Spandidos Publications Logo
  • About
    • About Spandidos
    • Aims and Scopes
    • Abstracting and Indexing
    • Editorial Policies
    • Reprints and Permissions
    • Job Opportunities
    • Terms and Conditions
    • Contact
  • Journals
    • All Journals
    • Oncology Letters
      • Oncology Letters
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • International Journal of Oncology
      • International Journal of Oncology
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Molecular and Clinical Oncology
      • Molecular and Clinical Oncology
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Experimental and Therapeutic Medicine
      • Experimental and Therapeutic Medicine
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • International Journal of Molecular Medicine
      • International Journal of Molecular Medicine
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Biomedical Reports
      • Biomedical Reports
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Oncology Reports
      • Oncology Reports
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Molecular Medicine Reports
      • Molecular Medicine Reports
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • World Academy of Sciences Journal
      • World Academy of Sciences Journal
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • International Journal of Functional Nutrition
      • International Journal of Functional Nutrition
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • International Journal of Epigenetics
      • International Journal of Epigenetics
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Medicine International
      • Medicine International
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
  • Articles
  • Information
    • Information for Authors
    • Information for Reviewers
    • Information for Librarians
    • Information for Advertisers
    • Conferences
  • Language Editing
Spandidos Publications Logo
  • About
    • About Spandidos
    • Aims and Scopes
    • Abstracting and Indexing
    • Editorial Policies
    • Reprints and Permissions
    • Job Opportunities
    • Terms and Conditions
    • Contact
  • Journals
    • All Journals
    • Biomedical Reports
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Experimental and Therapeutic Medicine
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • International Journal of Epigenetics
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • International Journal of Functional Nutrition
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • International Journal of Molecular Medicine
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • International Journal of Oncology
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Medicine International
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Molecular and Clinical Oncology
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Molecular Medicine Reports
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Oncology Letters
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Oncology Reports
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • World Academy of Sciences Journal
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
  • Articles
  • Information
    • For Authors
    • For Reviewers
    • For Librarians
    • For Advertisers
    • Conferences
  • Language Editing
Login Register Submit
  • This site uses cookies
  • You can change your cookie settings at any time by following the instructions in our Cookie Policy. To find out more, you may read our Privacy Policy.

    I agree
Search articles by DOI, keyword, author or affiliation
Search
Advanced Search
presentation
Oncology Letters
Join Editorial Board Propose a Special Issue
Print ISSN: 1792-1074 Online ISSN: 1792-1082
Journal Cover
November-2025 Volume 30 Issue 5

Full Size Image

Sign up for eToc alerts
Recommend to Library

Journals

International Journal of Molecular Medicine

International Journal of Molecular Medicine

International Journal of Molecular Medicine is an international journal devoted to molecular mechanisms of human disease.

International Journal of Oncology

International Journal of Oncology

International Journal of Oncology is an international journal devoted to oncology research and cancer treatment.

Molecular Medicine Reports

Molecular Medicine Reports

Covers molecular medicine topics such as pharmacology, pathology, genetics, neuroscience, infectious diseases, molecular cardiology, and molecular surgery.

Oncology Reports

Oncology Reports

Oncology Reports is an international journal devoted to fundamental and applied research in Oncology.

Experimental and Therapeutic Medicine

Experimental and Therapeutic Medicine

Experimental and Therapeutic Medicine is an international journal devoted to laboratory and clinical medicine.

Oncology Letters

Oncology Letters

Oncology Letters is an international journal devoted to Experimental and Clinical Oncology.

Biomedical Reports

Biomedical Reports

Explores a wide range of biological and medical fields, including pharmacology, genetics, microbiology, neuroscience, and molecular cardiology.

Molecular and Clinical Oncology

Molecular and Clinical Oncology

International journal addressing all aspects of oncology research, from tumorigenesis and oncogenes to chemotherapy and metastasis.

World Academy of Sciences Journal

World Academy of Sciences Journal

Multidisciplinary open-access journal spanning biochemistry, genetics, neuroscience, environmental health, and synthetic biology.

International Journal of Functional Nutrition

International Journal of Functional Nutrition

Open-access journal combining biochemistry, pharmacology, immunology, and genetics to advance health through functional nutrition.

International Journal of Epigenetics

International Journal of Epigenetics

Publishes open-access research on using epigenetics to advance understanding and treatment of human disease.

Medicine International

Medicine International

An International Open Access Journal Devoted to General Medicine.

Journal Cover
November-2025 Volume 30 Issue 5

Full Size Image

Sign up for eToc alerts
Recommend to Library

  • Article
  • Citations
    • Cite This Article
    • Download Citation
    • Create Citation Alert
    • Remove Citation Alert
    • Cited By
  • Similar Articles
    • Related Articles (in Spandidos Publications)
    • Similar Articles (Google Scholar)
    • Similar Articles (PubMed)
  • Download PDF
  • Download XML
  • View XML
Review Open Access

Molecular mechanisms and functions of guanylate‑binding protein 2 in inflammation and cancer (Review)

  • Authors:
    • Zeyu Liu
    • Shijun Peng
    • Jia Ouyang
  • View Affiliations / Copyright

    Affiliations: School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100083, P.R. China, Department of Neurosurgery, Peking University People's Hospital, Beijing 100044, P.R. China
    Copyright: © Liu et al. This is an open access article distributed under the terms of Creative Commons Attribution License.
  • Article Number: 534
    |
    Published online on: September 18, 2025
       https://doi.org/10.3892/ol.2025.15280
  • Expand metrics +
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Metrics: Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )
Cited By (CrossRef): 0 citations Loading Articles...

This article is mentioned in:



Abstract

The present review article aimed to summarize currently available research data on the roles and functions of human guanylate‑binding protein 2 (hGBP2) and murine (m)GBP2 in cancer and inflammation. In addition, its structure, hydrolytic mechanisms and molecular regulatory mechanisms are discussed. hGBP2 and mGBP2 are strongly induced by IFN‑γ through the IFN‑sensitive response element and IFN‑γ activation site sequences on their corresponding genes, whilst also being regulated by IFN‑α, IFN‑β and multiple STAT‑IFN‑regulatory factor complexes. In inflammation, hGBP2 primarily regulates activation of the NLR family pyrin domain‑containing 3 and absent in melanoma 2 inflammasome pathways and therefore the induction of pyroptosis. By contrast, in cancer, hGBP2 serves a dual role, such that it can either promote cancer progression or suppress cancer development in a context‑dependent manner. This leads to variations in the hGBP2 expression profile across different cancer types in addition to their corresponding prognostic outcomes. hGBP2 can also respond to paclitaxel, a notable anticancer drug. The present review aims to summarize the structural basis and regulatory mechanisms of hGBP2, elucidate the roles of both hGBP2 and mGBP2 in inflammation and cancer and propose prospective research directions to inform future fundamental investigations and clinical applications.

Introduction

Human guanylate-binding protein 2 (hGBP2) and murine (m)GBP2 were first isolated from human fibroblasts in 1982 (1) and macrophages in 1998 (2). GBP2 is a member of the IFN-inducible GTPase family that serves notable roles in cellular signaling. hGBP2 is reported to be highly expressed in various immune cells, including monocytes, lymphocytes and natural killer cells (3). Under basal conditions, hGBP2 and mGBP2 are primarily distributed diffusely in the cytoplasm and nucleus. However, upon stimulation by IFN-γ and in the GTP-bound state, such as hGBP1 and hGBP5, it can translocate to the Golgi apparatus (4,5). The genes encoding hGBP1-7 are located on human chromosome 1, whilst the genes encoding mGBP1-11 are located on mouse chromosome 3 (3), where mGBP2 is mapped to the distal end and is putatively associated with mGBP1 (2).

The inflammasome is a multi-protein complex that functions to eliminate abnormal cells and amplify inflammation. It is comprised of various sensor proteins, such as the NLR family pyrin domain-containing 3 [NLRP3; which primarily recognizes lipopolysaccharide (LPS)-induced signals], absent in melanoma 2 (AIM2; which detects intracellular double-stranded DNA), adaptor proteins, such as the apoptosis-associated speck-like protein containing a caspase recruitment domain (ASC) and caspase effector proteins. It also senses pathogen-associated molecular patterns (PAMPs) or damage-associated molecular patterns (DAMPs), thereby triggering pyroptosis, a type of programmed cell death characterized by gasdermin-D-mediated plasma membrane perforation and release of inflammatory cytokines. hGBP2 activates inflammasome assembly and pyroptosis primarily by binding and delivering LPS into the cytoplasm (6). Additionally, hGBP2 facilitates inflammasome assembly through direct binding to ASC and cooperates with hGBP5 (7).

In the context of cancer, paclitaxel (PTX), which has become an important chemotherapeutic agent since its discovery in 1967, is a natural diterpenoid compound that can be isolated from Taxus brevifolia. hGBP2 cooperates with PTX through the MCL-1 apoptosis regulator, myeloid cell leukemia-1 (MCL-1)/Bcl-2 antagonist/killer 1 (Bak) pathway (8) to activate the vascular endothelial growth factor (VEGF) pathway, thereby promoting angiogenesis and oxygen supply to tumor. Immunologically ‘cold’ tumors are characterized by poor responses to immunotherapies (9) and host a tumor immunosuppressive microenvironment (TIME) lacking infiltrating immune cells. Such tumors typically exhibit rapid progression and poor prognosis (10), posing a major challenge in cancer immunotherapy. Notably, hGBP2 may offer a promising therapeutic strategy for ‘cold’ tumors by activating a number of immune checkpoints, such as programmed cell death protein 1 (PD-1)/programmed death-ligand 1 (PD-L1) and lymphocyte activating 3 (LAG3) (11), thereby remodeling the TIME and inhibiting the activation of CD8+ T cells and CD4+ T cells (12).

Structure and hydrolase function of hGBP2

Structure of hGBP2

hGBP2 is a 67 kDa GTPase that is comprised of three distinct domains (13). The N-terminal region contains a large GTPase domain (LG; residues 1–309), followed by the middle domain (MD; residues 310–480) in the central region and the α12/13 helical domain (HD; residues 480–580) at the C-terminus. In its crystalline state, hGBP2 adopts a bi-molecular asymmetric unit configuration.

The LG domain serves as the nucleotide-binding site and is characterized by five conserved motifs involved in GTP binding and hydrolysis (Table I) (14). The G1/P-loop motif has a GxxxxGKS/T sequence (15) that encloses the β-phosphate of GTP. By contrast, the G2/SWITCH I motif serves a critical role in magnesium ion binding, which is a key feature for all GTPases, since it stabilizes the GTP molecule through coordination bonds with the phosphate groups. The G3/SWITCH II motif, with its conserved DTEG residue sequence, forms hydrogen bonds with the phosphate, aiding in its release. The G4 motif interacts specifically with the guanine base with its acidic Asp residue in the RDF residue sequence, facilitating the binding of GTP to the domain. The primary non-conserved region, located between residue 240 and 280, corresponds to a flexible region involved in membrane interactions and is situated within the G5 region. The G1-G5 motifs collectively constitute the ‘nucleotide-binding pocket’ for GTP (Table I).

Table I.

Key motifs (G1-G5) of the human guanylate-binding protein 2 hydrolysis site.

Table I.

Key motifs (G1-G5) of the human guanylate-binding protein 2 hydrolysis site.

Motif designationSequenceCorresponding residue numbers
G1/P-loopGLYRTGKS45-52
G2/SWITCH ITVKSHT70-75
G3/SWITCH IIDTEG97–100
G4RDF181-183
G5/Guanine cap WPAPKKYLAHLEQLKEEE236-255

The MD domain consists of 5 α-helices. The first helix group is formed of the N-terminal halves of α7, α8 and α9, where the second helix group is formed of the C-terminal half of α9, α10 and α11. By contrast, the α12/α13 domain contains a conserved CaaX sequence, where isoprenylation occurs, promoting the localization of hGBP2 on the membrane as a hydrophobic group. Another feature of α12/α13 domain is its interaction with LG domain. The α12/α13 domain is mainly negatively charged, while the LG domain mainly carries positive charge. This enables the two domains to contact through electrostatic interactions (13,16), where the α12/α13 domain is in a tightened state.

Hydrolase function of hGBP2

The hydrolase function of hGBP2 is essential for a number of mechanisms and processes. The membrane localization of hGBP2 requires homodimerization, which is dependent on the transition state of GTP hydrolysis (17).

The hGBP2 hydrolysate, which contains >75% GDP (18,19), yields only a small amount of GMP as the final product (13,16), which is obtained by the hydrolysis of GTP instead of GDP. This distinct characteristic, differing from hGBP1, is primarily attributed to the LG domain of hGBP2 rather than changes in the MD or HD domains. Specifically, this is due to differences in the adjustment of the active site in the LG domain of hGBP2 following the first hydrolysis event. In terms of tetramerization, hGBP2, unlike hGBP1 but like mGBP2 (18), can extensively tetramerize, although the tetramerization of hGBP2 does not contribute to the formation of GMP (20,21).

When hGBP2 binds GTP, GTP enters the nucleotide binding pocket formed by the G1-G5 motif, where hGBP2 undergoes homodimerization at the same time with the participation of LG and MD domains (22,23). The LG domain of hGBP2 is dimerized only 50% of the time in the presence of GTP, suggesting that the C-terminal domain of hGBP2 also participates in dimerization by providing a dimerization interface or otherwise stabilizing hGBP2 dimers in the GTP state (16). In addition, residue T75 in the G2/SWITCH motif and S52 in the G1 motif are involved in Mg2+ coordination (13), implying that Mg2+ may enter the nucleotide binding pocket and bind to the phosphate group and surrounding amino acid residues at this stage, which are necessary for hGBP2 activity. It has previously been reported that Mg2+ is necessary for the activation of hGBP2 at Ph 8.0, but not pH 6.0 (16), which suggests that Mg2+ and H3O+ may have a functional substitution relationship. However, this interaction requires further validation.

In the hGBP2-GDP conformation following phosphate group dissociation, the N-O bond of the G1/P-loop motif interacts with the β-phosphate. Concurrently, residue S52 forms N-O bonds with both the α- and β-phosphates, facilitating nucleotide positioning and binding (24,25). The Y47 side chain undergoes movement and participates in dimer interface formation, whilst the displacement of Y47 and R48 in the G1 motif induces notable movement of the G2 motif, with some Cα atoms shifting >10 Å (26). The G4/RD motif interacts with the guanine moiety through hydrogen bonding. W238 in the G5 motif undergoes allosteric rearrangement and interacts with W238 from another hGBP2 molecule (13). Consequently, the guanine base and ribose are enclosed within the space formed by the G1, G4 and G5 motifs. At the opposite end of the GDP, the diphosphate group is tightly positioned within the pocket formed by the G1/P-loop, G2/SWITCH I and G3/SWITCH II motifs. The β-phosphate further coordinates with the conserved residues K51, S52 and E99. When the E99-K51 interaction is absent, SWITCH I remains open. However, when these residues engage through hydrogen bonding, SWITCH I closes, simultaneously positioning T75 in the G2 motif for optimal interaction with Mg2+ (16).

Regulation of hGBP2 and mGBP2

Structural foundation of hGBP2 and mGbp2 genes

The hGBP2 gene, as an IFN-stimulated gene (ISG), contains a promoter region featuring two transcription factor binding sequences, namely the IFN-stimulated response element (ISRE), located at +42 (27) and the γ-activated sequence (GAS) (27,28), located at +53 and at −532 (27). Notably, GAS exhibits binding affinity for all STAT factors except for STAT2. Both sequences can independently respond to IFN signaling, whilst their combined presence results in enhanced transcriptional activity (29). By contrast, the mGbp2 gene is also an ISG, which contains one ISRE, located at −2 (27) and two GASs, located at −487 and −735 (27). However, Ramsauer et al (28) previously reported that the mGbpP2 gene has two ISREs, located at −30 and −440 and one GAS located at −530 (28).

Molecular regulatory mechanisms
IFN-independent hGBP2 and mGbp2 gene expression under normal conditions

STAT1, STAT2 and IFN-regulatory factor (IRF) 9 can form four types of oligomers. They are STAT1-STAT1 (binding to GAS, referred to as U-STAT1), STAT1-STAT2-IRF9 (binding to GAS, referred to as U-ISGF3), STAT2-STAT2-IRF9 (binding to ISRE, referred to as U-ST2) and IRF1-IRF1 (binding to ISRE) (29). These oligomers subsequently localize to hGBP2 or mGbp2 genes and activate their transcription (Fig. 1). In mice, the IRF1 dimer has been demonstrated to directly bind to the transcription complex containing RNA polymerase II to activate mGbp2 gene transcription (28), whilst STAT1 dimers not only bind to GAS on the hGBP2 gene promoter but also promote the acetylation of histone 4 in the hGBP2 gene, thereby providing active chromatin. Additionally, previous studies have shown that p53 can form a complex with IRF1 to upregulate hGBP2 expression (30,31).

GBP2 gene regulation under basal
conditions (without IFN-I/II stimulation). GBP2, guanylate-binding
protein 2; GAS, γ-activated sequence; ISRE, IFN-stimulated response
element; IFNAR, IFN-α/β receptor; IFNGR, IFN-γ receptor; IRF,
IFN-regulatory factor; ISGF, IFN-stimulated gene factor.

Figure 1.

GBP2 gene regulation under basal conditions (without IFN-I/II stimulation). GBP2, guanylate-binding protein 2; GAS, γ-activated sequence; ISRE, IFN-stimulated response element; IFNAR, IFN-α/β receptor; IFNGR, IFN-γ receptor; IRF, IFN-regulatory factor; ISGF, IFN-stimulated gene factor.

Highly activated phase under stimulation by IFN-I, IFN-II and IRF1

Upon pathogen invasion or intracellular abnormalities, the corresponding PAMPs or DAMPs are recognized by a series of pattern recognition receptors (PRRs). This type of recognition triggers the phosphorylation and activation of IRFs, thereby promoting the expression of IFN-I and IFN-II (the former mainly includes subtypes IFN-α and IFN-β) through the classical secretory pathway. These IFNs are secreted extracellularly and bind to their corresponding respective receptors. Mitochondrial outer membrane permeabilization (MOMP) under radiation conditions and apoptosis can lead to the release of mitochondrial double-stranded (ds)DNA, which activates the cyclic GMP-AMP synthase/stimulator of IFN genes pathway, subsequently inducing IFN-β and upregulating mGBP2 expression (32).

IFN-I can bind to the ubiquitously expressed IFN-α/β receptor (consisting of the IFNAR1 and IFNAR2 subunits), leading to the dimerization of the receptor subunits. This dimerization, through juxtaposition and trans-phosphorylation, enhances the kinase activity of Janus kinase (JAK) 1 and tyrosine kinase (TYK) 2. Subsequently, JAK1 and TYK2 phosphorylate tyrosine residues on IFNAR1 and IFNAR2, which serve as binding sites for STAT1 and STAT2 (33). Phosphorylation of STAT1 at Tyr701 and STAT2 at Tyr690 then occurs. By contrast, IFN-II, specifically IFN-γ, interacts with a tetrameric receptor complex composed of two IFNGR1 subunits and two IFNGR2 subunits. This receptor is associated with JAK1 and JAK2 kinases, which exclusively phosphorylate the STAT1 protein.

Similar to the pathway under normal conditions, phosphorylated (p-)STAT1, p-STAT2 and IRF1 can form four types of oligomers: p-STAT1-p-STAT1 (also known as γ-IFN activation factor, binds to GAS), p-STAT1-p-STAT2-IRF9 (binds GAS), pSTAT1-pSTAT2-IRF9 (also known as IFN-stimulated gene factor, binds to ISRE) and IRF1-IRF1 (binds ISRE). These oligomers subsequently localize to the hGBP2 or mGbp2 gene and activate its transcription through a similar mechanism (Fig. 2), exhibiting higher activity compared with their non-phosphorylated counterparts.

GBP2 gene regulation upon IFN-I and
IFN-II stimulation. GBP2, guanylate-binding protein 2; GAS,
γ-activated sequence; ISRE, IFN-stimulated response element; IFNAR,
IFN-α/β receptor; IFNGR, IFN-γ receptor; IRF, IFN-regulatory
factor; ISGF, IFN-stimulated gene factor; JAK1, Janus kinase 1;
TYK2, tyrosine kinase 2.

Figure 2.

GBP2 gene regulation upon IFN-I and IFN-II stimulation. GBP2, guanylate-binding protein 2; GAS, γ-activated sequence; ISRE, IFN-stimulated response element; IFNAR, IFN-α/β receptor; IFNGR, IFN-γ receptor; IRF, IFN-regulatory factor; ISGF, IFN-stimulated gene factor; JAK1, Janus kinase 1; TYK2, tyrosine kinase 2.

Function of hGBP2 and mGBP2 in inflammation

Activation of the inflammasome pathway by hGBP2 and mGBP2

The inflammasome pathway can be divided into two types, namely the canonical pathway and the non-canonical pathway. Their objective is to induce pyroptosis, which is mediated by the formation of plasma membrane pores through gasdermin-D. This leads to membrane rupture and alerting other cells, with the release of cytokines IL-1β and IL-18 through the gasdermin-D-formed membrane pores. hGBP2 and mGBP2 primarily serve supporting roles in inflammasome assembly.

In the canonical pathway, activation is primarily mediated by PRRs, such as nucleotide-binding oligomerization domain-like receptors, such as NLRP3, or AIM2-like receptors, including AIM. PAMPs or DAMPs are recognized by PRRs, such as NLRP3 or AIM2. During this process, mGBP2 can facilitate the release of dsDNA by lysing pathogen-containing vacuoles or pathogens, enabling AIM2 recognition (7,34,35). Upon activation, NLRP3 or AIM2 interacts with the adaptor protein ASC through their pyrin domains (36). In this step, mGBP2 and mGBP5 can form a heterocomplex, where mGBP2 binds ASC and mGBP5 binds NLRP3. This mGBP2-mGBP5 complex brings NLRP3 and ASC together, thereby promoting the assembly of the NLRP3 inflammasome (7). ASC in turn recruits caspase-1 through its caspase recruitment domain, forming the inflammasome. Subsequently, caspase-1 within the inflammasome cleaves gasdermin-D, releasing its N-terminal fragment to form plasma membrane pores and induce pyroptosis. Caspase-1 also processes pro-IL-1β and pro-IL-18 into their active forms, which are then released through the membrane pores to alert neighboring cells (7).

In the non-canonical pathway, activation in humans is primarily mediated by hGBPs through PRRs while in mice, it is mediated by caspase-11 with the assistance of mGBP2. In humans, when bacteria enter cells, hGBP1 binds bacterial LPS and recruits hGBP2, hGBP3 and hGBP4 to the surface of bacteria, especially gram-negative bacteria, forming a coating (6,37). hGBP2 and hGBP4 expose the lipid moiety of LPS, recruiting caspase-4 to the bacterial surface for LPS binding, while hGBP3 regulates caspase-4 activation (6,37). Activated caspase-4 then cleaves pro-IL-1β, pro-IL-18 and gasdermin-D, leading to pyroptosis and cytokine release.

In mice, caspase-11 directly recognizes the mGBP2-LPS complex, oligomerizes and activates, gaining the ability to cleave gasdermin-D and induce pyroptosis (38). Additionally, the formation of plasma membrane pores causes potassium ion efflux due to the intracellular potassium gradient, further promoting NLRP3 inflammasome assembly and creating a positive-feedback amplification loop (39). In this process, mGBP2 assists caspase-11 in LPS recognition. A previous study has reported that mGBP2 can interact with gasdermin-D and mGBP3, potentially serving as a novel component of this pathway, although the precise mechanisms remain unclear and require further investigation (7).

The severe consequences of pyroptosis, such as further inflammation cascade caused by the release of IL-18 and IL-1β (40), may explain the need for the continuity and regulation of the GBP2/caspase pathway. This type of regulation allows for the existence of multiple regulatory checkpoints before caspase activation, increasing the difficulty of activation and preventing unnecessary triggering (6).

Role of hGBP2 and mGBP2 in inflammatory diseases

The expression of hGBP2 is increased in the kidney tissues of patients with lupus nephritis, particularly in the glomeruli and renal tubulointerstitium (41). In diabetic nephropathy, macrophages at the injury site are predominantly of the M1 subtype, where hGBP2 can promote the polarization of macrophages toward the pro-inflammatory M1 subtype through the Notch 1 pathway. Through the hGBP2-mediated pathway, macrophages can be induced into the M1 phenotype by LPS or IFN-γ (42).

In allergic rhinitis (AR), mGBP2 can alleviate oxidative stress and abnormal lipid metabolism by inhibiting the hypoxia-inducible factor-1 (HIF-1) pathway. It can also inhibit mitochondrial fission whilst maintaining mitochondrial fusion to mitigate oxidative stress-induced damage to cells (43). A previous study has demonstrated that the overexpression of mGBP2 can notably reduce inflammatory cell infiltration into the nasal mucosa and markedly decrease the levels of various factors, such as total cholesterol, low-density lipoprotein-cholesterol, TNF-α, IL-5, IFN-γ and trimethylamine N-oxide, in AR mouse models whilst increasing high-density lipoprotein-cholesterol levels (44). However, the underlying mechanisms require further investigation.

LPS-induced macrophages-derived exosomes (L-Exo) can be transported from macrophages to lung epithelial cells, resulting in damage to the alveolar epithelial tissue. The mGBP2 content in L-Exo is higher compared with that in control Exo (45), leading to the hypothesis that the mGBP2 contained in L-Exo can trigger pyroptosis in the lung epithelial tissue, thereby causing injury. This may represent a potential therapeutic approach for sepsis-associated acute lung injury (46). In depression-like behaviors induced by neuroinflammation, reducing the expression of mGBP2 may also alleviate symptoms (47).

Role of hGBP2 and mGBP2 in oncogenesis and cancer

hGBP2 and mGBP2 regulate cancer progression through multiple signaling pathways

Previous studies have demonstrated that in glioblastoma (GBM) and low-grade glioma cells, hGBP2 can directly interact with kinesin family member 22 (KIF22) to post-transcriptionally regulate and elevate its levels, thereby enhancing KIF22-mediated EGFR internalization and signaling (Table II) (48–63). This in turn fosters cell proliferation (64). Unc-51 like autophagy activating kinase 1 (ULK1) is another critical regulator of autophagy (65), phosphorylating and inducing autophagy under low-nutrient conditions. hGBP2 can activate the ULK1 complex by suppressing PI3K/Akt/mTORC1 pathway (66), thereby enhancing cellular autophagy (66). Notably, the outcomes of this function may vary, where it may exert tumor-suppressive effects during early-stage cancer but potentially promote cancer progression at advanced stages (67).

Table II.

GBP2 expression level across different cancer types and corresponding mechanisms.

Table II.

GBP2 expression level across different cancer types and corresponding mechanisms.

Tumor typeCells or databases involvedSpeciesGBP2 expression levelMechanisms(Refs.)
GBMGlioblastoma cell lines, U87, U251, SNB19, GSC11 and G91; database, TCGAHumanUpregulatedExpression of various carcinogenesis-related genes in GBM, particularly in mesenchymal subtype, including fibronectin 1, MMP-1, MMP-3, MMP-14, urokinase, secreted protein acidic and rich in cysteine, TGFB1, macrophage colony-stimulating factor 1, CD44, IL-8, monocyte chemoattractant protein 1 and IL6, are elevated with hGBP2 involvement. Furthermore, cell proliferation is accelerated, the block of G0/G1 phase of cell cycle is prevented and apoptosis is decreased, due to hGBP2-activated kinesin family member 22/EGFR signaling pathway(48–51)
BC, particularly triple-negative BCPrimary BC cells isolated from tumor tissues from patients with BC and normal breast tissueHumanDownregulatedThe methylation of hGBP2 gene promoter decreases the levels of hGBP2, inhibiting the promotion of the immune response of CD8+ T cells, thus facilitating the development of BC(52–54)
SKCMHuman melanoma cell lines, A2058, A375; mouse melanoma cell lines, B16 and B16F10; human epidermal cell line, NHEKHuman and mouseDownregulatedIt has been revealed that overexpressing hGBP2 can upregulate E-cadherin and downregulate N-cadherin and vimentin, inhibiting the EMT, invasion and proliferation of SKCM. While whether hGBP2 downregulation, which occurs in SKCM cells, can facilitate EMT remains unstudied.hGBP2 also markedly downregulates the Wnt/β-catenin signaling pathway and related proteins, including transcription factor 4, c-Myc and cyclin D1, thus inhibiting cell proliferation, migration, invasion and promoting cell apoptosis. The mechanisms of mGBP2 in SKCM is unstudied(51,55)
ccRCCPrimary ccRCC cells isolated from patient tumor tissue and normal kidney tissue; monocyte cell line, THP-1; common RCC cell lines, 786-O, 769-P, CAKI-1, A498 and ACHN; renal tubular epithelium cell line, HK-2; databases, Clinical Proteomic Tumor Analysis Consortium and TCGAHumanUpregulatedhGBP2 activates the polarization of M0 macrophages toward the M2 macrophages through JAK/STAT3-dependent upregulation on IL-18 secretion. Simultaneously, M2 macrophages can induce the expression of GBP2 in tumor cells by secreting IL-10 and TGF-β, which in turn activates the expression of hGBP2, forming a loop. hGBP2 can also enhance the levels of regulatory T cells and exhausted T cells, facilitating ccRCC tumor immune evasion and proliferation(51,56,57)
PAADDatabases, TCGA and GEO; primary PAAD cells isolated from tumor tissue from patients with PAAD and adjacent normal tissueHumanUpregulatedhGBP2 levels are markedly elevated in active CD4 memory T cells, resting dendritic cells and M1 macrophages, indicating that these three types of cells can possibly induce the expression of hGBP2. Subsequently, hGBP2 can further regulate the TIME(11,51)
Thyroid carcinomaDatabase, TCGAHumanUpregulatedRemains unstudied(51)
Uveal melanomaDatabase, TCGAHumanUpregulatedRemains unstudied(51)
Hepatocellular carcinomaDatabases, TCGA and GEOHumanUpregulatedhGBP2 can induce tumor cell infiltration, especially by upregulating macrophages and downregulating Th17 and neutrophils in the TIME(51,58)
ESCCHuman ESCC cell lines, TE-1, TE-7, TE-10 and TE-13HumanUpregulatedhGBP2 is involved in a p53/IRF-1/hGBP2 pathway and is thus overexpressed in ESCC. Downstream mechanisms remain unstudied(30)
CRCDatabases: GEO, UCSC Xena Browser, TCGA; human MSS CRC cell lines, HT29 and SW480; murine MSS CRC cell line, CT26Human and mouseThe level of hGBP2 varies: Upregulated in MSS CRC of IC and dMMR/MSI CRC; downregulated in pMMR/MSS CRC of non-IChGBP2 can activate STAT1 by competing with SHP1, which inhibits the phosphorylation and activation of STAT1, for binding to STAT1(59,60)
Gastric cancerDatabases: GEO, TIDE, TCGA, UCSC Xena Browser, KEGGHumanUpregulatedRemains unstudied(61)
HNSCCDatabases, ONCOMINE, TCGA, HPAHumanStably upregulated across different stages of HNSCCRemains unstudied(31)
OCHuman cervical cancer cell line, HeLa; murine breast cancer cell line, 4T1; murine macrophage cell line, RAW264.7; murine preadipocyte cell line, 3T3-L1; murine ovarian cancer cell line, ID8Human and mouseDownregulatedmGBP2/Pin1/NF-κB pathway, which is possibly inhibited in OC, induces the polarization of M0 to M1, thus the anticancer macrophage subtype M1 is at low levels in OC(62)
Prostate cancerDatabases, KEGG, GEO, TCGAHumanUpregulatedRemains unstudied(63)

[i] GBP2, guanylate-binding protein 2; h, human; GBM, glioblastoma; TCGA, The Cancer Genome Atlas; BC, breast cancer; SKCM, skin cutaneous melanoma; EMT, epithelial-mesenchymal transition; ccRCC, clear cell renal cell carcinoma; JAK, Janus kinase; GEO, Gene Expression Omnibus; PAAD, pancreatic adenocarcinoma; TIME, tumor immunosuppressive microenvironment; ESCC, esophageal squamous cell carcinoma; CRC, colorectal cancer; MSS, microsatellite stable; MSI, microsatellite instability; dMMR, deficient DNA mismatch repair; pMMR, proficient DNA mismatch repair; IC, immune class; SHP1, tyrosine-protein phosphatase non-receptor type 6; TIDE, Tumor Immune Dysfunction and Exclusion; KEGG, Kyoto Encyclopedia of Genes and Genomes; HNSCC, head and neck squamous cell carcinoma; HPA, Human Protein Atlas; OC, ovarian cancer; Pin1, peptidyl-prolyl cis-trans isomerase NIMA-interacting 1.

hGBP2 can induce the assembly of inflammasomes, leading to a positive feedback loop of inflammatory responses. This forms the hGBP2/STAT3/fibronectin (FN1) pathway (49,53), where hGBP2 induces FN1, which is an extracellular glycoprotein involved in cell migration (68), on both transcriptional and translational levels to enhance glioblastoma migration and invasion without influencing proliferation in vitro (49). In addition, STAT3 contributes to maintaining the mesenchymal subtype of GBM (50) and is indispensable for hGBP2-induced FN1 elevation (49). FN1 can also promote MMP-9 secretion in a focal adhesion kinase (FAK)- and Ras-dependent manner (69) or through the FN1/MMP-2/MMP-9 pathway (70). However, hGBP2 can induce FN1 in vivo but lacks this capability in vitro (49). Furthermore, MMP-9 is an inducer of epithelial-mesenchymal transition (EMT) (71). Thus, hGBP2 should activate, rather than inhibit, MMP-9 through the STAT3/FN1/FAK or STAT3/FN1/MMP-2 signaling pathways. However, the regulatory effect of hGBP2 on migration has not been experimentally demonstrated.

Bak is a member of the Bcl-2 family that is crucial for the activation of apoptosis. It promotes MOMP, thereby triggering the apoptotic process (72). hGBP2 acts as a dual stimulator of Bak. It can not only release Bak but can also promote its expression (8). The LG domain of hGBP2 specifically binds to the BH3 domain of Mcl-1, a member of the Bcl-2 family, thereby preventing the interaction between Bak and Mcl-1. This then releases Bak, which oligomerizes to induce MOMP, ultimately promoting apoptosis (8). Simultaneously, hGBP2 upregulates the expression of Bak through its inhibitory effect on the PI3K/Akt pathway (8). In summary, hGBP2 can promote MOMP by activating Bak and enhancing its expression, thereby facilitating apoptosis. PTX, as an anticancer drug, occupies the upstream position in the hGBP2/Mcl-1/Bak pathway. Through this pathway, hGBP2 enhances cellular sensitivity to PTX (8,66). Notably, hGBP2 upregulation is a common feature of PTX treatment and other chemotherapeutic agents for hematologic malignancies (such as doxorubicin, cytarabine, vincristine, etoposide and IFN-γ). However, among these antineoplastic agents, only PTX possesses the unique ability to elevate Bak levels (8).

In breast cancer (BC) cells, hGBP2 has been demonstrated to directly interact with dynamin-related protein 1 (Drp1) in a K51-dependent manner, preventing its translocation from the cytoplasm to the mitochondria. This interaction inhibits Drp1-dependent mitochondrial fission and elongation, thereby blocking mitochondrial division and cell metastasis in cancer cells (73). Notably, the activity of hGBP2 binding to Drp1 is influenced by all three major structural domains of hGBP2, where it is possible that hGBP2 and Drp1 bind to each other to form hetero-oligomers, although further investigations are required (73). hGBP2 also exerts its anticancer effects by inhibiting the Wnt/β-catenin/EMT pathway (55,71,74) and cancer cell metastasis and invasion, which has been demonstrated in cancers such as skin cutaneous melanoma (SKCM) and colon cancer, but the specific molecular mechanisms remain unclear. PTX primarily inhibits tumor metastasis and progression by blocking angiogenesis in tumor tissues, whilst hGBP2 enhances the sensitivity of colon cancer cells to PTX by inhibiting the Wnt/β-catenin pathway (75,76). Additionally, a previous study has indicated that inhibiting the Wnt/β-catenin pathway in BC and triple-negative BC (TNBC) can promote ferroptosis in BC cells and suppress cell cycle and growth regulatory proteins, such as cyclin D1 and c-Myc. Therefore, it can be speculated that this may be a potential function of hGBP2, warranting further investigation.

In mouse dendritic cells, mGBP2 has been reported to interact with the Akt-p110 complex, inhibiting the phosphorylation and activation of Akt (77,78). This in turn prevents the phosphorylation of TSC complex subunit 2, disrupting its ability to form a complex with tuberous sclerosis 1 that inhibits mTORC1 (77–80). VEGF promotes endothelial cell proliferation and migration by binding to VEGFRs on endothelial cells, thereby stimulating angiogenesis. Under basal conditions, the HIF-1α subunit is hydroxylated by prolyl hydroxylases and recognized by the von Hippel-Lindau protein complex, leading to its ubiquitination and degradation. Under hypoxic conditions, however, the HIF-1α subunit is stabilized, where it translocates to the nucleus and dimerizes with the HIF-1β subunit to form the HIF-1 transcription factor, which then translocates to the nucleus and binds to hypoxia-response element in the promoter of VEGF gene (81,82), regulating VEGF transcription (43). In oxygen-induced retinopathy mice model, mGBP2 has been shown to inhibit the HIF-1α/VEGF pathway by suppressing mTORC1 (78), thereby attenuating angiogenesis. This reduces blood supply to cancer lesions, maintaining hypoxic conditions and potentially curbing cancer progression.

mGBP2 can activate MMP-9 through distinct pathways, thereby influencing collagen degradation near the basement membrane (33), extracellular matrix remodeling, angiogenesis in cancer lesions and cancer cell invasion and metastasis (77). Previous studies in NIH 3T3 fibroblasts have shown that mGBP2 can inhibit Rac, disrupting its regulation of the cytoskeleton. This inhibition suppresses the TNF-α-mediated activation of the NF-κB pathway, thereby reducing NF-κB-induced MMP-9 transcription (83). mGBP2 has been demonstrated in NIH 3T3 fibroblasts to directly interact with the p65 (RelA) protein in the NF-κB pathway, preventing its binding to the MMP-9 promoter and subsequently lowering MMP-9 expression (83). In ovarian cancer, mGBP2 promotes the recruitment of the peptidyl-prolyl cis-trans isomerase NIMA-interacting 1 (Pin1) protein, which activates the NF-κB pathway (62). Based on this, it can be hypothesized that mGBP2 may upregulate MMP-9 expression through the Pin1/NF-κB and Rac/NF-κB pathways, although further experimental validations are required.

hGBP2 and mGBP2 modulate cancer progression by regulating the TIME

hGBP2 exerts a dual influence on the TIME. It can promote immune activation, transforming the TIME into a ‘hot’ state, thereby addressing the immunologically ‘cold’ tumors, such as TNBC and proficient mismatch repair/microsatellite stable colorectal cancer (CRC) of the immune class (59). Simultaneously, it can also reduce immune cell infiltration through certain mechanisms, enhancing tumor immune evasion. Additionally, the expression of immunotherapy biomarkers is positively associated with hGBP2 expression (61). In CRC, hGBP2 expression is positively associated with CD8+ T cell infiltration, CD8, PD-L1, C-X-C motif chemokine ligand (CXCL) 9, CXCL10, CXCL11, CXCL13, HLA-I expression, antigen processing and presentation machinery and a variety of antitumor immunity steps, including the release of cancer cell antigens, cancer antigen presentation, priming and activation and trafficking of immune cells to tumors (59). By contrast, it is negatively associated with the cell count of cytokeratin (59). However, specific mechanisms of the regulation of hGBP2 in CRC require further study.

In gastric cancer, hGBP2 is reported to be positively associated with an inflamed TIME, immunophenoscore (IPS), abundance of PD-1+ cells and the expression of immunotherapy biomarkers, such as PD-L1, PD-L2, IFN-γ, CD8A, secreted and transmembrane protein 1 and IFN-induced transmembrane protein 3 (61). In GBM, it has been demonstrated that urokinase, secreted protein acidic and rich in cysteine, TGFB1, FN1 and colony-stimulating factor 1 are induced by elevated hGBP2 expression (49). In SKCM, previous studies have demonstrated that hGBP2 expression is positively associated with the infiltration of B cells, CD8+ T cells, CD4+ T cells, macrophages, neutrophils and dendritic cells (11,12,57,61), This may serve a role in therapies that block PD-1/PD-L1 interactions to prevent tumor immune evasion, since CD8+ T cells are the primary effector cells in tumor killing (84). In BC, hGBP2 is positively associated with T cell infiltration levels and can serve as a marker for T cell infiltration (53). In clear cell renal cell carcinoma (ccRCC), hGBP2 promotes the infiltration of CD8+ T cells, regulator T cells and both M1 and M2 macrophages (57).

From the perspective of reducing immune infiltration and promoting tumor immune evasion, hGBP2 can exert oncogenic effects by increasing the phosphorylation of STAT2 and STAT3, modulating the JAK/STAT signaling pathway and reducing tumor immune infiltration (51). hGBP2 can also induce immune checkpoints, such as PD-1/PD-L1, cytotoxic T-lymphocyte-associated protein 4 (CTLA-4), T cell immunoreceptor with Ig and ITIM domains, LAG3, indoleamine 2,3-dioxygenase 2 and V-domain Ig suppressor of T cell activation (VISTA) (11), which are inhibitory immune checkpoints. Additionally, hGBP2 can promote PD-L1 expression on the transcriptional level by binding to and phosphorylating STAT1, thereby reducing tumor immune infiltration and facilitating immune evasion (85). hGBP2 can also induce PD-L2; however, the relevant mechanisms remain unstudied (61). Based on these functions, hGBP2 will likely be a crucial target for restoring the TIME for the treatment of immunologically ‘cold’ tumors, such as TNBC.

mGBP2 has been found to upregulate the secretion of IL-6, IL-12 and TNF-α (77). Furthermore, mGBP2 promotes the maturation of dendritic cells and enhances their antigen-presenting capacity, thereby boosting T cell activation (77). mGBP2 also promotes the polarization of macrophages into M1 and M2 subtypes by promoting STAT3 pathway and by activating the NF-κB signaling pathway (62), while hGBP2 promotes polarization by stimulating the secretion of IL-18 (56). M2 macrophages, in turn, can enhance the migration and invasion of tumor cells by secreting IL-10 and TGF-β, which upregulate the hGBP2/STAT3 and ERK axes (part of the MAPK signaling pathway) as demonstrated in ccRCC (56).

Notably, a number of downstream factors promoted by hGBP2 can exhibit both pro-tumor and antitumor effects. CD80 in pancreatic adenocarcinoma can bind to either CD28 (activates T cells) or CTLA-4 (inhibits T cells), thereby bidirectionally modulating immune responses (11). Notably, hGBP2 is reported to promote the polarization of M0 to M2 in ccRCC by activating the secretion of IL-18 (56) and to M1 in ovarian cancer (62) and in diabetic nephropathy by activating the Notch 1 signaling pathway (42). By contrast, mGBP2 has only been reported to promote the polarization of M0 to M1 under the activation of the PLGA-CpG@ID8-M nano vaccine (62). The underlying mechanisms of these outcomes and whether mGBP2 can also promote the polarization of M0 to M2 require further investigation.

Regulation of hGBP2 in cancer

In primary CRC, hGBP2 has been found to exhibit a high mutation rate alongside genes such as G protein subunit β1 and GATA zinc finger domain containing 2A. However, only hGBP2 showed an even higher mutation rate in CRC with liver metastasis (60). Additionally, in CRC with liver metastasis, the hGBP2 gene undergoes methylation at four specific sites (m1A, m5C, m6A and m7G) (60). Peroxisome proliferator-activated receptor α, an anticancer factor, can inhibit hGBP2 expression (58). In BC (52) and SKCM (12), the methylation level of hGBP2 increases, leading to a decrease in hGBP2 expression.

Future directions

Prior reviews on GBP2 have predominantly emphasized their roles in host defense against bacterial and viral pathogens (6,86,87), with limited attention to the regulatory mechanisms of GBP2 expression or its functions in cancer and oncogenesis. Furthermore, species-specific distinctions-particularly the differences between hGBP2 and mGBP2-have been overlooked (86,87). This review bridges these gaps by providing a comprehensive analysis of the regulation of GBP2 expression and by systematically comparing hGBP2 and mGBP2 functions in oncogenesis. Whether a potential functional substitution relationship between Mg2+ and H3O+ during hGBP2 activation exists and its possible mechanisms could offer further insight into the hydrolysis of hGBP2, and requires further investigation. Although hGBP2 can form tetramers extensively, this tetramerization cannot facilitate the hydrolysis from GTP to GMP, rendering the physiological relevance of its tetramerization unknown. Moreover, while the context-dependent dual role of hGBP2 in cancer and oncogenesis has been extensively documented, the underlying mechanisms have not yet been systematically elucidated. For instance, although hGBP2 drives M0-to-M1 polarization via the Notch1 pathway in diabetic nephropathy, it mediates an M0-to-M2 polarization in ccRCC; the determinants of these opposing outcomes warrant further investigation.

Clinically, agonistic monoclonal antibodies which promote hGBP2 oligomerization and activation could be exploited to convert immunologically ‘cold’ tumors into ‘hot’ ones, thereby augmenting existing immunotherapies. Across multiple cancer types, hGBP2 abundance is positively associated with levels of immune-related biomarkers, such as PD-1/PD-L1 and VISTA, immune-cell infiltration and IPS, revealing its potential as a robust tumor biomarker for assessing the activation level of TIME and prognosis. Additionally, the methylation status of the hGBP2 promoter also has also emerged as a promising diagnostic indicator in SKCM and BC.

Acknowledgements

Not applicable.

Funding

The present study was Supported by the National Natural Science Foundation of China (grant no. 82104603), Beijing Natural Science Foundation (grant no. 7204322), Peking University People's Hospital Research and Development Fund (grant nos. RDZH2022-04, RS2021-12 and RDX2021-08).

Availability of data and materials

Not applicable.

Authors' contributions

ZL performed project administration, methodology, data curation and wrote the draft. SP performed visualization and reviewed the manuscript. JO conceived and designed the review, and was involved in funding acquisition, supervision, proofreading and revision of the manuscript. All authors read and approved the final version of the manuscript. Data authentication is not applicable.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

Glossary

Abbreviations

Abbreviations:

GBP2

guanylate-binding protein 2

Pin1

peptidyl-prolyl cis-trans isomerase NIMA-interacting 1

CRC

colorectal cancer

IRF

IFN-regulatory factor

TIME

tumor immunosuppressive microenvironment

ccRCC

clear cell renal cell carcinoma

SKCM

skin cutaneous melanoma

BC

breast cancer

GBM

glioblastoma

References

1 

Cheng YS, Colonno RJ and Yin FH: Interferon induction of fibroblast proteins with guanylate binding activity. J Biol Chem. 258:7746–7750. 1983. View Article : Google Scholar : PubMed/NCBI

2 

Vestal DJ, Buss JE, McKercher SR, Jenkins NA, Copeland NG, Kelner GS, Asundi VK and Maki RA: Murine GBP-2: A new IFN-gamma-induced member of the GBP family of GTPases isolated from macrophages. J Interferon Cytokine Res. 18:977–985. 1998. View Article : Google Scholar : PubMed/NCBI

3 

Quan ST, Jiao WW, Xu F, Sun L, Qi H and Shen A: Advances in the regulation of inflammasome activation by GBP family in infectious diseases. Yi Chuan. 45:1007–1017. 2023.PubMed/NCBI

4 

Britzen-Laurent N, Bauer M, Berton V, Fischer N, Syguda A, Reipschläger S, Naschberger E, Herrmann C and Stürzl M: Intracellular trafficking of guanylate-binding proteins is regulated by heterodimerization in a hierarchical manner. PLoS One. 5:e142462010. View Article : Google Scholar : PubMed/NCBI

5 

Modiano N, Lu YE and Cresswell P: Golgi targeting of human guanylate-binding protein-1 requires nucleotide binding, isoprenylation, and an IFN-gamma-inducible cofactor. Proc Natl Acad Sci USA. 102:8680–8685. 2005. View Article : Google Scholar : PubMed/NCBI

6 

Kirkby M, Enosi Tuipulotu D, Feng S, Lo Pilato J and Man SM: Guanylate-binding proteins: Mechanisms of pattern recognition and antimicrobial functions. Trends Biochem Sci. 48:883–893. 2023. View Article : Google Scholar : PubMed/NCBI

7 

Kim BH, Chee JD, Bradfield CJ, Park ES, Kumar P and MacMicking JD: Interferon-induced guanylate-binding proteins in inflammasome activation and host defense. Nat Immunol. 17:481–489. 2016. View Article : Google Scholar : PubMed/NCBI

8 

Luo Y, Jin H, Kim JH and Bae J: Guanylate-binding proteins induce apoptosis of leukemia cells by regulating MCL-1 and BAK. Oncogenesis. 10:542021. View Article : Google Scholar : PubMed/NCBI

9 

Liu YT and Sun ZJ: Turning cold tumors into hot tumors by improving T-cell infiltration. Theranostics. 11:5365–5386. 2021. View Article : Google Scholar : PubMed/NCBI

10 

Galon J and Bruni D: Approaches to treat immune hot, altered and cold tumours with combination immunotherapies. Nat Rev Drug Discov. 18:197–218. 2019. View Article : Google Scholar : PubMed/NCBI

11 

Liu B, Huang R, Fu T, He P, Du C, Zhou W, Xu K and Ren T: GBP2 as a potential prognostic biomarker in pancreatic adenocarcinoma. PeerJ. 9:e114232021. View Article : Google Scholar : PubMed/NCBI

12 

Zhang S, Chen K, Zhao Z, Zhang X, Xu L, Liu T and Yu S: Lower expression of GBP2 associated with less immune cell infiltration and poor prognosis in skin cutaneous melanoma (SKCM). J Immunother. 45:274–283. 2022. View Article : Google Scholar : PubMed/NCBI

13 

Tang JH: Structural study of human guanylate binding protein GBP2. 2019.

14 

Ban T, Heymann JA, Song Z, Hinshaw JE and Chan DC: OPA1 disease alleles causing dominant optic atrophy have defects in cardiolipin-stimulated GTP hydrolysis and membrane tubulation. Hum Mol Genet. 19:2113–2122. 2010. View Article : Google Scholar : PubMed/NCBI

15 

Daumke O and Praefcke GJK: Mechanisms of GTP hydrolysis and conformational transitions in the dynamin superfamily. Biopolymers. 109:e230792018. View Article : Google Scholar : PubMed/NCBI

16 

Roy S, Wang B, Roy K, Tian Y, Bhattacharya M, Williams S and Yin Q: Crystal structures reveal nucleotide-induced conformational changes in G motifs and distal regions in human guanylate-binding protein 2. Commun Biol. 8:2822025. View Article : Google Scholar : PubMed/NCBI

17 

Kravets E, Degrandi D, Ma Q, Peulen TO, Klümpers V, Felekyan S, Kühnemuth R, Weidtkamp-Peters S, Seidel CA and Pfeffer K: Guanylate binding proteins directly attack Toxoplasma gondii via supramolecular complexes. Elife. 5:e114792016. View Article : Google Scholar : PubMed/NCBI

18 

Kravets E, Degrandi D, Weidtkamp-Peters S, Ries B, Konermann C, Felekyan S, Dargazanli JM, Praefcke GJ, Seidel CA, Schmitt L, et al: The GTPase activity of murine Guanylate-binding protein 2 (mGBP2) controls the intracellular localization and recruitment to the parasitophorous vacuole of toxoplasma gondii. J Biol Chem. 287:27452–27466. 2012. View Article : Google Scholar : PubMed/NCBI

19 

Neun R, Richter MF, Staeheli P and Schwemmle M: GTPase properties of the interferon-induced human Guanylate-binding protein 2. FEBS Lett. 390:69–72. 1996. View Article : Google Scholar : PubMed/NCBI

20 

Rajan S, Pandita E, Mittal M and Sau AK: Understanding the lower GMP formation in large GTPase hGBP-2 and role of its individual domains in regulation of GTP hydrolysis. FEBS J. 286:4103–4121. 2019. View Article : Google Scholar : PubMed/NCBI

21 

Honkala AT, Tailor D and Malhotra SV: Guanylate-binding protein 1: An emerging target in inflammation and cancer. Front Immunol. 10:31392019. View Article : Google Scholar : PubMed/NCBI

22 

Cui W, Braun E, Wang W, Tang J, Zheng Y, Slater B, Li N, Chen C, Liu Q, Wang B, et al: Structural basis for GTP-induced dimerization and antiviral function of guanylate-binding proteins. Proc Natl Acad Sci USA. 118:e20222691182021. View Article : Google Scholar : PubMed/NCBI

23 

Abdullah N, Balakumari M and Sau AK: Dimerization and its role in GMP formation by human guanylate binding proteins. Biophys J. 99:2235–2244. 2010. View Article : Google Scholar : PubMed/NCBI

24 

Walker JE, Saraste M, Runswick MJ and Gay NJ: Distantly related sequences in the alpha- and beta-subunits of ATP synthase, myosin, kinases and other ATP-requiring enzymes and a common nucleotide binding fold. EMBO J. 1:945–951. 1982. View Article : Google Scholar : PubMed/NCBI

25 

Wittinghofer A and Vetter IR: Structure-function relationships of the G domain, a canonical switch motif. Annu Rev Biochem. 80:943–971. 2011. View Article : Google Scholar : PubMed/NCBI

26 

Ghosh A, Praefcke GJ, Renault L, Wittinghofer A and Herrmann C: How guanylate-binding proteins achieve assembly-stimulated processive cleavage of GTP to GMP. Nature. 440:101–104. 2006. View Article : Google Scholar : PubMed/NCBI

27 

Olszewski MA, Gray J and Vestal DJ: In silico genomic analysis of the human and murine Guanylate-binding protein (GBP) gene clusters. J Interferon Cytokine Res. 26:328–352. 2006. View Article : Google Scholar : PubMed/NCBI

28 

Ramsauer K, Farlik M, Zupkovitz G, Seiser C, Kröger A, Hauser H and Decker T: Distinct modes of action applied by transcription factors STAT1 and IRF1 to initiate transcription of the IFN-gamma-inducible gbp2 gene. Proc Natl Acad Sci USA. 104:2849–2854. 2007. View Article : Google Scholar : PubMed/NCBI

29 

Michalska A, Blaszczyk K, Wesoly J and Bluyssen HAR: A positive feedback amplifier circuit that regulates interferon (IFN)-Stimulated gene expression and controls type I and type II IFN responses. Front Immunol. 9:11352018. View Article : Google Scholar : PubMed/NCBI

30 

Guimarães DP, Oliveira IM, de Moraes E, Paiva GR, Souza DM, Barnas C, Olmedo DB, Pinto CE, Faria PA, De Moura Gallo CV, et al: Interferon-inducible guanylate binding protein (GBP)-2: A novel p53-regulated tumor marker in esophageal squamous cell carcinomas. Int J Cancer. 124:272–279. 2009. View Article : Google Scholar : PubMed/NCBI

31 

Wu ZH, Cai F and Zhong Y: Comprehensive analysis of the expression and prognosis for GBPs in head and neck squamous cell carcinoma. Sci Rep. 10:60852020. View Article : Google Scholar : PubMed/NCBI

32 

Du CH, Wu YD, Yang K, Liao WN, Ran L, Liu CN, Zhang SZ, Yu K, Chen J, Quan Y, et al: Apoptosis-resistant megakaryocytes produce large and hyperreactive platelets in response to radiation injury. Mil Med Res. 10:662023.PubMed/NCBI

33 

Mondal S, Adhikari N, Banerjee S, Amin SA and Jha T: Matrix metalloproteinase-9 (MMP-9) and its inhibitors in cancer: A minireview. Eur J Med Chem. 194:1122602020. View Article : Google Scholar : PubMed/NCBI

34 

Ngo CC and Man SM: Mechanisms and functions of guanylate-binding proteins and related interferon-inducible GTPases: Roles in intracellular lysis of pathogens. Cell Microbiol. 192017.doi: 10.1111/cmi.12791.

35 

Meunier E and Broz P: Interferon-inducible GTPases in cell autonomous and innate immunity. Cell Microbiol. 18:168–180. 2016. View Article : Google Scholar : PubMed/NCBI

36 

Fu J and Wu H: Structural mechanisms of NLRP3 inflammasome assembly and activation. Annu Rev Immunol. 41:301–316. 2023. View Article : Google Scholar : PubMed/NCBI

37 

Wandel MP, Kim BH, Park ES, Boyle KB, Nayak K, Lagrange B, Herod A, Henry T, Zilbauer M, Rohde J, et al: Guanylate-binding proteins convert cytosolic bacteria into caspase-4 signaling platforms. Nat Immunol. 21:880–891. 2020. View Article : Google Scholar : PubMed/NCBI

38 

Kayagaki N, Stowe IB, Lee BL, O'Rourke K, Anderson K, Warming S, Cuellar T, Haley B, Roose-Girma M, Phung QT, et al: Caspase-11 cleaves gasdermin D for non-canonical inflammasome signalling. Nature. 526:666–671. 2015. View Article : Google Scholar : PubMed/NCBI

39 

Huang S, Dong W, Lin X, Xu K, Li K, Xiong S, Wang Z, Nie X and Bian JS: Disruption of the Na+/K+-ATPase-purinergic P2X7 receptor complex in microglia promotes Stress-induced anxiety. Immunity. 57:495–512.e11. 2024. View Article : Google Scholar : PubMed/NCBI

40 

Garlanda C, Dinarello CA and Mantovani A: The interleukin-1 family: Back to the future. Immunity. 39:1003–1018. 2013. View Article : Google Scholar : PubMed/NCBI

41 

Zhang Y, Liao Y, Hang Q, Sun D and Liu Y: GBP2 acts as a member of the interferon signalling pathway in lupus nephritis. BMC Immunology. 23:442022. View Article : Google Scholar : PubMed/NCBI

42 

Li X, Liu J, Zeng M, Yang K, Zhang S, Liu Y, Yin X, Zhao C, Wang W and Xiao L: GBP2 promotes M1 macrophage polarization by activating the notch1 signaling pathway in diabetic nephropathy. Front Immunol. 14:11276122023. View Article : Google Scholar : PubMed/NCBI

43 

Schori C, Trachsel C, Grossmann J, Barben M, Klee K, Storti F, Samardzija M and Grimm C: A chronic hypoxic response in photoreceptors alters the vitreous proteome in mice. Exp Eye Res. 185:1076902019. View Article : Google Scholar : PubMed/NCBI

44 

An Y, Xu J, Hu X, Xu M, Yang X and Liu T: GBP2 regulates lipid metabolism by inhibiting the HIF-1 pathway to alleviate the progression of allergic rhinitis. Cell Biochem Biophys. 83:1689–1701. 2025. View Article : Google Scholar : PubMed/NCBI

45 

Wang G, Jin S, Ling X, Li Y, Hu Y, Zhang Y, Huang Y, Chen T, Lin J, Ning Z, et al: Proteomic profiling of LPS-induced Macrophage-derived exosomes indicates their involvement in acute liver injury. Proteomics. 19:e18002742019. View Article : Google Scholar : PubMed/NCBI

46 

Huang W, Zhang Y, Zheng B, Ling X, Wang G, Li L and Meng Y: GBP2 upregulated in LPS-stimulated macrophages-derived exosomes accelerates septic lung injury by activating epithelial cell NLRP3 signaling. Int Immunopharmacol. 124:1110172023. View Article : Google Scholar : PubMed/NCBI

47 

Gao R, Ali T, Liu Z, Li A, Hao L, He L, Yu X and Li S: Ceftriaxone averts neuroinflammation and relieves depressive-like behaviors via GLT-1/TrkB signaling. Biochem Biophys Res Commun. 701:1495502024. View Article : Google Scholar : PubMed/NCBI

48 

Ren Y, Yang B, Guo G, Zhang J, Sun Y, Liu D, Guo S, Wu Y, Wang X, Wang S, et al: GBP2 facilitates the progression of glioma via regulation of KIF22/EGFR signaling. Cell Death Discov. 8:2082022. View Article : Google Scholar : PubMed/NCBI

49 

Yu S, Yu X, Sun L, Zheng Y, Chen L, Xu H, Jin J, Lan Q, Chen CC and Li M: GBP2 enhances glioblastoma invasion through Stat3/fibronectin pathway. Oncogene. 39:5042–5055. 2020. View Article : Google Scholar : PubMed/NCBI

50 

Verdugo E, Puerto I and Medina MÁ: An update on the molecular biology of glioblastoma, with clinical implications and progress in its treatment. Cancer Commun (Lond). 42:1083–111. 2022. View Article : Google Scholar : PubMed/NCBI

51 

Meng K, Li YY, Liu DY, Hu LL, Pan YL, Zhang CZ and He QY: A five-protein prognostic signature with GBP2 functioning in immune cell infiltration of clear cell renal cell carcinoma. Comput Struct Biotechnol J. 21:2621–2630. 2023. View Article : Google Scholar : PubMed/NCBI

52 

Rahvar F, Salimi M and Mozdarani H: Plasma GBP2 promoter methylation is associated with advanced stages in breast cancer. Genet Mol Biol. 43:e201902302020. View Article : Google Scholar : PubMed/NCBI

53 

Godoy P, Cadenas C, Hellwig B, Marchan R, Stewart J, Reif R, Lohr M, Gehrmann M, Rahnenführer J, Schmidt M and Hengstler JG: Interferon-inducible guanylate binding protein (GBP2) is associated with better prognosis in breast cancer and indicates an efficient T cell response. Breast Cancer. 21:491–499. 2014. View Article : Google Scholar : PubMed/NCBI

54 

Li NN, Qiu XT, Xue JS, Yi LM, Chen ML and Huang ZJ: Predicting the prognosis and immunotherapeutic response of Triple-negative breast cancer by constructing a prognostic model based on CD8+ T Cell-related immune genes. Biomed Environ Sci. 37:581–593. 2024.PubMed/NCBI

55 

Ji G, Luo B, Chen L, Shen G and Tian T: GBP2 is a favorable prognostic marker of skin cutaneous melanoma and affects its progression via the Wnt/β-catenin pathway. Ann Clin Lab Sci. 51:772–782. 2021.PubMed/NCBI

56 

Zheng W, Ye S, Liu B, Liu D, Yan R, Guo H, Yu H, Hu X, Zhao H, Zhou K and Li G: Crosstalk between GBP2 and M2 macrophage promotes the ccRCC progression. Cancer Science. 115:3570–3586. 2024. View Article : Google Scholar : PubMed/NCBI

57 

Tian Y, Wang H, Guan W, Tu X, Zhang X, Sun Y, Qian C, Song X, Peng B and Cui X: GBP2 serves as a novel prognostic biomarker and potential immune microenvironment indicator in renal cell carcinoma. Mol Carcinog. 61:1082–1098. 2022. View Article : Google Scholar : PubMed/NCBI

58 

AmeliMojarad M, AmeliMojarad M and Cui X: Weighted gene co-expression network analysis identified GBP2 connected to PPARα activity and liver cancer. Sci Rep. 14:207452024. View Article : Google Scholar : PubMed/NCBI

59 

Wang H, Zhou Y, Zhang Y, Fang S, Zhang M, Li H, Xu F, Liu L, Liu J, Zhao Q and Wang F: Subtyping of microsatellite stability colorectal cancer reveals guanylate binding protein 2 (GBP2) as a potential immunotherapeutic target. J Immunother Cancer. 10:e0043022022. View Article : Google Scholar : PubMed/NCBI

60 

Qi F, Gao N, Li J, Zhou C, Jiang J, Zhou B, Guo L, Feng X, Ji J, Cai Q, et al: A multidimensional recommendation framework for identifying biological targets to aid the diagnosis and treatment of liver metastasis in patients with colorectal cancer. Mol Cancer. 23:2392024. View Article : Google Scholar : PubMed/NCBI

61 

Wang Y, Pan J, An F, Chen K, Chen J, Nie H, Zhu Y, Qian Z and Zhan Q: GBP2 is a prognostic biomarker and associated with immunotherapeutic responses in gastric cancer. BMC Cancer. 23:9252023. View Article : Google Scholar : PubMed/NCBI

62 

Xiong J, Huang J, Xu H, Wu Q, Zhao J, Chen Y, Fan G, Guan H, Xiao R, He Z, et al: CpG-based nanovaccines enhance ovarian cancer immune response by Gbp2-mediated remodeling of tumor-associated macrophages. Adv Sci (Weinh). 12:e24128812025. View Article : Google Scholar : PubMed/NCBI

63 

Feng D, Zhu W, Shi X, Wang Z, Wei W, Wei Q, Yang L and Han P: Immune-related gene index predicts metastasis for prostate cancer patients undergoing radical radiotherapy. Exp Hematol Oncol. 12:82023. View Article : Google Scholar : PubMed/NCBI

64 

Uribe ML, Marrocco I and Yarden Y: EGFR in cancer: Signaling mechanisms, drugs, and acquired resistance. Cancers (Basel). 13:27482021. View Article : Google Scholar : PubMed/NCBI

65 

Kim J, Kundu M, Viollet B and Guan KL: AMPK and mTOR regulate autophagy through direct phosphorylation of Ulk1. Nat Cell Biol. 13:132–141. 2011. View Article : Google Scholar : PubMed/NCBI

66 

Zhang W, Tang X, Peng Y, Xu Y, Liu L and Liu S: GBP2 enhances paclitaxel sensitivity in triple-negative breast cancer by promoting autophagy in combination with ATG2 and inhibiting the PI3K/AKT/mTOR pathway. Int J Oncol. 64:342024. View Article : Google Scholar : PubMed/NCBI

67 

Li X, He S and Ma B: Autophagy and autophagy-related proteins in cancer. Mol Cancer. 19:122020. View Article : Google Scholar : PubMed/NCBI

68 

Zhang H, Sun Z, Li Y, Fan D and Jiang H: MicroRNA-200c binding to FN1 suppresses the proliferation, migration and invasion of gastric cancer cells. Biomed Pharmacother. 88:285–292. 2017. View Article : Google Scholar : PubMed/NCBI

69 

Shibata K, Kikkawa F, Nawa A, Thant AA, Naruse K, Mizutani S and Hamaguchi M: Both focal adhesion kinase and c-Ras are required for the enhanced matrix metalloproteinase 9 secretion by fibronectin in ovarian cancer cells. Cancer Res. 58:900–1093. 1998.PubMed/NCBI

70 

Xu TP, Huang MD, Xia R, Liu XX, Sun M, Yin L, Chen WM, Han L, Zhang EB, Kong R, et al: Decreased expression of the long non-coding RNA FENDRR is associated with poor prognosis in gastric cancer and FENDRR regulates gastric cancer cell metastasis by affecting fibronectin1 expression. J Hematol Oncol. 7:632014. View Article : Google Scholar : PubMed/NCBI

71 

Xue W, Yang L, Chen C, Ashrafizadeh M, Tian Y and Sun R: Wnt/β-catenin-driven EMT regulation in human cancers. Cell Mol Life Sci. 81:792024. View Article : Google Scholar : PubMed/NCBI

72 

Peña-Blanco A and García-Sáez AJ: Bax, Bak and beyond-mitochondrial performance in apoptosis. FEBS J. 285:416–431. 2018. View Article : Google Scholar : PubMed/NCBI

73 

Zhang J, Zhang Y, Wu W, Wang F, Liu X, Shui G and Nie C: Guanylate-binding protein 2 regulates Drp1-mediated mitochondrial fission to suppress breast cancer cell invasion. Cell Death Dis. 8:e31512017. View Article : Google Scholar : PubMed/NCBI

74 

Liu W, Chen Y, Xie H, Guo Y, Ren D, Li Y, Jing X, Li D, Wang X, Zhao M, et al: TIPE1 suppresses invasion and migration through down-regulating Wnt/β-catenin pathway in gastric cancer. J Cell Mol Med. 22:1103–1117. 2018. View Article : Google Scholar : PubMed/NCBI

75 

Wang J, Min H, Hu B, Xue X and Liu Y: Guanylate-binding protein-2 inhibits colorectal cancer cell growth and increases the sensitivity to paclitaxel of paclitaxel-resistant colorectal cancer cells by interfering Wnt signaling. J Cell Biochem. 121:1250–129. 2020. View Article : Google Scholar : PubMed/NCBI

76 

Song JX, Wang Y, Hua ZP, Huang Y, Hu LF, Tian MR, Qiu L, Liu H and Zhang J: FATS inhibits the Wnt pathway and induces apoptosis through degradation of MYH9 and enhances sensitivity to paclitaxel in breast cancer. Cell Death Dis. 15:8352024. View Article : Google Scholar : PubMed/NCBI

77 

Zhang SW, Feng TB, Ning YL, Zhang XH and Qi CJ: Guanylate-binding protein 2 regulates the maturation of mouse dendritic cells induced by β-glucan. Xi Bao Yu Fen Zi Mian Yi Xue Za Zhi. 33:1153–1159. 2017.(In Chinese). PubMed/NCBI

78 

Xu X, Ding X, Wang Z, Ye S, Xu J, Liang Z, Luo R, Xu J, Li X and Ren Z: GBP2 inhibits pathological angiogenesis in the retina via the AKT/mTOR/VEGFA axis. Microvasc Res. 154:1046892024. View Article : Google Scholar : PubMed/NCBI

79 

Du F, Liu M, Wang J, Hu L, Zeng D, Zhou S, Zhang L, Wang M, Xu X, Li C, et al: Metformin coordinates with mesenchymal cells to promote VEGF-mediated angiogenesis in diabetic wound healing through Akt/mTOR activation. Metabolism. 140:1553982023. View Article : Google Scholar : PubMed/NCBI

80 

Inoki K, Li Y, Zhu T, Wu J and Guan KL: TSC2 is phosphorylated and inhibited by Akt and suppresses mTOR signalling. Nat Cell Biol. 4:648–6457. 2002. View Article : Google Scholar : PubMed/NCBI

81 

Ruchko MV, Gorodnya OM, Pastukh VM, Swiger BM, Middleton NS, Wilson GL and Gillespie MN: Hypoxia-induced oxidative base modifications in the VEGF hypoxia-response element are associated with transcriptionally active nucleosomes. Free Radic Biol Med. 46:352–359. 2009. View Article : Google Scholar : PubMed/NCBI

82 

Wenger RH, Stiehl DP and Camenisch G: Integration of oxygen signaling at the consensus HRE. Sci STKE. 2005:re122005. View Article : Google Scholar : PubMed/NCBI

83 

Balasubramanian S, Fan M, Messmer-Blust AF, Yang CH, Trendel JA, Jeyaratnam JA, Pfeffer LM and Vestal DJ: The interferon-gamma-induced GTPase, mGBP-2, inhibitsc (TNF-alpha) induction of matrix metalloproteinase-9 (MMP-9) by inhibiting NF-kappaB and Rac protein. J Biol Chem. 286:20054–20064. 2011. View Article : Google Scholar : PubMed/NCBI

84 

Raskov H, Orhan A, Christensen JP and Gögenur I: Cytotoxic CD8+ T cells in cancer and cancer immunotherapy. Br J Cancer. 124:359–367. 2021. View Article : Google Scholar : PubMed/NCBI

85 

Ye S, Li S, Qin L, Zheng W, Liu B, Li X, Ren Z, Zhao H, Hu X, Ye N and Li G: GBP2 promotes clear cell renal cell carcinoma progression through immune infiltration and regulation of PD-L1 expression via STAT1 signaling. Oncol Rep. 49:492023. View Article : Google Scholar : PubMed/NCBI

86 

Kutsch M and Coers J: Human guanylate binding proteins: Nanomachines orchestrating host defense. FEBS J. 288:5826–5849. 2021. View Article : Google Scholar : PubMed/NCBI

87 

Chakraborty S, Kasirajan A, Mariappan V, Green SR and Pillai AKB: Guanylate binding proteins (GBPs) as novel therapeutic targets against single-stranded RNA viruses. Mol Biol Rep. 52:7802025. View Article : Google Scholar : PubMed/NCBI

Related Articles

  • Abstract
  • View
  • Download
  • Twitter
Copy and paste a formatted citation
Spandidos Publications style
Liu Z, Peng S and Ouyang J: Molecular mechanisms and functions of guanylate‑binding protein 2 in inflammation and cancer (Review). Oncol Lett 30: 534, 2025.
APA
Liu, Z., Peng, S., & Ouyang, J. (2025). Molecular mechanisms and functions of guanylate‑binding protein 2 in inflammation and cancer (Review). Oncology Letters, 30, 534. https://doi.org/10.3892/ol.2025.15280
MLA
Liu, Z., Peng, S., Ouyang, J."Molecular mechanisms and functions of guanylate‑binding protein 2 in inflammation and cancer (Review)". Oncology Letters 30.5 (2025): 534.
Chicago
Liu, Z., Peng, S., Ouyang, J."Molecular mechanisms and functions of guanylate‑binding protein 2 in inflammation and cancer (Review)". Oncology Letters 30, no. 5 (2025): 534. https://doi.org/10.3892/ol.2025.15280
Copy and paste a formatted citation
x
Spandidos Publications style
Liu Z, Peng S and Ouyang J: Molecular mechanisms and functions of guanylate‑binding protein 2 in inflammation and cancer (Review). Oncol Lett 30: 534, 2025.
APA
Liu, Z., Peng, S., & Ouyang, J. (2025). Molecular mechanisms and functions of guanylate‑binding protein 2 in inflammation and cancer (Review). Oncology Letters, 30, 534. https://doi.org/10.3892/ol.2025.15280
MLA
Liu, Z., Peng, S., Ouyang, J."Molecular mechanisms and functions of guanylate‑binding protein 2 in inflammation and cancer (Review)". Oncology Letters 30.5 (2025): 534.
Chicago
Liu, Z., Peng, S., Ouyang, J."Molecular mechanisms and functions of guanylate‑binding protein 2 in inflammation and cancer (Review)". Oncology Letters 30, no. 5 (2025): 534. https://doi.org/10.3892/ol.2025.15280
Follow us
  • Twitter
  • LinkedIn
  • Facebook
About
  • Spandidos Publications
  • Careers
  • Cookie Policy
  • Privacy Policy
How can we help?
  • Help
  • Live Chat
  • Contact
  • Email to our Support Team