Molecular Mechanisms of the CIRBP Family in Tumors: Current Status and Future Perspectives

Abstract

The Cold-Inducible RNA-Binding Protein (CIRBP) family plays a pivotal role in cellular stress responses and tumorigenesis. Recent studies have increasingly highlighted the expression alterations of CIRBP family members across various cancer types and their potential molecular mechanisms. This review provides a comprehensive overview of the structural characteristics and functions of the CIRBP family, alongside their expression profiles in tumors and the regulatory molecular mechanisms involved. By synthesizing current knowledge, this review aims to offer new insights and directions for future cancer therapies, emphasizing the importance of CIRBP proteins in oncological research.

Share and Cite:

Cai, Y. , Wang, T. and Zhan, Y. (2025) Molecular Mechanisms of the CIRBP Family in Tumors: Current Status and Future Perspectives. Journal of Cancer Therapy, 16, 63-76. doi: 10.4236/jct.2025.162006.

1. Introduction

The CIRBP (Cold-Inducible RNA-Binding Protein) family is a group of RNA-binding proteins that play significant roles in cellular stress responses, particularly in the context of cold shock and other environmental stressors. These proteins are crucial in regulating various cellular processes, including mRNA stability, translation, and cellular proliferation. CIRBP proteins are known to interact with RNA in a sequence-specific manner, influencing gene expression patterns that are vital for cell survival under stress conditions. They are implicated in numerous biological processes, including cellular differentiation, apoptosis, and inflammation, highlighting their importance in cellular biology [1]. Understanding the molecular mechanisms by which CIRBP family members operate is essential for elucidating their roles in tumor biology and potential therapeutic applications.

The relationship between tumorigenesis and cellular stress responses is a complex and multifaceted area of research. Tumor cells often encounter various forms of stress, including oxidative stress, hypoxia, and nutrient deprivation, which can influence their growth and survival. The CIRBP family plays a pivotal role in mediating cellular responses to these stressors, thereby impacting tumor progression and metastasis. As tumors grow, they create a unique microenvironment characterized by altered pH, oxygen levels, and nutrient availability, which can induce stress responses in cancer cells. The ability of CIRBP proteins to modulate these responses may contribute to the adaptation and survival of tumor cells in adverse conditions [2]. Studies have shown that circ_0007386, a circular RNA, targets miR-383-5p to influence CIRBP, ultimately affecting the proliferation and apoptosis of Non-Small Cell Lung Cancer (NSCLC) cells through the PI3K/AKT signaling pathway [3]. CIRBP also plays an important role in nasopharyngeal carcinoma, especially in terms of hyperthermia resistance [4].

Research into the CIRBP family within the context of cancer has revealed their potential as biomarkers and therapeutic targets. For instance, the expression levels of CIRBP proteins have been associated with various types of malignancies, and their modulation can influence tumor cell behavior, including proliferation, migration, and apoptosis resistance. This suggests that CIRBP proteins could serve as promising targets for developing novel cancer therapies aimed at restoring normal stress responses in tumor cells [5]. Moreover, insights into the molecular pathways regulated by CIRBP proteins can provide a deeper understanding of tumor biology, paving the way for innovative diagnostic and therapeutic strategies in oncology [6].

In summary, the CIRBP family represents a critical component of the cellular machinery that responds to stress, with significant implications for tumor biology. As research continues to uncover the intricate roles of these proteins in cancer, their potential applications in diagnosis and treatment are becoming increasingly apparent. Future studies that explore the specific mechanisms by which CIRBP proteins influence tumorigenesis will be essential for harnessing their therapeutic potential and improving outcomes for cancer patients.

2. Main Body

2.1. Structure and Function of the CIRBP Family

The cold-inducible RNA-binding protein (CIRBP) family consists of proteins that play critical roles in cellular stress responses and the regulation of gene expression. CIRBP is characterized by its ability to bind RNA and is involved in various biological processes, including cell proliferation, apoptosis, and inflammation. The family is primarily composed of CIRBP and its homologs, which share structural features such as RNA recognition motifs and RG/RGG-rich domains. These domains are essential for the protein’s function in RNA binding and the formation of biomolecular condensates. The CIRBP family is particularly important in vertebrates, where it is activated under stress conditions like cold exposure, hypoxia, and heat shock, allowing cells to adapt and survive in adverse environments. The structural diversity among CIRBP family members suggests that they may have evolved to perform distinct yet overlapping functions in cellular stress responses and the maintenance of cellular homeostasis [7].

2.1.1. Classification of CIRBP Family Members

CIRBP family members can be classified based on their structural characteristics and functional roles. The primary member, CIRBP, is known for its involvement in cold shock responses, while other homologs, such as CIRBP-A and CIRBP-B, exhibit similar RNA-binding capabilities but may differ in their specific regulatory functions. These proteins are often categorized into two main groups: those primarily involved in stress response and those that participate in developmental processes. For instance, CIRBP is heavily implicated in spermatogenesis and the regulation of cell cycle progression, particularly under heat stress conditions. Other family members may have specialized roles in different tissues or developmental stages, indicating a complex interplay between structure and function within the CIRBP family. Understanding these classifications can provide insights into the specific biological pathways influenced by each member and their potential therapeutic targets in diseases such as cancer and neurodegeneration [8].

2.1.2. Major Biological Functions of CIRBP

CIRBP serves several critical biological functions that are essential for cellular adaptation to stress. One of its primary roles is the regulation of mRNA stability and translation during stress conditions. CIRBP binds to target mRNAs, influencing their degradation and translation efficiency, which is crucial for the timely expression of stress-responsive genes. This regulation is particularly important in contexts such as cancer, where CIRBP has been shown to promote cell proliferation and survival by modulating the expression of oncogenes and tumor suppressors. Additionally, CIRBP is involved in the cellular response to hypoxia and heat stress, where it helps maintain cellular homeostasis by coordinating the expression of proteins that protect against damage. The ability of CIRBP to interact with various cellular pathways underscores its importance as a multifunctional protein that integrates signals from the environment to regulate essential biological processes [9].

CIRBP is involved in numerous processes such as cell growth, proliferation, development, and differentiation. Cell proliferation is closely related to the activation of various signaling pathways, and CIRBP plays an important role in these pathways, such as ERK/MAPK, PI3K/AKT, Wnt, and NF-κB [10] [11], and promotes the transition to the next stage of the cell proliferation cycle. Firstly, CIRBP can enhance the activation of the ERK pathway, leading to the formation of phosphorylated ERK1/2 (P-ERK1/2). P-ERK1/2 promotes cell proliferation through a series of downstream signaling molecules, for example, by facilitating the transition from the G1 (first gap/DNA synthesis preparation) to S (DNA synthesis) phase in normal somatic and germline cells [12].

Secondly, CIRBP can promote cell proliferation and differentiation by regulating the Wnt signaling pathway. It enhances the activation of the Wnt pathway by binding to TCF-3 mRNA and increasing its stability, reducing its degradation, thereby regulating the development and growth of organs and tissues such as the nervous system. Finally, CIRBP also regulates the growth, proliferation, and differentiation of cells in organs and tissues by regulating certain target mRNAs. For instance, by binding to the mRNA of the Wilms’ tumor 1 (WT1) gene, CIRBP regulates the formation and development of the kidney in Xenopus laevis embryos [13], or by binding to adhesion molecule genes such as C- and E-cadherin, and αE- and β-catenin, the transcribed mRNAs promote the development and migration of embryonic cells [14]. According to current research findings, CIRBP primarily regulates cell growth and proliferation through the ERK/MAPK and Wnt pathways. Additionally, it exerts specific regulatory effects through the regulation of individual target genes.

2.1.3. Mechanisms of CIRBP in Cellular Stress Responses

CIRBP plays a pivotal role in mediating cellular responses to various stressors through multiple mechanisms. Upon exposure to stress, CIRBP undergoes post-translational modifications, such as phosphorylation and methylation, which can alter its localization and activity. For instance, these modifications enable CIRBP to translocate from the nucleus to the cytoplasm, where it can interact with stress granules and regulate mRNA dynamics. Furthermore, CIRBP’s ability to form biomolecular condensates allows it to concentrate and coordinate the response to stress, facilitating the efficient processing of target mRNAs. In the context of heat stress, CIRBP has been shown to regulate the expression of key cell cycle proteins, such as cyclin B1, thereby influencing cell cycle progression and apoptosis in spermatogenic cells. This regulatory capacity is critical for protecting cells from damage and ensuring proper recovery following stress exposure. Overall, the mechanisms by which CIRBP operates highlight its central role in maintaining cellular integrity and function under adverse conditions [7].

2.2. CIRBP Expression Changes in Tumors

The cold-inducible RNA-binding protein (CIRBP) family has emerged as a significant player in the context of various tumors, with its expression levels showing notable variations across different cancer types. CIRBP is known to be involved in the regulation of mRNA stability and translation, particularly under stress conditions. In various studies, CIRBP expression has been found to differ significantly between tumor tissues and adjacent normal tissues, indicating its potential role as a biomarker for tumorigenesis. For instance, in pancreatic cancer, lower levels of CIRBP were associated with poorer survival outcomes, suggesting that high expression of CIRBP could be protective against tumor progression [15]. Similarly, in hepatocellular carcinoma (HCC), CIRBP was identified as part of a prognostic model that correlated high expression with improved overall survival rates [16]. The differential expression of CIRBP across tumor types underscores its potential as a therapeutic target and a prognostic marker, emphasizing the need for further research to elucidate its precise roles in cancer biology.

2.2.1. CIRBP Expression Patterns in Different Tumor Types

CIRBP expression patterns vary significantly across different tumor types, reflecting its multifaceted role in cancer biology. In endometrial cancer, for example, low CIRBP expression has been linked to increased cell proliferation and a worse prognosis, highlighting its potential as a prognostic biomarker [17]. In contrast, pancreatic cancer studies have shown that high nuclear expression of CIRBP correlates with longer survival, indicating a protective role against tumor growth [15]. Furthermore, in hepatocellular carcinoma, CIRBP was identified as a critical factor in a prognostic model, where its expression was associated with advanced disease stages and poor outcomes [16]. These findings suggest that CIRBP may exert context-dependent effects, acting as a tumor suppressor in some cancers while potentially promoting malignancy in others. The complexity of CIRBP’s role in different tumor microenvironments necessitates comprehensive studies to unravel the underlying mechanisms that dictate its expression and function in various cancers.

2.2.2. Correlation Studies of CIRBP with Tumor Stage and Prognosis

Research has increasingly focused on the correlation between CIRBP expression and tumor stage as well as patient prognosis. In endometrial cancer, a study constructed a nomogram incorporating CIRBP expression alongside clinical factors, demonstrating that low CIRBP levels were significantly associated with advanced tumor stages and poorer overall survival [18]. This correlation suggests that CIRBP could serve as a valuable prognostic marker in clinical settings. Similarly, in hepatocellular carcinoma, high CIRBP expression was linked to better survival outcomes and lower tumor mutational burden, indicating its potential as an independent prognostic factor [17]. These studies collectively highlight the importance of CIRBP in assessing tumor progression and patient prognosis, suggesting that it could be integrated into clinical practice to enhance personalized treatment strategies.

2.2.3. Regulatory Mechanisms of CIRBP Expression

The expression of CIRBP is tightly regulated through various mechanisms, which can significantly influence its role in tumor biology. CIRBP undergoes transcriptional regulation influenced by environmental stressors such as hypoxia and heat stress, which can induce its expression [19]. Understanding the regulatory mechanisms governing CIRBP expression and function is crucial for harnessing its potential in clinical applications. CIRBP is regulated at multiple levels, including transcriptional, post-transcriptional, and post-translational modifications. Transcriptionally, CIRBP expression is influenced by various stressors, including cold shock, hypoxia, and oxidative stress, which activate specific signaling pathways that enhance its mRNA stability and translation [5]. Post-transcriptionally, CIRBP interacts with a range of RNA-binding proteins and microRNAs, modulating the stability and translation of its target mRNAs, which are often involved in critical cellular processes such as apoptosis and inflammation [8]. Additionally, alternative splicing events can lead to different CIRBP isoforms, potentially altering its functional roles in various cellular contexts [7]. Post-translational modifications, such as phosphorylation and methylation, further influence CIRBP’s localization and activity, dictating its participation in stress granule formation and other cellular responses. Moreover, influencing its interactions with target mRNAs involved in key cellular processes like apoptosis and cell proliferation [7]. Understanding these regulatory mechanisms is essential for elucidating how CIRBP contributes to tumorigenesis and could provide insights into novel therapeutic approaches targeting its expression and function in cancer.

2.3. CIRBP Molecular Mechanism Research

The Cold-Inducible RNA Binding Protein (CIRBP) family has garnered attention for its multifaceted roles in various biological processes, particularly in cancer biology. Understanding the molecular mechanisms by which CIRBP influences tumor progression is critical for developing novel therapeutic strategies. CIRBP is known to be involved in cellular responses to cold stress, but its implications in cancer cell proliferation, migration, invasion, and interactions with the tumor microenvironment reveal a more complex role. This section will delve into the specific functions of CIRBP in tumor biology, emphasizing its relationship with cancer cell proliferation, migration, invasion, and its interactions within the tumor microenvironment.

2.3.1. The Relationship between CIRBP and Tumor Cell Proliferation

CIRBP has been implicated in the modulation of tumor cell proliferation across various cancer types. In pancreatic ductal adenocarcinoma (PDAC), studies have shown that CIRBP expression is significantly upregulated in tumor tissues compared to adjacent normal tissues, correlating with poor prognosis [14]. Knockdown of CIRBP in PDAC cell lines, such as PANC-1 and SW1990, resulted in a marked decrease in cell proliferation, migration, and invasion in vitro, as well as reduced tumor growth in vivo. This indicates that CIRBP plays a pivotal role in promoting the malignant characteristics of PDAC cells. Furthermore, the mechanism underlying this effect involves the upregulation of dual-specificity tyrosine-Y-phosphorylation regulated kinase 1B (DYRK1B), which is associated with enhanced chemoresistance to gemcitabine treatment. The inhibition of CIRBP not only diminishes DYRK1B expression but also alters the ERK/p38 signaling pathway, further highlighting the significance of CIRBP in regulating tumor cell proliferation and treatment response [16]. Additionally, in non-small cell lung cancer (NSCLC), the circRNA circ_0007386 has been identified as a regulator of CIRBP, suggesting that CIRBP’s role in proliferation may be influenced by complex regulatory networks involving non-coding RNAs [3]. Overall, these findings underscore the critical function of CIRBP in tumor cell proliferation, making it a potential target for therapeutic intervention.

2.3.2. The Role of CIRBP in Tumor Cell Migration and Invasion

CIRBP not only influences tumor cell proliferation but also plays a crucial role in the migration and invasion of cancer cells. In the context of nasopharyngeal carcinoma (NPC), CIRBP expression has been shown to affect the sensitivity of cancer cells to hyperthermia, a treatment modality that can induce cell death. Specifically, the inhibition of CIRBP enhances the anti-tumor effects of hyperthermia, indicating that CIRBP overexpression may confer resistance to this treatment [20]. This resistance is linked to the regulation of cancer stem-like cells, which are associated with increased migratory and invasive capabilities. Moreover, in gastric cancer, CIRBP expression levels have been correlated with the tumor microenvironment, where it may influence the behavior of cancer cells in response to various stimuli [21]. The interplay between CIRBP and the tumor microenvironment is critical, as it can modulate the invasive potential of cancer cells through pathways that regulate cell adhesion, migration, and the epithelial-mesenchymal transition (EMT). Thus, CIRBP serves as a significant player in the mechanisms underlying tumor cell migration and invasion, presenting opportunities for targeted therapies aimed at disrupting these processes.

2.3.3. The Interaction of CIRBP with the Tumor Microenvironment

The tumor microenvironment (TME) is a complex ecosystem that significantly influences cancer progression and therapeutic response. CIRBP has been shown to interact with various components of the TME, including immune cells, stromal cells, and extracellular matrix components. In gastric cancer, single-cell RNA sequencing has revealed that CIRBP expression varies among different cell populations within the TME, suggesting its role in modulating the immune landscape and cellular interactions [14]. Furthermore, studies indicate that CIRBP may regulate the expression of genes involved in ferroptosis, a form of regulated cell death that can affect tumor growth and response to therapy. By influencing the expression of key proteins within the TME, CIRBP can alter the dynamics of immune cell infiltration and the overall tumorigenic process. Additionally, the interaction of CIRBP with signaling pathways, such as the PI3K/AKT pathway, further emphasizes its role in shaping the TME and its impact on cancer cell behavior [3]. Understanding these interactions provides valuable insights into the potential of CIRBP as a therapeutic target, as modulating its activity could enhance the efficacy of existing treatments by altering the TME to favor anti-tumor responses.

2.4. Potential Applications of the CIRBP Family in Cancer Treatment

The Cold-Inducible RNA Binding Protein (CIRBP) family has emerged as a significant player in the landscape of cancer biology and treatment. As research progresses, the potential applications of CIRBP in cancer therapy are increasingly recognized, particularly in its role as a biomarker, a therapeutic target, and in personalized treatment strategies. The multifunctional nature of CIRBP, which is involved in cellular stress responses, apoptosis, and gene expression regulation, positions it as a promising candidate for enhancing cancer treatment efficacy and patient outcomes.

Research indicates that in specific cancers, including non-small cell lung cancer and nasopharyngeal carcinoma, CIRBP expression is closely linked to cancer cell proliferation, apoptosis, and treatment resistance. Scientists have found that regulating CIRBP expression affects the growth and survival of cancer cells, which supports the development of targeted therapies. Additionally, studies have explored the relationship between CIRBP and the PI3K/AKT signaling pathway, which plays a crucial role in cancer progression. Interfering with the interaction between CIRBP and these pathways may lead to the development of new strategies for more effective cancer treatment [22] [23]. Current research on CIRBP as a therapeutic target is still in its early stages, necessitating more experimental evidence and clinical studies to verify its feasibility and safety.

2.4.1. Prospects of CIRBP as a Tumor Biomarker

CIRBP has garnered attention as a potential biomarker in various malignancies due to its differential expression patterns associated with tumor progression and response to treatment. Recent studies have indicated that CIRBP levels correlate with the aggressiveness of tumors and patient prognosis, making it a valuable tool for early detection and monitoring of cancer. For instance, in gastric cancer, the expression of CIRBP has been linked to the tumor microenvironment and cellular heterogeneity, suggesting that it may serve as a prognostic marker for disease outcomes [23]. Furthermore, the ability of CIRBP to modulate cellular responses to stress and its involvement in processes such as ferroptosis highlight its relevance in understanding tumor biology. As a biomarker, CIRBP could facilitate the stratification of patients based on their likelihood of responding to specific therapies, thereby guiding treatment decisions and improving clinical management strategies.

2.4.2. Targeting CIRBP in Therapeutic Strategies

The therapeutic potential of targeting CIRBP is being explored in various cancer types, particularly in enhancing the efficacy of existing treatments. Research has shown that CIRBP plays a critical role in mediating cellular responses to stress and can influence the effectiveness of therapies such as radiotherapy. For example, in lung cancer models, inhibition of CIRBP has been associated with reduced radioresistance, suggesting that targeting CIRBP could sensitize tumors to radiation therapy [24]. Additionally, CIRBP’s involvement in mitochondrial autophagy presents another avenue for therapeutic intervention, as manipulating its expression could alter cancer cell survival and treatment resistance. By developing drugs or strategies that specifically target CIRBP, there is potential to improve treatment outcomes and overcome challenges associated with therapy resistance in cancer patients.

2.4.3. Applications of CIRBP in Personalized Treatment

The integration of CIRBP into personalized cancer treatment approaches holds significant promise for enhancing patient outcomes. Given its role as a biomarker and therapeutic target, CIRBP can be utilized to tailor treatment regimens based on individual patient profiles. For instance, patients exhibiting high levels of CIRBP may benefit from specific therapeutic strategies that target this protein, potentially leading to improved responses to treatment. Moreover, the ability to assess CIRBP expression levels could aid in predicting patient responses to various therapies, allowing for more informed decisions regarding treatment plans. Personalized treatment strategies that incorporate CIRBP could ultimately lead to more effective and targeted interventions, reducing the likelihood of adverse effects and improving overall survival rates in cancer patients. As research continues to elucidate the mechanisms by which CIRBP influences cancer biology, its role in personalized medicine is likely to expand, paving the way for more innovative and effective cancer therapies.

2.5. Future Research Directions and Challenges

The study of the Cold-Inducible RNA-Binding Protein (CIRBP) family has gained significant attention in recent years due to its involvement in various cellular processes, including stress responses, cell proliferation, and differentiation. However, despite the progress made, there remain considerable gaps in our understanding of the CIRBP family. Current research primarily focuses on the molecular mechanisms of CIRBP in response to cold shock and its role in regulating gene expression. Yet, the functional diversity of CIRBP family members and their interactions with other cellular proteins are not fully elucidated. Furthermore, the implications of CIRBP in various diseases, such as cancer and neurodegenerative disorders, are still underexplored, highlighting a critical need for comprehensive studies that address these gaps. The limitations in existing research methodologies, including the lack of advanced imaging techniques and the need for more robust in vivo models, also pose challenges to the field. As such, future research must prioritize these areas to enhance our understanding of CIRBP’s multifaceted roles in health and disease.

2.5.1. Current Status and Limitations of CIRBP Family Research CIRBP

The current state of research on the CIRBP family reveals both advancements and limitations. Although significant strides have been made in identifying the biological functions of CIRBP, particularly in stress response mechanisms, there is a notable deficiency in understanding the full spectrum of its biological roles. Most studies have concentrated on the well-characterized members of the CIRBP family, leaving other potential members relatively unexplored. Moreover, the existing literature often lacks comprehensive analyses that integrate findings across different biological contexts, which could provide insights into the functional redundancy and specificity of CIRBP family members. Additionally, the methodologies employed in CIRBP research often do not account for the complexity of cellular environments, leading to oversimplified interpretations of data. This gap in research underscores the necessity for a more holistic approach that encompasses various experimental models and techniques to unravel the intricate biological functions of the CIRBP family.

2.5.2. Key Areas for Future Research

Future research on the CIRBP family should focus on several key areas to advance our understanding of its biological significance. Firstly, investigating the role of CIRBP in various disease contexts, particularly in cancer and neurodegenerative diseases, is crucial. Understanding how CIRBP contributes to tumorigenesis or neurodegeneration could unveil novel therapeutic targets. Secondly, exploring the post-translational modifications of CIRBP proteins and their impact on function and stability will provide insights into the regulatory mechanisms governing CIRBP activity. Furthermore, the development of advanced in vivo models that accurately reflect human disease conditions will enhance the translational relevance of CIRBP research. Lastly, a multi-omics approach integrating genomics, proteomics, and metabolomics could offer a comprehensive view of CIRBP’s role in cellular networks, facilitating the identification of novel pathways and interactions. By addressing these areas, future studies can significantly contribute to the field and potentially lead to innovative therapeutic strategies.

2.5.3. Challenges in Clinical Translational Research

The translation of CIRBP research into clinical applications faces several challenges that need to be addressed to realize its potential in therapeutic settings. One of the primary obstacles is the gap between basic research findings and clinical implementation. Many promising discoveries regarding CIRBP’s role in disease mechanisms have yet to be validated in clinical cohorts, which is essential for establishing their relevance in patient care. Additionally, the heterogeneity of diseases, particularly in cancer, complicates the development of standardized treatment protocols based on CIRBP modulation. There is also the challenge of identifying reliable biomarkers associated with CIRBP activity that can be used for patient stratification and monitoring treatment responses. Moreover, the regulatory landscape for translating laboratory findings into clinical practice can be cumbersome, often delaying the progression of promising therapies. Addressing these challenges requires collaborative efforts between researchers, clinicians, and regulatory bodies to streamline the process of translating CIRBP-related discoveries into effective clinical interventions.

3. Conclusions

In summary, the Cold-Inducible RNA-Binding Protein (CIRBP) family has emerged as a significant player in cancer biology, influencing various molecular mechanisms associated with tumor progression and resistance. The multifaceted role of CIRBP in cancer encompasses its involvement in cellular stress responses, regulation of gene expression, and modulation of RNA metabolism, thereby impacting cell survival, proliferation, and differentiation. As our understanding of CIRBP’s functions continues to evolve, it becomes increasingly clear that this family of proteins holds considerable promise as a therapeutic target.

CIRBP’s potential as a therapeutic target is underscored by its ability to influence critical pathways that are often dysregulated in cancer. By targeting CIRBP, there is the possibility of not only inhibiting tumor growth but also overcoming resistance to conventional therapies. However, translating these findings into clinical applications poses several challenges. One of the primary hurdles is the need for a comprehensive understanding of the diverse roles CIRBP plays in different cancer types, as its expression and function may vary significantly depending on the tumor microenvironment and genetic context.

Future research should focus on elucidating the specific mechanisms by which CIRBP contributes to tumor biology. This includes investigating how CIRBP interactions with other cellular proteins and RNAs influence cancer-related processes. Additionally, there is a need for more in vivo studies that can validate CIRBP’s role in tumorigenesis and its potential as a biomarker for treatment response. Understanding the balance between CIRBP’s tumor-promoting and tumor-suppressive roles will be crucial for developing targeted therapies.

Moreover, the integration of multi-omics approaches could enhance our comprehension of CIRBP’s function in the broader landscape of cancer biology. This integrative strategy may help identify novel CIRBP-related pathways and allow for the identification of patient populations that would benefit most from CIRBP-targeted therapies.

In conclusion, while the CIRBP family presents a promising avenue for cancer treatment, significant research is still required to navigate the complexities of its role in tumor biology. By addressing these challenges and focusing on the multifaceted nature of CIRBP, we can pave the way for innovative therapeutic strategies that harness the potential of this protein family in combating cancer.

Declaration Section

Publication Consent

There are no disputes regarding copyright or publication rights.

Author Contributions

Yu Cai was primarily responsible for collecting and reading literature, as well as writing articles.

Acknowledgements

With deep gratitude, I extend my heartfelt thanks to PubMed and all other academic websites.

NOTES

*First Author.

Conflicts of Interest

All authors of this study declare that there are no conflicts of interest that may affect the impartiality of the study results. We commit to maintaining an objective and impartial attitude in writing the research report and publishing the research findings, without being influenced by any external factors.

References

[1] Damaghi, M., Mori, H., Byrne, S., Xu, L., Chen, T., Johnson, J., et al. (2020) Collagen Production and Niche Engineering: A Novel Strategy for Cancer Cells to Survive Acidosis in DCIS and Evolve. Evolutionary Applications, 13, 2689-2703.
https://doi.org/10.1111/eva.13075
[2] Wu, T., Liao, C., Lu, W., Chang, C., Tsai, F., Jiang, S., et al. (2022) Identification of Distinct Slow Mode of Reversible Adaptation of Pancreatic Ductal Adenocarcinoma to the Prolonged Acidic Ph Microenvironment. Journal of Experimental & Clinical Cancer Research, 41, Article No. 137.
https://doi.org/10.1186/s13046-022-02329-x
[3] Liao, Y., Feng, J., Sun, W., Wu, C., Li, J., Jing, T., et al. (2021) CIRP Promotes the Progression of Non-Small Cell Lung Cancer through Activation of Wnt/β-Catenin Signaling via CTNNB1. Journal of Experimental & Clinical Cancer Research, 40, Article No. 275.
https://doi.org/10.1186/s13046-021-02080-9
[4] Lin, T., Chen, Y., Jia, J., Zhou, C., Lian, M., Wen, Y., et al. (2019) Loss of CIRBP Expression Is Correlated with the Malignant Progression and Poor Prognosis in Nasopharyngeal Carcinoma. Cancer Management and Research, 11, 6959-6969.
https://doi.org/10.2147/cmar.s211389
[5] Wisowski, G., Pudełko, A., Olczyk, K., Paul-Samojedny, M. and Koźma, E.M. (2022) Dermatan Sulfate Affects Breast Cancer Cell Function via the Induction of Necroptosis. Cells, 11, Article 173.
https://doi.org/10.3390/cells11010173
[6] Wilczyński, J.R., Wilczyński, M. and Paradowska, E. (2022) Cancer Stem Cells in Ovarian Cancer—A Source of Tumor Success and a Challenging Target for Novel Therapies. International Journal of Molecular Sciences, 23, Article 2496.
https://doi.org/10.3390/ijms23052496
[7] Corre, M. and Lebreton, A. (2024) Regulation of Cold-Inducible RNA-Binding Protein (CIRBP) in Response to Cellular Stresses. Biochimie, 217, 3-9.
https://doi.org/10.1016/j.biochi.2023.04.003
[8] Indacochea, A., Guerrero, S., Ureña, M., Araujo, F., Coll, O., LLeonart, M.E., et al. (2020) Cold-Inducible RNA Binding Protein Promotes Breast Cancer Cell Malignancy by Regulating Cystatin C Levels. RNA, 27, 190-201.
https://doi.org/10.1261/rna.076422.120
[9] Gong, E., Jung, D., Woo, H., Song, J., Choi, E., Jo, S., et al. (2024) Genomic Analysis Uncovers That Cold-Inducible RNA Binding Protein Is Associated with Estrogen Receptor in Breast Cancer. Genes & Genomics, 46, 899-907.
https://doi.org/10.1007/s13258-024-01530-w
[10] Wang, Q., Zhang, Q., Wu, D., Yin, X. and Zhang, G. (2007) Neuroprotection of Selenite against Ischemic Brain Injury through Negatively Regulating Early Activation of ASK1/JNK Cascade via Activation of PI3K/AKT Pathway. Acta Pharmacologica Sinica, 28, 19-27.
https://doi.org/10.1111/j.1745-7254.2007.00469.x
[11] Qi, Z., Yan, F., Shi, W., Zhang, C., Dong, W., Zhao, Y., et al. (2014) AKT-Related Autophagy Contributes to the Neuroprotective Efficacy of Hydroxysafflor Yellow A against Ischemic Stroke in Rats. Translational Stroke Research, 5, 501-509.
https://doi.org/10.1007/s12975-014-0346-x
[12] Meloche, S. and Pouysségur, J. (2007) The ERK1/2 Mitogen-Activated Protein Kinase Pathway as a Master Regulator of the G1-to S-Phase Transition. Oncogene, 26, 3227-3239.
https://doi.org/10.1038/sj.onc.1210414
[13] Peng, Y., Kok, K.H., Xu, R., Kwok, K.H.H., Tay, D., Fung, P.C.W., et al. (2000) Maternal Cold Inducible RNA Binding Protein Is Required for Embryonic Kidney Formation in Xenopus laevis. FEBS Letters, 482, 37-43.
https://doi.org/10.1016/s0014-5793(00)02019-6
[14] Peng, Y., Yang, P., Tanner, J.A., Huang, J., Li, M., Lee, H.F., et al. (2006) Cold-Inducible RNA Binding Protein Is Required for the Expression of Adhesion Molecules and Embryonic Cell Movement in Xenopus Laevis. Biochemical and Biophysical Research Communications, 344, 416-424.
https://doi.org/10.1016/j.bbrc.2006.03.086
[15] Gao, H., Xie, R., Huang, R., Wang, C., Wang, Y., Wang, D., et al. (2022) CIRBP Regulates Pancreatic Cancer Cell Ferroptosis and Growth by Directly Binding to P53. Journal of Immunology Research, 2022, Article ID: 2527210.
https://doi.org/10.1155/2022/2527210
[16] Chen, C., Wang, S., Tang, Y., Liu, H., Tu, D., Su, B., et al. (2024) Identifying Epithelial-Mesenchymal Transition-Related Genes as Prognostic Biomarkers and Therapeutic Targets of Hepatocellular Carcinoma by Integrated Analysis of Single-Cell and Bulk-RNA Sequencing Data. Translational Cancer Research, 13, 4257-4277.
https://doi.org/10.21037/tcr-24-521
[17] Yuan, S., Sun, X. and Wang, L. (2022) Prognostic Values from Integrated Analysis of the Nomogram Based on RNA-Binding Proteins and Clinical Factors in Endometrial Cancer. Clinical Medicine Insights: Oncology, 16, 1-14.
https://doi.org/10.1177/11795549221123620
[18] Chen, X., Xie, H., Wang, X., Zheng, Z. and Jin, S. (2021) CIRBP Knockdown Attenuates Tumourigenesis and Improves the Chemosensitivity of Pancreatic Cancer via the Downregulation of DYRK1B. Frontiers in Cell and Developmental Biology, 9, Article 667551.
https://doi.org/10.3389/fcell.2021.667551
[19] Hozumi, H., Kataoka, K., Kondoh, Y., Isayama, T., Okada, J., Sugiura, K., et al. (2021) Clinical Significance of Cold-Inducible RNA-Binding Protein in Idiopathic Pulmonary Fibrosis. Chest, 160, 2149-2157.
https://doi.org/10.1016/j.chest.2021.06.067
[20] Lin, T., Jia, J., Luo, W., Lin, X., Xiao, S., Yang, J., et al. (2024) Thermomir-377-3p-induced Suppression of Cirbp Expression Is Required for Effective Elimination of Cancer Cells and Cancer Stem-Like Cells by Hyperthermia. Journal of Experimental & Clinical Cancer Research, 43, Article No. 62.
https://doi.org/10.1186/s13046-024-02983-3
[21] Zhang, S., Li, Z., Hu, G. and Chen, H. (2025) Integrative Single-Cell and Multi-Omics Analyses Reveal Ferroptosis-Associated Gene Expression and Immune Microenvironment Heterogeneity in Gastric Cancer. Discover Oncology, 16, Article No. 57.
https://doi.org/10.1007/s12672-025-01798-8
[22] Li, L., Liu, D., Chen, T., Wei, C., Qiao, Y., Liu, W., et al. (2024) Hypoxia-Enhanced YAP1-EIF4A3 Interaction Drives Circ_0007386 Circularization by Competing with CRIM1 Pre-mRNA Linear Splicing and Promotes Non-Small Cell Lung Cancer Progression. Journal of Experimental & Clinical Cancer Research, 43, Article No. 200.
https://doi.org/10.1186/s13046-024-03116-6
[23] Li, Y., Wu, X., Miao, S. and Cao, Q. (2022) MiR-383-5p Promotes Apoptosis of Ovarian Granulosa Cells by Targeting CIRP through the PI3K/AKT Signaling Pathway. Archives of Gynecology and Obstetrics, 306, 501-512.
https://doi.org/10.1007/s00404-022-06461-z
[24] Wu, S., Li, Z., Li, H. and Liao, K. (2022) Dihydroartemisinin Reduces Irradiation-Induced Mitophagy and Radioresistance in Lung Cancer A549 Cells via CIRBP Inhibition. Life, 12, Article 1129.
https://doi.org/10.3390/life12081129

Copyright © 2025 by authors and Scientific Research Publishing Inc.

Creative Commons License

This work and the related PDF file are licensed under a Creative Commons Attribution 4.0 International License.