Skip to main content
Biologics : Targets & Therapy logoLink to Biologics : Targets & Therapy
. 2025 May 30;19:341–350. doi: 10.2147/BTT.S522325

Targeting CREBRF in Cancer: Mechanistic Insights and Future Directions

Baixue Lv 1, Dongdong Zhang 1,
PMCID: PMC12132502  PMID: 40463837

Abstract

Luman/CREB3 recruitment factor (LRF), also known as CREBRF, was initially identified as a cellular binding protein of Luman through yeast two-hybrid screening of a human brain cDNA library. CREBRF plays a critical role in various biological processes, with its functions garnering significant attention in the field of oncology. Notably, CREBRF is involved in endoplasmic reticulum (ER) stress and regulates the unfolded protein response (UPR), leading to an accumulation of misfolded proteins. This can ultimately result in cellular dysfunction, apoptosis, and even tumorigenesis. In solid tumors, hypoxia is a common condition, and CREBRF has been implicated in hypoxia-induced autophagy, which promotes tumor cell proliferation. Depending on the tumor type and microenvironment, CREBRF exerts diverse effects by modulating distinct signaling pathways. This review summarizes CREBRF’s involvement in ER stress, cell cycle regulation, autophagy, and the mechanisms through which it influences tumor initiation and progression across various cancer types. Furthermore, the potential of CREBRF as a therapeutic target in cancer treatment is discussed, providing insights into future research and clinical applications.

Keywords: CREBRF, ER, hypoxia, tumor, autophagy, therapeutic target

Introduction

Tumorigenesis and cancer progression are intricate processes governed by diverse biological mechanisms. Autophagy and endoplasmic reticulum (ER) stress are interconnected processes that serve dual roles in tumor biology, acting to either suppress or promote tumors depending on the context.1 Autophagy is a cellular mechanism that degrades and recycles damaged organelles and proteins, maintaining cellular homeostasis.2 During early tumorigenesis, autophagy acts as a tumor suppressor by removing damaged components and preventing genomic instability.3 In established tumors, however, autophagy supports tumor survival by supplying nutrients during stress conditions like hypoxia or nutrient deprivation, commonly found in the tumor microenvironment.4 ER stress occurs when the ER’s protein-folding capacity is overwhelmed, triggering activation of the unfolded protein response (UPR). The UPR initially restores homeostasis by enhancing protein folding and degradation pathways.5 Tumors frequently exploit the UPR to adapt to hostile microenvironments, fostering growth and therapeutic resistance.5,6 Prolonged or unresolved ER stress can trigger apoptosis or activate additional mechanisms that foster tumorigenesis, such as the regulation of the PI3K/AKT/mTOR signaling pathway.7 Autophagy and ER stress are interconnected. During ER stress, autophagy is activated as an adaptive mechanism to mitigate misfolded protein accumulation and relieve cellular stress.8

CREBRF (Luman/CREB3 Recruiting Factor, LRF) plays a pivotal role in regulating cellular stress responses and autophagy.9,10 CREBRF mediates ER stress, initiating adaptive cellular changes. Prolonged ER stress may lead to cellular dysfunction, inflammation, apoptosis, and potentially drive tumorigenesis.11 Under hypoxic conditions, CREBRF promotes autophagy through the CREB3/ATG5 pathway.12 Aberrant CREBRF expression plays a crucial role in the development and progression of various cancers. This finding has been validated in studies on various tumor types, including gastric cancer,13 acute myeloid leukemia (AML) and cervical cancer.14 CREBRF exhibits diverse effects across tumor types, with its role varying depending on tumor type and microenvironment. In gastric cancer,13 CREBRF promotes tumor progression; whereas in AML, it functions as a tumor suppressor.15 In gliomas, under normal conditions, elevated CREBRF expression activates the AKT pathway, driving tumor cell proliferation.16 Under hypoxic conditions, elevated CREBRF expression reduces autophagy levels, subsequently suppressing tumor cell proliferation.12 Therefore, CREBRF’s functional roles may be influenced by alterations in the tumor microenvironment. Additionally, a CREBRF mutation is associated with increased obesity and a significant reduction in type 2 diabetes risk, potentially influencing cancer risk.17,18

Research on the CREBRF gene is still in its early stages, and its exact role in tumor development and progression remains unclear. Evaluating its potential as a therapeutic target in cancer is essential. Exploring how this target can be effectively utilized for cancer diagnosis and treatment is equally critical. This review explores the biological functions of CREBRF, its role in tumor initiation and progression, and the molecular mechanisms driving its involvement in cancer development. It aims to provide a theoretical foundation for the development of CREBRF-targeted anti-cancer therapies.

Overview of CREBRF

Luman/CREB3 Recruiting Factor (LRF/CREBRF) is a Luman-associated protein activated during ER stress. It was first identified using yeast two-hybrid technology in a human brain cDNA library.11 The CREBRF gene encodes a 639-amino-acid protein containing a highly acidic region, a bZIP domain, and a leucine zipper-like motif. This protein is highly conserved, sharing over 95% sequence homology with mouse and rat homologs (GenBank Gene IDs 77128 and 303016, respectively), underscoring its evolutionary and functional significance.11

cAMP response element-binding protein 3 (CREB3/Luman/CREB3/LZIP) is a central member of the CREB3 family, which includes CREB3L1/OASIS, CREB3L2/BBF2H7, CREB3L3/CREBH, and CREB3L4/AIbZIP.19,20 These proteins exhibit substantial homology and conservation in their functional domains. CREB3 family members are single-pass transmembrane proteins localized to the ER membrane.19 Their domains are arranged sequentially from the N-terminus (cytoplasmic side) to the C-terminus (luminal side), including: (1) Transactivation domain (TAD) – mediates sequence-specific DNA binding; (2) ATB domain – a conserved ~30-amino-acid region near the bZIP domain, unique to the CREB3 family; (3) bZIP domain – enables DNA binding and dimerization; and (4) Transmembrane domain (TMD) – anchors the protein to the ER membrane.19,21

During cellular stress, including endoplasmic reticulum (ER) stress, CREB3 family proteins move from the ER to the Golgi apparatus, where Site-1 protease (S1P) and Site-2 protease (S2P) sequentially cleave them.19 This cleavage generates an N-terminal fragment with a bZIP domain that functions as a transcription factor. The N-terminal fragment is transported to the nucleus, where it binds to the cAMP response element (CRE) and activates the transcription of target genes.19,22–24 CREBRF mainly acts as a negative regulator of CREB3. It does so by recruiting the CREB3 transcription factor to specific nuclear foci, suppressing its activity and promoting its degradation.11 The biological functions of CREBRF are detailed in Figure 1.

Figure 1.

Figure 1

CREBRF as a Modulator of ER Stress Response Under ER stress conditions, CREB3 undergoes proteolytic cleavage and translocates to the nucleus, where it functions as a transcription factor. CREBRF plays a critical regulatory role by inhibiting CREB3 activity, recruiting it to distinct nuclear foci and modulating its transcriptional function. By Figdraw.

CREBRF-Mediated Endoplasmic Reticulum Stress

Secretory and membrane proteins are synthesized on ribosomes attached to the ER membrane, where they fold and mature in the ER lumen.25 Genetic mutations or changes in cellular conditions can disrupt this process, causing misfolded or unfolded proteins to accumulate in the ER lumen. The ER’s limited capacity to handle proteins leads to their excessive accumulation, causing ER stress.26 Three stress sensors—inositol-requiring enzyme 1 (IRE1), activating transcription factor 6 (ATF6), and protein kinase R-like ER kinase (PERK)-detect unfolded proteins during ER stress. These sensors activate the UPR, a mechanism that alleviates stress and restores ER homeostasis. If ER stress remains unresolved, the UPR may promote apoptosis, preventing prolonged cellular dysfunction.5,19,27 In cancer cells, ER stress plays a dual role. ER stress helps tumor cells adapt to hypoxia and nutrient deprivation by activating the UPR or autophagy, promoting survival and growth. Conversely, excessive autophagy induces cell death, while prolonged severe ER stress triggers apoptosis in tumor cells.5,28

CREB3 family members, as ER stress transducers, share significant homology with ATF6, leading to similar mechanisms of transmembrane cleavage and transcriptional activation.29 During ER stress, CREB3 moves to the Golgi apparatus from the ER, where it undergoes regulated intramembrane proteolysis (RIP). This activation enables CREB3 to act as a transcription factor, regulating stress-responsive genes.22 Similarly, CREBRF is UPR-sensitive and prone to proteasomal degradation. CREBRF primarily acts as a negative regulator of CREB3, suppressing its activity.11 However, studies in Neuro2a cells indicate that CREBRF can also positively regulate CREB3, modulating ER stress.30 This dual regulation underscores the intricate interplay between CREB3 family members and ER stress in diverse cellular contexts.

CREBRF and Cell Cycle Regulation

The eukaryotic cell cycle, consisting of G1, S, G2, and M phases, is stringently regulated by checkpoints that ensure DNA integrity and halt progression upon detecting damage.31 The G1/S checkpoint inhibits CDK4/6-Cyclin D and CDK2-Cyclin E complexes to prevent damaged DNA replication. The G2/M checkpoint ensures DNA integrity and replication completeness, blocking mitotic entry if errors persist.32 Malfunctioning checkpoints impair DNA repair, leading to genomic instability and an increased risk of cancer.33

Checkpoint dysregulation is a key feature of many cancers.34–36 CREBRF regulates key cell cycle factors and transitions, playing a crucial role in biological processes like cancer progression and decidualization. In gastric cancer (GC), CREBRF activates the AKT pathway, upregulating CDK2, Cyclin D1, and Cyclin A to promote the G1/S transition and drive tumor cell proliferation.13 CREBRF also influences decidualization by regulating the proliferation and differentiation of endometrial stromal cells (ESCs) into decidual cells. Silencing CREBRF downregulates Cyclin A and Cyclin B1, sparing Cyclin D3 and Cyclin E, causing S-phase arrest and reduced ESC proliferation.37,38

CREBRF and Autophagy

Autophagy degrades and recycles cellular components via the lysosomal system, selectively or non-selectively breaking down organelles and proteins to maintain metabolic balance and cellular homeostasis.2 In tumor cells, autophagy supports metabolic demands, aiding their growth and survival.3,39 Autophagy sustains cell survival during starvation, hypoxia, immune responses, and radio-chemotherapy by providing essential resources. Late-stage cancer cells heavily depend on autophagy to survive in the nutrient-deprived tumor microenvironment.3,40–42

In breast cancer, CREBRF is significantly upregulated during drug-induced autophagy, and its high expression correlates with better clinical outcomes, suggesting its potential as an autophagy biomarker.43 Moreover, FOXC1, a transcription factor frequently overexpressed in various cancers, has been shown to bind to the promoter region of CREBRF and suppress its expression in four distinct triple-negative breast cancer (TNBC) cell lines. This finding implies that CREBRF may function as a tumor suppressor in TNBC.44 In endometrial epithelial cells, hormonal regulation upregulates CREBRF, which activates autophagy via the CREBRF-mTOR-autophagy pathway.45 Human melanoma is characterized by constant MEK/ERK pathway activation.46,47 CREB upregulates Noxa transcription through MEK/ERK signaling, inducing autophagy in melanoma cells and delaying apoptosis during nutrient starvation.48 This indicates that CREBRF may be involved in regulating apoptosis in melanoma cells. In glioblastoma, the IL6-p-STAT3-miR155-3p-CREBRF-CREB3-ATG5 axis promotes hypoxia-induced autophagy, while IL6 receptor blockade suppresses this process.10 Additionally, MicroRNA-124-3p promotes autophagy in gliomas by targeting CREBRF for downregulation.16

Solid tumors, especially malignant ones, commonly exhibit hypoxic conditions with oxygen levels significantly lower than their tissue of origin.49–51 Hypoxic tumor regions exhibit extensive autophagy.52 In glioblastoma, hypoxia downregulates CREBRF expression, correlating with elevated HIF-1α levels and promoting autophagy via the CREB3/ATG5 pathway.12 Combined knockdown of CREB3 and inhibition of ATG5 suppresses hypoxia-induced autophagy.

CREBRF in Other Aspects

CREBRF is a versatile regulatory protein engaged in diverse physiological and pathological processes. In reproductive biology, CREBRF is crucial for hormonal signaling during pregnancy, influencing implantation and embryonic development.53 It also affects maternal behavior by inhibiting the glucocorticoid receptor, which modulates the hypothalamic-pituitary-adrenal (HPA) axis and prolactin signaling.54 MicroRNA miR-181d-5p targets CREBRF, influencing placental cell function and possibly contributing to pregnancy-related disorders.55 Additionally, CREBRF is a critical regulator of muscle energy metabolism, and its dysfunction may affect tumor metabolic states.56

CREBRF and Tumors

Recently, the potential role of CREBRF in cancer therapy has gained attention (Figure 2), especially for its involvement in regulating tumor metabolism and improving therapeutic efficacy. CREBRF generally regulates tumors via the LncRNA/CircRNA-miR-CREBRF axis or the AKT signaling pathway.57–59 CREBRF promotes tumorigenesis in cervical and gastric cancers but acts as a tumor suppressor in gallbladder cancer and AML. In gliomas, particularly glioblastomas, its role varies based on cellular physiological state and microenvironment, exhibiting dual biological functions.13,14,16,60–65 Table 1 summarizes the dual roles of CREBRF in tumorigenesis, demonstrating both inhibitory and promotive effects across different cancer types.

Figure 2.

Figure 2

CREBRF plays a critical role in modulating regulatory pathways that influence the progression and therapeutic response of various cancers. By Figdraw.

Abbreviations: HNSCC, Head and neck squamous cell carcinoma; GC, Gastric cancer; AML, acute myeloid leukemia; CC, Cervical cancer.

Table 1.

The Dual Roles of CREBRF in Tumorigenesis

Cancer Types Expression Levels (Tumor vs Normal Tissue Comparison) Biological Effects Functional Impact on Tumor Progression
Glioma Upregulation
Downregulation (under hypoxic conditions)
Suppress autophagy
Induce autophagy and promote cell proliferation
Promote
Inhibit
HNSCC Downregulation Induce autophagy and enhance invasiveness and migratory capacity Inhibit
GC Upregulation Stimulate cell proliferation Promote
AML Downregulation Stimulate cell proliferation Inhibit
Gallbladder cancer Downregulation Induce autophagy and stimulate cell proliferation Inhibit
CC Upregulation Stimulate cell proliferation and enhance invasiveness and migration potential Promote

Abbreviations: GC, Gastric cancer; CC, Cervical cancer.

Cervical Cancer (CC)

Cervical cancer (CC) is the most common gynecological malignancy and ranks as the fourth leading cause of cancer-related deaths among women. CREBRF, an essential transcription factor, is upregulated in CC and strongly associated with enhanced tumor proliferation, migration, and invasiveness. Numerous CircRNAs are upregulated in CC, functioning as miRNA sponges to suppress specific miRNAs and elevate CREBRF expression, thereby promoting CC progression. For example, circ_0009035 influences CC progression by regulating the miR-1305/CREBRF axis.62 hsa_circ_0102171 targets the miR-4465/CREBRF axis, potentially promoting proliferation, reducing apoptosis, and enhancing invasion and migration in CC cells, thereby accelerating tumor progression and severity.63 Additionally, the circ_0081723/miR-545-3p/CREBRF axis and CircVIRMA/miR-452-5p/CREBRF axis are implicated in CC progression.14,60 These studies highlight the significant role of non-coding RNAs in regulating CREBRF expression and driving CC progression.

Head and Neck Squamous Cell Carcinoma (HNSCC)

Chemoradiotherapy failure is a significant contributor to recurrence, progression, and poor prognosis in head and neck squamous cell carcinoma (HNSCC).66 MicroRNAs, particularly miR-124-3p, have been identified as critical mediators of drug resistance in HNSCC. A recent study showed that miR-124-3p is overexpressed in chemoradiotherapy-resistant HNSCC tumors compared to responsive tumors. Suppressing miR-124-3p improved the sensitivity of HNSCC cell lines to standard treatments, including 5-fluorouracil, cisplatin, and radiotherapy. MiR-124-3p downregulates the transcription factor CREBRF by directly targeting it. This downregulation activates the CREB3/ATG5 signaling axis, promoting aggressive tumor behavior. Inhibiting miR-124-3p restores CREBRF expression, which may reduce tumor invasiveness and migration, enhance chemoradiotherapy efficacy, and improve survival rates in HNSCC patients.61 Therefore, targeting miR-124-3p and restoring CREBRF expression holds great promise for overcoming drug resistance in HNSCC.

AML

AML is a hematological malignancy marked by the accumulation and impaired differentiation of clonal myeloid precursor cells in the bone marrow.67 Studies have demonstrated that hsa_circ_0001947 is frequently downregulated in the bone marrow of AML patients. hsa_circ_0001947 acts as a molecular sponge for hsa-miR-329-5p, which directly targets and suppresses CREBRF mRNA expression. When hsa-miR-329-5p is sequestered by hsa_circ_0001947, CREBRF suppression is relieved, resulting in increased CREBRF expression and inhibition of AML cell proliferation.64 Furthermore, lncRNAs can reduce the levels of free miRNAs, thereby increasing the expression of miRNA-repressed target proteins.68 NEAT1 is a lncRNA downregulated in leukemia patients and cell lines, while miR-338-3p is overexpressed in AML.69,70 NEAT1 binds to miR-338-3p, mitigating its suppression of CREBRF and thereby enhancing CREBRF expression. Elevated CREBRF expression may inhibit CREB3 activity, thereby suppressing AML cell proliferation and survival while promoting apoptosis.15

Gallbladder Cancer

Gallbladder cancer is a malignant tumor derived from the epithelial cells of the gallbladder, notable for its diverse metastatic pathways and poor prognosis.71 miR-181b has demonstrated oncogenic roles in various cancers.72–74 Research suggests that in gallbladder cancer, miR-181b directly targets and suppresses CREBRF, inhibiting CREB3 degradation and promoting the expression of autophagy-related genes, including LC3 and ATG5. This mechanism enables tumor cells to manage stress, thereby enhancing their survival and proliferation.65 Rg3, a potential autophagy inhibitor, exhibits antitumor effects in several malignancies, such as colorectal and lung cancers.75,76 In gallbladder cancer, Rg3 inhibits autophagic flux and suppresses tumor progression by blocking the miR-181b/CREBRF/CREB3 pathway. Exogenous overexpression of miR-181b diminishes the tumor-suppressive effects of ginsenoside Rg3, ultimately accelerating gallbladder cancer progression.65

Glioma

Glioma is the most common primary malignant brain tumor in adults.77 In glioma cells, miR-124-3p is significantly downregulated, while CREBRF is upregulated. Overexpression of CREBRF increases p62 expression and activates the AKT pathway while suppressing autophagy-related proteins, including Beclin1 and LC3-II/I. These findings suggest that CREBRF promotes glioma cell survival by suppressing apoptosis and autophagy. Studies reveal that miR-124-3p directly targets CREBRF, inhibiting AKT activation and enhancing apoptosis and autophagy in glioma cells.16 Under hypoxic conditions, the expression of CREBRF is diminished, while CREB3 levels are upregulated, thereby promoting autophagy, inhibiting apoptosis, and driving the proliferation of glioma cells.12 Thus, understanding CREBRF function necessitates analyzing its expression levels in context with biophysiological conditions. This context-dependent regulation allows tumor cells to adapt to environmental changes, presenting potential opportunities for therapeutic intervention.

Gastric Cancer

Gastric cancer (GC) is a malignant tumor originating in the stomach and remains a major cause of cancer-related mortality worldwide.78 CREBRF is highly upregulated in GC cells, activating the AKT pathway and significantly increasing the expression of cell cycle regulators CDK2, Cyclin D1, and Cyclin A. Cyclin D1, Cyclin A, and CDK2 are essential for the G1-to-S phase transition during the cell cycle.79 Silencing CREBRF causes significant cell cycle arrest at the G1/G0 phase, thereby inhibiting GC cell proliferation. AKT pathway activation can counteract this effect and stimulate GC cell proliferation. These findings suggest that CREBRF may be a promising therapeutic target for gastric cancer.13

Future Directions of CREBRF in Cancer Treatment

CREBRF is pivotal in processes like ER stress, cell cycle regulation, and autophagy, underscoring its substantial impact on cancer progression. Its dual role as a tumor suppressor and oncogene highlights its complexity and therapeutic potential. However, databases like the Drug Signatures Database on Enrichr reveal a lack of drugs targeting CREBRF, raising concerns about its druggability.

Further research is essential to clarify CREBRF’s context-dependent roles in different tumor types. This involves studying how CREBRF regulates ER stress, cell cycle checkpoints, and autophagy in hypoxic or nutrient-deprived microenvironments. Exploring CREBRF’s post-translational modifications and their effects on its tumor-promoting or suppressive functions may uncover new regulatory mechanisms and therapeutic opportunities.

Structural biology techniques like cryo-electron microscopy and molecular docking should be utilized to identify potential binding sites on CREBRF.80 If direct targeting of CREBRF proves difficult, its upstream regulators or downstream effectors may serve as alternative therapeutic targets. For example, targeting pathways linked to ER stress or autophagy could indirectly modulate CREBRF activity. High-throughput screening of small molecules or peptides to modulate CREBRF function may identify promising candidates for therapy.81

CREBRF’s context-dependent role, as a tumor suppressor in some cancers and an oncogene in others, highlights the need for patient stratification. Profiling CREBRF expression or functional status may facilitate precision medicine strategies. Integrating CREBRF-targeting strategies with therapies like immune checkpoint inhibitors, chemotherapy, or autophagy modulators could improve efficacy and overcome resistance. Relevant models like patient-derived organoids, xenografts, and genetically engineered mouse models are crucial for understanding CREBRF’s role in cancer and its therapeutic potential.82

Despite the challenges in directly targeting CREBRF, its pivotal role in cancer biology, especially its potential function as a tumor suppressor, underscores the importance of further research on CREBRF. A thorough understanding of CREBRF’s molecular functions, paired with innovative drug discovery and validation strategies, may unlock its potential as a cancer therapy target. Multidisciplinary collaboration will be essential to translate these insights into effective therapies.

Conclusion

CREBRF has been identified as a critical regulator in cancer biology, displaying both tumor-suppressive and tumor-promoting roles depending on the tumor type and microenvironment. These dual roles are governed by complex molecular mechanisms, emphasizing the importance of further research to elucidate CREBRF’s context-specific contributions to cancer progression. Despite its considerable therapeutic potential, CREBRF remains relatively underexplored. Direct targeting is difficult because of the lack of clearly identified druggable sites. Alternative strategies, including targeting upstream regulators, downstream effectors, or related pathways, as well as employing advanced drug discovery technologies, present promising opportunities for intervention.

The context-specific roles of CREBRF underscore the importance of precision medicine. Patient stratification based on CREBRF expression profiles or functional states could facilitate personalized therapeutic strategies. Although challenges remain, CREBRF’s crucial role in cancer biology makes it a promising target for therapeutic development. Expanding knowledge of CREBRF’s molecular functions, regulatory networks, and druggability, supported by multidisciplinary collaboration, is vital for translating these findings into clinical applications. Addressing these gaps could lead to the development of innovative and effective cancer therapies targeting CREBRF.

Acknowledgment

We would like to thank the Figdraw website for the assistance provided in image sketching.

Funding Statement

This study was supported by National Natural Science Foundation of China (Grants number: 82200214).

Data Sharing Statement

The clinical data supporting the conclusions of this manuscript will be made available by the authors.

Author Contributions

All authors made a significant contribution to the work reported, whether that is in the conception, study design, execution, acquisition of data, analysis and interpretation, or in all these areas; took part in drafting, revising or critically reviewing the article; gave final approval of the version to be published; have agreed on the journal to which the article has been submitted; and agree to be accountable for all aspects of the work.

Disclosure

The authors declare that they have no competing interests in this work.

References

  • 1.Liao H, Liu S, Ma Q, et al. Endoplasmic reticulum stress induced autophagy in cancer and its potential interactions with apoptosis and ferroptosis. Biochim Biophys Acta Mol Cell Res. 2025;1872(1):119869. [DOI] [PubMed] [Google Scholar]
  • 2.Gomez-Virgilio L, Silva-Lucero MD, Flores-Morelos DS, et al. Autophagy: a key regulator of homeostasis and disease: an overview of molecular mechanisms and modulators. Cells. 2022;11(15). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Russell RC, Guan KL. The multifaceted role of autophagy in cancer. EMBO J. 2022;41(13):e110031. doi: 10.15252/embj.2021110031 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Poillet-Perez L, Sarry JE, Joffre C. Autophagy is a major metabolic regulator involved in cancer therapy resistance. Cell Rep. 2021;36(7):109528. doi: 10.1016/j.celrep.2021.109528 [DOI] [PubMed] [Google Scholar]
  • 5.Walter P, Ron D. The unfolded protein response: from stress pathway to homeostatic regulation. Science. 2011;334(6059):1081–1086. doi: 10.1126/science.1209038 [DOI] [PubMed] [Google Scholar]
  • 6.Akman M, Belisario DC, Salaroglio IC, et al. Hypoxia, endoplasmic reticulum stress and chemoresistance: dangerous liaisons. J Exp Clin Cancer Res. 2021;40(1):28. doi: 10.1186/s13046-020-01824-3 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Mafi S, Ahmadi E, Meehan E, et al. The mTOR signaling pathway interacts with the ER stress response and the unfolded protein response in cancer. Cancer Res. 2023;83(15):2450–2460. doi: 10.1158/0008-5472.CAN-22-3032 [DOI] [PubMed] [Google Scholar]
  • 8.Chipurupalli S, Samavedam U, Robinson N. Crosstalk between ER stress, autophagy and inflammation. Front Med. 2021;8:758311. doi: 10.3389/fmed.2021.758311 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Penney J, Taylor T, MacLusky N, Lu R. LUMAN/CREB3 plays a dual role in stress responses as a cofactor of the glucocorticoid receptor and a regulator of secretion. Front Mol Neurosci. 2018;11:352. doi: 10.3389/fnmol.2018.00352 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Xue H, Yuan G, Guo X, et al. A novel tumor-promoting mechanism of IL6 and the therapeutic efficacy of tocilizumab: hypoxia-induced IL6 is a potent autophagy initiator in glioblastoma via the p-STAT3-MIR155-3p-CREBRF pathway. Autophagy. 2016;12(7):1129–1152. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Audas TE, Li Y, Liang G, Lu R. A novel protein, Luman/CREB3 recruitment factor, inhibits Luman activation of the unfolded protein response. Mol Cell Biol. 2008;28(12):3952–3966. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Xue H, Zhang JS, Guo X, et al. CREBRF is a potent tumor suppressor of glioblastoma by blocking hypoxia-induced autophagy via the CREB3/ATG5 pathway. Int J Oncol. 2016;49(2):519–528. doi: 10.3892/ijo.2016.3576 [DOI] [PubMed] [Google Scholar]
  • 13.Han JM, Zhang L, Zhang J, et al. CREBRF promotes the proliferation of human gastric cancer cells via the AKT signaling pathway. Cell Mol Biol. 2018;64(5):40–45. doi: 10.14715/cmb/2018.64.5.6 [DOI] [PubMed] [Google Scholar]
  • 14.Ma QY, Yu WW, Li ZB, Zhang XL, Zhang LH. Circ_0081723 enhances cervical cancer progression and modulates CREBRF via sponging miR-545-3p. Naunyn-Schmiedebergs Arch Pharmacol. 2024;397:8839–8852. doi: 10.1007/s00210-024-03175-8 [DOI] [PubMed] [Google Scholar]
  • 15.Feng S, Liu N, Chen XG, Liu YF, An JD. Long non-coding RNA NEAT1/miR-338-3p axis impedes the progression of acute myeloid leukemia via regulating CREBRF. Can Cell Inter. 2020;20(1). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Zeng H, Huang M, Gong X. MicroRNA-124-3p promotes apoptosis and autophagy of glioma cells by down-regulating CREBRF. Neurol Res. 2022;44(12):1094–1103. doi: 10.1080/01616412.2022.2112374 [DOI] [PubMed] [Google Scholar]
  • 17.Minster RL, Hawley NL, Su CT, et al. A thrifty variant in CREBRF strongly influences body mass index in Samoans. Nat Genet. 2016;48(9):1049–1054. doi: 10.1038/ng.3620 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Loos RJ. CREBRF variant increases obesity risk and protects against diabetes in Samoans. Nat Genet. 2016;48(9):976–978. doi: 10.1038/ng.3653 [DOI] [PubMed] [Google Scholar]
  • 19.Yuxiong W, Faping L, Bin L, et al. Regulatory mechanisms of the cAMP-responsive element binding protein 3 (CREB3) family in cancers. Biomed Pharmacother. 2023;166:115335. [DOI] [PubMed] [Google Scholar]
  • 20.Kondo S, Murakami T, Tatsumi K, et al. OASIS, a CREB/ATF-family member, modulates UPR signalling in astrocytes. Nat Cell Biol. 2005;7(2):186–194. [DOI] [PubMed] [Google Scholar]
  • 21.Bian C, Marchetti A, Dias M, Perrin J, Cosson P. Short transmembrane domains target type II proteins to the Golgi apparatus and type I proteins to the endoplasmic reticulum. J Cell Sci. 2024;137(15). doi: 10.1242/jcs.261738 [DOI] [PubMed] [Google Scholar]
  • 22.Brown MS, Ye J, Rawson RB, Goldstein JL. Regulated intramembrane proteolysis: a control mechanism conserved from bacteria to humans. Cell. 2000;100(4):391–398. doi: 10.1016/S0092-8674(00)80675-3 [DOI] [PubMed] [Google Scholar]
  • 23.Luciano RL, Wilson AC. N-terminal transcriptional activation domain of LZIP comprises two LxxLL motifs and the host cell factor-1 binding motif. Proc Natl Acad Sci U S A. 2000;97(20):10757–10762. doi: 10.1073/pnas.190062797 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Liang G, Audas TE, Li Y, et al. Luman/CREB3 induces transcription of the endoplasmic reticulum (ER) stress response protein Herp through an ER stress response element. Mol Cell Biol. 2006;26(21):7999–8010. doi: 10.1128/MCB.01046-06 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Karagoz GE, Acosta-Alvear D, Walter P. The unfolded protein response: detecting and responding to fluctuations in the protein-folding capacity of the endoplasmic reticulum. Cold Spring Harb Perspect Biol. 2019;11(9):a033886. doi: 10.1101/cshperspect.a033886 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Hemagirri M, Chen Y, Gopinath SCB, Sahreen S, Adnan M, Sasidharan S. Crosstalk between protein misfolding and endoplasmic reticulum stress during ageing and their role in age-related disorders. Biochimie. 2024;221:159–181. doi: 10.1016/j.biochi.2023.10.019 [DOI] [PubMed] [Google Scholar]
  • 27.Ron D, Walter P. Signal integration in the endoplasmic reticulum unfolded protein response. Nat Rev Mol Cell Biol. 2007;8(7):519–529. doi: 10.1038/nrm2199 [DOI] [PubMed] [Google Scholar]
  • 28.Wang J, Kang R, Huang H, et al. Hepatitis C virus core protein activates autophagy through EIF2AK3 and ATF6 UPR pathway-mediated MAP1LC3B and ATG12 expression. Autophagy. 2014;10(5):766–784. doi: 10.4161/auto.27954 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Lu R, Yang P, O’Hare P, Misra V. Luman, a new member of the CREB/ATF family, binds to herpes simplex virus VP16-associated host cellular factor. Mol Cell Biol. 1997;17(9):5117–5126. doi: 10.1128/MCB.17.9.5117 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Oh-Hashi K, Hasegawa T, Naruse Y, Hirata Y. Molecular characterization of mouse CREB3 regulatory factor in Neuro2a cells. Mol Biol Rep. 2021;48(7):5411–5420. doi: 10.1007/s11033-021-06543-2 [DOI] [PubMed] [Google Scholar]
  • 31.Yam CQX, Lim HH, Surana U. DNA damage checkpoint execution and the rules of its disengagement. Front Cell Dev Biol. 2022;10:1020643. doi: 10.3389/fcell.2022.1020643 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Baserga R. The relationship of the cell cycle to tumor growth and control of cell division: a review. Cancer Res. 1965;25:581–595. [PubMed] [Google Scholar]
  • 33.Harper JW, Elledge SJ. The DNA damage response: ten years after. Mol Cell. 2007;28(5):739–745. doi: 10.1016/j.molcel.2007.11.015 [DOI] [PubMed] [Google Scholar]
  • 34.Oropeza E, Seker S, Carrel S, et al. Molecular portraits of cell cycle checkpoint kinases in cancer evolution, progression, and treatment responsiveness. Sci Adv. 2023;9(26):eadf2860. doi: 10.1126/sciadv.adf2860 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Sotillo R, Dubus P, Martin J, et al. Wide spectrum of tumors in knock-in mice carrying a Cdk4 protein insensitive to INK4 inhibitors. EMBO J. 2001;20(23):6637–6647. doi: 10.1093/emboj/20.23.6637 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Lobrich M, Jeggo PA. The impact of a negligent G2/M checkpoint on genomic instability and cancer induction. Nat Rev Cancer. 2007;7(11):861–869. doi: 10.1038/nrc2248 [DOI] [PubMed] [Google Scholar]
  • 37.Dey SK, Lim H, Das SK, et al. Molecular cues to implantation. Endocr Rev. 2004;25(3):341–373. doi: 10.1210/er.2003-0020 [DOI] [PubMed] [Google Scholar]
  • 38.Li X, Lin P, Chen F, et al. Luman recruiting factor is involved in stromal cell proliferation during decidualization in mice. Cell Tissue Res. 2016;365:437–447. doi: 10.1007/s00441-016-2392-z [DOI] [PubMed] [Google Scholar]
  • 39.Zhang P, Cheng S, Sheng X, Dai H, He K, Du Y. The role of autophagy in regulating metabolism in the tumor microenvironment. Genes Dis. 2023;10(2):447–456. doi: 10.1016/j.gendis.2021.10.010 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Zaarour RF, Azakir B, Hajam EY, et al. Role of hypoxia-mediated autophagy in tumor cell death and survival. Cancers. 2021;13(3):533. doi: 10.3390/cancers13030533 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Gewirtz DA. The four faces of autophagy: implications for cancer therapy. Cancer Res. 2014;74(3):647–651. doi: 10.1158/0008-5472.CAN-13-2966 [DOI] [PubMed] [Google Scholar]
  • 42.Guo JY, Xia B, White E. Autophagy-mediated tumor promotion. Cell. 2013;155(6):1216–1219. doi: 10.1016/j.cell.2013.11.019 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Mascia F, Mazo I, Alterovitz W-L, et al. In search of autophagy biomarkers in breast cancer: receptor status and drug agnostic transcriptional changes during autophagy flux in cell lines. PLoS One. 2022;17(1):e0262134. doi: 10.1371/journal.pone.0262134 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Ramachandran R, Ibragimova S, Woods LM, et al. Conserved role of FOXC1 in TNBC is parallel to FOXA1 in ER+ breast cancer. iScience. 2024;27(8):110500. doi: 10.1016/j.isci.2024.110500 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Yang D, Jiang T, Liu J, et al. CREB3 regulatory factor -mTOR-autophagy regulates goat endometrial function during early pregnancy. Biol Reprod. 2018;98(5):713–721. doi: 10.1093/biolre/ioy044 [DOI] [PubMed] [Google Scholar]
  • 46.Davies H, Bignell GR, Cox C, et al. Mutations of the BRAF gene in human cancer. Nature. 2002;417(6892):949–954. doi: 10.1038/nature00766 [DOI] [PubMed] [Google Scholar]
  • 47.Kim S, Carvajal R, Kim M, Yang HW. Kinetics of RTK activation determine ERK reactivation and resistance to dual BRAF/MEK inhibition in melanoma. Cell Rep. 2023;42(6):112570. doi: 10.1016/j.celrep.2023.112570 [DOI] [PubMed] [Google Scholar]
  • 48.Liu YL, Lai F, Wilmott JS, et al. Noxa upregulation by oncogenic activation of MEK/ERK through CREB promotes autophagy in human melanoma cells. Oncotarget. 2014;5(22):11237–11251. doi: 10.18632/oncotarget.2616 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Emami Nejad A, Najafgholian S, Rostami A, et al. The role of hypoxia in the tumor microenvironment and development of cancer stem cell: a novel approach to developing treatment. Cancer Cell Int. 2021;21(1):62. doi: 10.1186/s12935-020-01719-5 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Shi R, Liao C, Zhang Q. Hypoxia-driven effects in cancer: characterization, mechanisms, and therapeutic implications. Cells. 2021;10(3):678. doi: 10.3390/cells10030678 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Brown JM, Wilson WR. Exploiting tumour hypoxia in cancer treatment. Nat Rev Cancer. 2004;4(6):437–447. doi: 10.1038/nrc1367 [DOI] [PubMed] [Google Scholar]
  • 52.Hu YL, Jahangiri A, De Lay M, Aghi MK. Hypoxia-induced tumor cell autophagy mediates resistance to anti-angiogenic therapy. Autophagy. 2012;8(6):979–981. doi: 10.4161/auto.20232 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Yang YZ, Jin YP, Martyn AC, et al. Expression pattern implicates a potential role for luman recruitment factor in the process of implantation in uteri and development of preimplantation embryos in mice. J Reprod Dev. 2013;59(3):245–251. doi: 10.1262/jrd.2012-137 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Martyn AC, Choleris E, Gillis DJ, et al. Luman/CREB3 recruitment factor regulates glucocorticoid receptor activity and is essential for prolactin-mediated maternal instinct. Mol Cell Biol. 2012;32(24):5140–5150. doi: 10.1128/MCB.01142-12 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Wu ZH, Li FF, Ruan LL, et al. miR-181d-5p, which is upregulated in fetal growth restriction placentas, inhibits trophoblast fusion via CREBRF. J Assist Reprod Genet. 2023;40(11):2725–2737. doi: 10.1007/s10815-023-02917-6 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Saavedra P, Dumesic PA, Hu Y, et al. REPTOR and CREBRF encode key regulators of muscle energy metabolism. Nat Commun. 2023;14(1):4943. doi: 10.1038/s41467-023-40595-1 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Zhang C, Zhang F, Li Y, Yang P, Liu Y, Yang W. The circular RNA Circ_0043947 promoted gastric cancer progression by sponging miR-384 to regulate CREB1 expression. Gut Liver. 2024;18(6):977–991. doi: 10.5009/gnl230173 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Vivanco I, Sawyers CL. The phosphatidylinositol 3-Kinase AKT pathway in human cancer. Nat Rev Cancer. 2002;2(7):489–501. doi: 10.1038/nrc839 [DOI] [PubMed] [Google Scholar]
  • 59.Manning BD, Cantley LC. AKT/PKB signaling: navigating downstream. Cell. 2007;129(7):1261–1274. doi: 10.1016/j.cell.2007.06.009 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Hao CL, Han JJ, Xiang KC, et al. CircVIRMA enhances cell malignant behavior by governing the miR-452-5p/CREBRF pathway in cervical cancer. Naunyn-Schmiedebergs Arch Pharmacol. 2024;397:8825–8838. doi: 10.1007/s00210-024-03159-8 [DOI] [PubMed] [Google Scholar]
  • 61.Shibata T, Cao DY, Dar TB, et al. miR766-3p and miR124-3p dictate drug resistance and clinical outcome in HNSCC. Cancers. 2022;14(21):5273. doi: 10.3390/cancers14215273 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Qu XD, Tao JZ, Xie JR. Circ_0009035 regulates the progression of cervical cancer by targeting miR-1305/CREBRF axis. Anti-Cancer Drugs. 2022;33(6):539–552. doi: 10.1097/CAD.0000000000001278 [DOI] [PubMed] [Google Scholar]
  • 63.Tang X, Wen XM, Li ZY, et al. Hsa_circ_0102171 aggravates the progression of cervical cancer through targeting miR-4465/CREBRF axis. J Cell Physiol. 2021;236(7):4973–4984. doi: 10.1002/jcp.30210 [DOI] [PubMed] [Google Scholar]
  • 64.Han F, Zhong C, Li W, et al. hsa_circ_0001947 suppresses acute myeloid leukemia progression via targeting hsa-miR-329-5p/CREBRF axis. Epigenomics. 2020;12(11):935–953. doi: 10.2217/epi-2019-0352 [DOI] [PubMed] [Google Scholar]
  • 65.Wu KR, Huang J, Xu T, et al. MicroRNA-181b blocks gensenoside Rg3-mediated tumor suppression of gallbladder carcinoma by promoting autophagy flux via CREBRF/CREB3 pathway. Am J Transl Res. 2019;11(9):5776–5787. [PMC free article] [PubMed] [Google Scholar]
  • 66.Cossu Rocca M, Lorini L, Szturz P, Bossi P, Vermorken JB. Recurrent/metastatic head and neck squamous cell carcinoma in older patients: are new agents bringing new hope? Drugs Aging. 2023;40(2):135–143. doi: 10.1007/s40266-022-01000-6 [DOI] [PubMed] [Google Scholar]
  • 67.Dohner H, Weisdorf DJ, Bloomfield CD. Acute myeloid leukemia. N Engl J Med. 2015;373(12):1136–1152. doi: 10.1056/NEJMra1406184 [DOI] [PubMed] [Google Scholar]
  • 68.Entezari M, Taheriazam A, Orouei S, et al. LncRNA-miRNA axis in tumor progression and therapy response: an emphasis on molecular interactions and therapeutic interventions. Biomed Pharmacother. 2022;154:113609. doi: 10.1016/j.biopha.2022.113609 [DOI] [PubMed] [Google Scholar]
  • 69.Yao FY, Zhao C, Zhong FM, et al. m(6)A modification of lncRNA NEAT1 regulates chronic myelocytic leukemia progression via miR-766-5p/CDKN1A axis. Front Oncol. 2021;11:679634. doi: 10.3389/fonc.2021.679634 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Yu K, Wang J, Hou J, Zhang L, Liang H. miR-338-3p plays a significant role in casticin-induced suppression of acute myeloid leukemia via targeting PI3K/Akt pathway. Biomed Res Int. 2022;2022:9214130. doi: 10.1155/2022/9214130 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Yang Y, Tu Z, Ye C, et al. Site-specific metastases of gallbladder adenocarcinoma and their prognostic value for survival: a SEER-based study. BMC Surg. 2021;21(1):59. doi: 10.1186/s12893-021-01068-8 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Jiang Y, Qiu Q, Jing X, et al. Cancer-associated fibroblast-derived exosome miR-181b-3p promotes the occurrence and development of colorectal cancer by regulating SNX2 expression. Biochem Biophys Res Commun. 2023;641:177–185. doi: 10.1016/j.bbrc.2022.12.026 [DOI] [PubMed] [Google Scholar]
  • 73.Li B, Tao Q, Hu X, Li T, Bao Y. [miR-181b-5p promotes cell proliferation and induces apoptosis in human acute myeloid leukemia by targeting PAX9]. Xi Bao Yu Fen Zi Mian Yi Xue Za Zhi. 2023;39(12):1074–1082. Wolof [PubMed] [Google Scholar]
  • 74.Maryam M, Naemi M, Hasani SS. A comprehensive review on oncogenic miRNAs in breast cancer. J Genet. 2021;100. [PubMed] [Google Scholar]
  • 75.Tang YC, Zhang Y, Zhou J, et al. Ginsenoside Rg3 targets cancer stem cells and tumor angiogenesis to inhibit colorectal cancer progression in vivo. Int J Oncol. 2018;52(1):127–138. doi: 10.3892/ijo.2017.4183 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.Liu Y, Li G, Ning J, Zhao Y. Unveiling the experimental proof of the anticancer potential of ginsenoside Rg3 (Review). Oncol Lett. 2024;27(4):182. doi: 10.3892/ol.2024.14315 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.Gusyatiner O, Hegi ME. Glioma epigenetics: from subclassification to novel treatment options. Semin Cancer Biol. 2018;51:50–58. doi: 10.1016/j.semcancer.2017.11.010 [DOI] [PubMed] [Google Scholar]
  • 78.Siegel RL, Miller KD, Jemal A. Cancer statistics, 2016. CA Cancer J Clin. 2016;66(1):7–30. doi: 10.3322/caac.21332 [DOI] [PubMed] [Google Scholar]
  • 79.van den Heuvel S, Harlow E. Distinct roles for cyclin-dependent kinases in cell cycle control. Science. 1993;262(5142):2050–2054. doi: 10.1126/science.8266103 [DOI] [PubMed] [Google Scholar]
  • 80.Duan J, He XH, Li SJ, Xu HE. Cryo-electron microscopy for GPCR research and drug discovery in endocrinology and metabolism. Nat Rev Endocrinol. 2024;20(6):349–365. doi: 10.1038/s41574-024-00957-1 [DOI] [PubMed] [Google Scholar]
  • 81.Blay V, Tolani B, Ho SP, Arkin MR. High-throughput screening: today’s biochemical and cell-based approaches. Drug Discovery Today. 2020;25(10):1807–1821. doi: 10.1016/j.drudis.2020.07.024 [DOI] [PubMed] [Google Scholar]
  • 82.Herpers B, Eppink B, James MI, et al. Functional patient-derived organoid screenings identify MCLA-158 as a therapeutic EGFR × LGR5 bispecific antibody with efficacy in epithelial tumors. Nat Cancer. 2022;3(4):418–436. doi: 10.1038/s43018-022-00359-0 [DOI] [PubMed] [Google Scholar]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Data Availability Statement

The clinical data supporting the conclusions of this manuscript will be made available by the authors.


Articles from Biologics : Targets & Therapy are provided here courtesy of Dove Press

RESOURCES