Basso, G. (2009). miR-34b targets cyclic AMP-responsive element binding protein in acute myeloid leukemia. Cancer Res.. 69, 2471-2478.
- Pignataro, A., Borreca, A., Ammassari-Teule, M., Middei, S. (2015). CREB regulates experience-dependent spine formation and enlargement in mouse barrel cortex. Neural Plast.. 2015, 651469.
- Pittenger, C., Huang, Y.Y., Paletzki, R.F., Bourtchouladze, R., Scanlin, H., Vronskaya, S., Kandel, E.R. (2002). Reversible inhibition of CREB/ATF transcription factors in region CA1 of the dorsal hippocampus disrupts hippocampus-dependent spatial memory. Neuron. 34, 447-462.
- Planavila, A., Rodríguez-Calvo, R., Jové, M., Michalik, L., Wahli, W., Laguna, J.C., Vázquez-Carrera, M. (2005). Peroxisome proliferator-activated receptor β/δ activation inhibits hypertrophy in neonatal rat cardiomyocytes. Cardiovasc. Res.. 65, 832-841.
- Portilla, D., Dai, G., McClure, T., Bates, L., Kurten, R., Megyesi, J., Price, P., Li, S. (2002). Alterations of PPARα and its coactivator PGC-1 in cisplatin-induced acute renal failure. Kidney Int.. 62, 1208-1218.
- Purves, D., Augustine, G.J., Fitzpatrick, D., Hall, W.C., LaMantia, A.S., McNamara, J.O., White, L.E. (2008). . Neuroscience, , ed. (Sunderland:Sinauer Associates), pp. .
- Quinn, P.G., Granner, D.K. (1990). Cyclic AMP-dependent protein kinase regulates transcription of the phosphoenolpyruvate carboxykinase gene but not binding of nuclear factors to the cyclic AMP regulatory element. Mol. Cell. Biol.. 10, 3357-3364.
- Rall, T.W., Sutherland, E.W., Wosilait, W.D. (1956). The relationship of epinephrine and glucagon to liver phosphorylase: iii. Reactivation of liver phosphorylase in slices and in extracts. J. Biol. Chem.. 218, 483-495.
- Riccio, A., Ahn, S., Davenport, C.M., Blendy, J.A., Ginty, D.D. (1999). Mediation by a CREB family transcription factor of NGF-dependent survival of sympathetic neurons. Science. 286, 2358-2361.
- Rowe, G.C., Arany, Z. (2014). Genetic models of PGC-1 and glucose metabolism and homeostasis. Rev. Endocr. Metab. Disord.. 15, 21-29.
- Rudolph, D., Tafuri, A., Gass, P., Hämmerling, G.J., Arnold, B., Schütz, G. (1998). Impaired fetal T cell development and perinatal lethality in mice lacking the cAMP response element binding protein. Proc. Natl. Acad. Sci. U. S. A.. 95, 4481-4486.
- Rui, L. (2014). Energy metabolism in the liver. Compr. Physiol.. 4, 177-197.
- Sakamoto, K.M., Frank, D.A. (2009). CREB in the pathophysiology of cancer: implications for targeting transcription factors for cancer therapy. Clin. Cancer Res.. 15, 2583-2587.
- Sandoval, S., Pigazzi, M., Sakamoto, K.M. (2009). CREB: a key regulator of normal and neoplastic hematopoiesis. Adv. Hematol.. 2009, 634292.
- Sands, W.A., Palmer, T.M. (2008). Regulating gene transcription in response to cyclic AMP elevation. Cell. Signal.. 20, 460-466.
- Sano, M., Wang, S.C., Shirai, M., Scaglia, F., Xie, M., Sakai, S., Tanaka, T., Kulkarni, P.A., Barger, P.M., Youker, K.A. (2004). Activation of cardiac Cdk9 represses PGC-1 and confers a predisposition to heart failure. EMBO J.. 23, 3559-3569.
- Sapio, L., Salzillo, A., Ragone, A., Illiano, M., Spina, A., Naviglio, S. (2020). Targeting CREB in cancer therapy: a key candidate or one of many? An update. Cancers (Basel). 12, 3166.
- Saraiva, M., O'Garra, A. (2010). The regulation of IL-10 production by immune cells. Nat. Rev. Immunol.. 10, 170-181.
- Sassone-Corsi, P. (1995). Transcription factors responsive to cAMP. Annu. Rev. Cell Dev. Biol.. 11, 355-377.
- Saura, C.A., Valero, J. (2011). The role of CREB signaling in Alzheimer's disease and other cognitive disorders. Rev. Neurosci.. 22, 153-169.
- Schaefer, K.L., Brachwitz, K., Wai, D.H., Braun, Y., Diallo, R., Korsching, E., Eisenacher, M., Voss, R., Van Valen, F., Baer, C. (2004). Expression profiling of t(12;22) positive clear cell sarcoma of soft tissue cell lines reveals characteristic up-regulation of potential new marker genes including ERBB3. Cancer Res.. 64, 3395-3405.
- Schulte, J.S., Seidl, M.D., Nunes, F., Freese, C., Schneider, M., Schmitz, W., Müller, F.U. (2012). CREB critically regulates action potential shape and duration in the adult mouse ventricle. Am. J. Physiol. Heart Circ. Physiol.. 302, H1998-H2007.
- Scoditti, E., Massaro, M., Carluccio, M.A., Distante, A., Storelli, C., De Caterina, R. (2010). PPARγ agonists inhibit angiogenesis by suppressing PKCα-and CREB-mediated COX-2 expression in the human endothelium. Cardiovasc. Res.. 86, 302-310.
- Sekeres, M.J., Neve, R.L., Frankland, P.W., Josselyn, S.A. (2010). Dorsal hippocampal CREB is both necessary and sufficient for spatial memory. Learn. Mem.. 17, 280-283.
- Shankar, D.B., Cheng, J.C., Kinjo, K., Federman, N., Moore, T.B., Gill, A., Rao, N.P., Landaw, E.M., Sakamoto, K.M. (2005). The role of CREB as a proto-oncogene in hematopoiesis and in acute myeloid leukemia. Cancer Cell. 7, 351-362.
- Smolik, S.M., Rose, R.E., Goodman, R.H. (1992). A cyclic AMP-responsive element-binding transcriptional activator in Drosophilamelanogaster, dCREB-A, is a member of the leucine zipper family. Mol. Cell. Biol.. 12, 4123-4131.
- Steffan, J.S., Bodai, L., Pallos, J., Poelman, M., McCampbell, A., Apostol, B.L., Kazantsev, A., Schmidt, E., Zhu, Y.Z., Greenwald, M. (2001). Histone deacetylase inhibitors arrest polyglutamine-dependent neurodegeneration in Drosophila. Nature. 413, 739-743.
- Stepulak, A., Sifringer, M., Rzeski, W., Brocke, K., Gratopp, A., Pohl, E.E., Turski, L., Ikonomidou, C. (2007). AMPA antagonists inhibit the extracellular signal regulated kinase pathway and suppress lung cancer growth. Cancer Biol. Ther.. 6, 1908-1915.
- Steven, A., Friedrich, M., Jank, P., Heimer, N., Budczies, J., Denkert, C., Seliger, B. (2020). What turns CREB on? And off? And why does it matter?. Cell. Mol. Life Sci.. 77, 4049-4067.
- Suresh, R., Mosser, D.M. (2013). Pattern recognition receptors in innate immunity, host defense, and immunopathology. Adv. Physiol. Educ.. 37, 284-291.
- Sutherland, E.W. (1972). Studies on the mechanism of hormone action. Science. 177, 401-408.
- Tabuchi, A., Sakaya, H., Kisukeda, T., Fushiki, H., Tsuda, M. (2002). Involvement of an upstream stimulatory factor as well as cAMP-responsive element-binding protein in the activation of brain-derived neurotrophic factor gene promoter I. J. Biol. Chem.. 277, 35920-35931.
- Tan, X., Wang, S., Zhu, L., Wu, C., Yin, B., Zhao, J., Yuan, J., Qiang, B., Peng, X. (2012). cAMP response element-binding protein promotes gliomagenesis by modulating the expression of oncogenic microRNA-23a. Proc. Natl. Acad. Sci. U. S. A.. 109, 15805-15810.
- Tokunou, T., Shibata, R., Kai, H., Ichiki, T., Morisaki, T., Fukuyama, K., Ono, H., Iino, N., Masuda, S., Shimokawa, H. (2003). Apoptosis induced by inhibition of cyclic AMP response element-binding protein in vascular smooth muscle cells. Circulation. 108, 1246-1252.
- Tran, M., Tam, D., Bardia, A., Bhasin, M., Rowe, G.C., Kher, A., Zsengeller, Z.K., Reza Akhavan-Sharif, M., Khankin, E.V., Saintgeniez, M. (2011). PGC-1α promotes recovery after acute kidney injury during systemic inflammation in mice. J. Clin. Invest.. 121, 4003-4014.
- Trivier, E., De Cesare, D., Jacquot, S., Pannetier, S., Zackai, E., Young, I., Mandel, J.L., Sassone-Corsi, P., Hanauer, A. (1996). Mutations in the kinase Rsk-2 associated with Coffin-Lowry syndrome. Nature. 384, 567-570.
- Truong, V., Anand-Srivastava, M.B., Srivastava, A.K. (2021). Role of cyclic AMP response element binding protein (CREB) in angiotensin II-induced responses in vascular smooth muscle cells. Can. J. Physiol. Pharmacol.. 99, 30-35.
- Tully, T., Bourtchouladze, R., Scott, R., Tallman, J. (2003). Targeting the CREB pathway for memory enhancers. Nat. Rev. Drug Discov.. 2, 267-277.
- Usui, T., Smolik, S.M., Goodman, R.H. (1993). Isolation of Drosophila CREB-B: a novel CRE-binding protein. DNA Cell Biol.. 12, 589-595.
- Vitolo, O.V., Sant'Angelo, A., Costanzo, V., Battaglia, F., Arancio, O., Shelanski, M. (2002). Amyloid β-peptide inhibition of the PKA/CREB pathway and long-term potentiation: reversibility by drugs that enhance cAMP signaling. Proc. Natl. Acad. Sci. U. S. A.. 99, 13217-13221.
- Vo, N., Klein, M.E., Varlamova, O., Keller, D.M., Yamamoto, T., Goodman, R.H., Impey, S. (2005). A cAMP-response element binding protein-induced microRNA regulates neuronal morphogenesis. Proc. Natl. Acad. Sci. U. S. A.. 102, 16426-16431.
- Voropaev, H., Gimmelshein Vatkin, M., Shneor, D., Luski, S., Honigman, A., Frenkel, S. (2019). Infectious knockdown of CREB and HIF-1 for the treatment of metastatic uveal melanoma. Cancers (Basel). 11, 1056.
- Wang, G., Cheng, Z., Liu, F., Zhang, H., Li, J., Li, F. (2015a). CREB is a key negative regulator of carbonic anhydrase IX (CA9) in gastric cancer. Cell. Signal.. 27, 1369-1379.
- Wang, H., Xu, J., Lazarovici, P., Quirion, R., Zheng, W. (2018). cAMP response element-binding protein (CREB): a possible signaling molecule link in the pathophysiology of schizophrenia. Front. Mol. Neurosci.. 11, 255.
- Wang, R., Zhang, Y.W., Zhang, X., Liu, R., Zhang, X., Hong, S., Xia, K., Xia, J., Zhang, Z., Xu, H. (2006). Transcriptional regulation of APH-1A and increased γ-secretase cleavage of APP and Notch by HIF-1 and hypoxia. FASEB J.. 20, 1275-1277.
- Wang, T., Wiater, E., Zhang, X., Thomas, J.B., Montminy, M. (2021). Crtc modulates fasting programs associated with 1-C metabolism and inhibition of insulin signaling. Proc. Natl. Acad. Sci. U. S. A.. 118, e2024865118.
- Wang, Y., Viscarra, J., Kim, S.J., Sul, H.S. (2015b). Transcriptional regulation of hepatic lipogenesis. Nat. Rev. Mol. Cell Biol.. 16, 678-689.
- Wen, A.Y., Sakamoto, K.M., Miller, L.S. (2010). The role of the transcription factor CREB in immune function. J. Immunol.. 185, 6413-6419.
- Xia, Y., Zhan, C., Feng, M., Leblanc, M., Ke, E., Yeddula, N., Verma, I.M. (2018). Targeting CREB pathway suppresses small cell lung cancer. Mol. Cancer Res.. 16, 825-832.
- Xiang, H., Wang, J., Boxer, L.M. (2006). Role of the cyclic AMP response element in the bcl-2 promoter in the regulation of endogenous Bcl-2 expression and apoptosis in murine B cells. Mol. Cell. Biol.. 26, 8599-8606.
- Xiao, X., Li, B.X., Mitton, B., Ikeda, A., Sakamoto, K.M. (2010). Targeting CREB for cancer therapy: friend or foe. Curr. Cancer Drug Targets. 10, 384-391.
- Xie, F., Li, B.X., Kassenbrock, A., Xue, C., Wang, X., Qian, D.Z., Sears, R.C., Xiao, X. (2015). Identification of a potent inhibitor of CREB-mediated gene transcription with efficacious in vivo anticancer activity. J. Med. Chem.. 58, 5075-5087.
- Xing, J., Ginty, D.D., Greenberg, M.E. (1996). Coupling of the RAS-MAPK pathway to gene activation by RSK2, a growth factor-regulated CREB kinase. Science. 273, 959-963.
- Yamamoto, K., Ohki, R., Lee, R.T., Ikeda, U., Shimada, K. (2001). Peroxisome proliferator-activated receptor γ activators inhibit cardiac hypertrophy in cardiac myocytes. Circulation. 104, 1670-1675.
- Yin, J.C., Tully, T. (1996). CREB and the formation of long-term memory. Curr. Opin. Neurobiol.. 6, 264-268.
- Yin, J.C., Wallach, J.S., Del Vecchio, M., Wilder, E.L., Zhou, H., Quinn, W.G., Tully, T. (1994). Induction of a dominant negative CREB transgene specifically blocks long-term memory in Drosophila. Cell. 79, 49-58.
- Yin, J.C.P., Del Vecchio, M., Zhou, H., Tully, T. (1995). CREB as a memory modulator: induced expression of a dCREB2 activator isoform enhances long-term memory in Drosophila. Cell. 81, 107-115.
- Yin, P., Li, D., Zhao, Q., Cai, M., Wu, Z., Shi, Y., Su, L. (2021). Gsα deficiency facilitates cardiac remodeling via CREB/ Bmp10-mediated signaling. Cell Death Discov.. 7, 391.
- Zhang, C., Wu, B., Beglopoulos, V., Wines-Samuelson, M., Zhang, D., Dragatsis, I., Südhof, T.C., Shen, J. (2009). Presenilins are essential for regulating neurotransmitter release. Nature. 460, 632-636.
- Zhang, J., Yao, Q., Kuang, Y., Ma, Y., Yang, L., Huang, H., Cheng, J., Yang, T., Liu, E., Liang, L. (2014). Prognostic value of coexistence of abnormal expression of micro-RNA-200b and cyclic adenosine monophosphate-responsive element-binding protein 1 in human astrocytoma. Hum. Pathol.. 45, 2154-2161.
- Zhang, L., Guo, X., Zhang, D., Fan, Y., Qin, L., Dong, S., Zhang, L. (2017). Upregulated miR-132 in Lgr5(+) gastric cancer stem cell-like cells contributes to cisplatin-resistance via SIRT1/CREB/ABCG2 signaling pathway. Mol. Carcinog.. 56, 2022-2034.
- Zhang, Q., Koser, S.L., Donkin, S.S. (2016). Propionate induces mRNA expression of gluconeogenic genes in bovine calf hepatocytes. J. Dairy Sci.. 99, 3908-3915.
- Zhang, X., Yu, K., Ma, L., Qian, Z., Tian, X., Miao, Y., Niu, Y., Xu, X., Guo, S., Yang, Y. (2021). Endogenous glutamate determines ferroptosis sensitivity via ADCY10-dependent YAP suppression in lung adenocarcinoma. Theranostics. 11, 5650-5674.
- Zhang, Y., Yang, J., Cui, X., Chen, Y., Zhu, V.F., Hagan, J.P., Wang, H., Yu, X., Hodges, S.E., Fang, J. (2013). A novel epigenetic CREB-miR-373 axis mediates ZIP4-induced pancreatic cancer growth. EMBO Mol. Med.. 5, 1322-1334.
- Zhang, Y., Zheng, D., Zhou, T., Song, H., Hulsurkar, M., Su, N., Liu, Y., Wang, Z., Shao, L., Ittmann, M. (2018). Androgen deprivation promotes neuroendocrine differentiation and angiogenesis through CREB-EZH2-TSP1 pathway in prostate cancers. Nat. Commun.. 9, 4080.