CXCL2, also known as C-X-C motif chemokine ligand 2 or macrophage inflammatory protein 2 alpha (MIP-2α), is a small secreted cytokine belonging to the ELR+ subfamily of CXC chemokines that functions primarily as a potent chemoattractant for neutrophils.[1][2]Encoded by the CXCL2 gene located on human chromosome 4q21, the protein is synthesized as a 107-amino-acid precursor that undergoes proteolytic cleavage to yield a mature form spanning amino acids 5–73, enabling its role in guiding leukocyte migration during immune responses.[2] This chemokine exhibits both chemokine and cytokine activities, interacting specifically with the G protein-coupled receptor CXCR2 on target cells to mediate signaling pathways such as ERK/MAPK, NF-κB, PI3K/AKT, and JAK/STAT3, which drive neutrophil recruitment and activation at sites of inflammation.[1][2]CXCL2 is broadly expressed across human tissues, with the highest levels observed in the liver (RPKM 52.2) and gallbladder (RPKM 27.8), and is produced by various cell types including monocytes, macrophages, endothelial cells, and epithelial cells.[1] Its expression is tightly regulated and upregulated by proinflammatory stimuli such as tumor necrosis factor alpha (TNFα), interleukin-1 beta (IL-1β), and lipopolysaccharide (LPS) through NF-κB and MAPK pathways, allowing rapid deployment in response to infection or injury.[2]In physiological contexts, CXCL2 contributes to antimicrobial humoral immunity, acute inflammation, and tissue repair by facilitating neutrophil infiltration and coordinating early immune defense mechanisms.[1][2] However, dysregulated CXCL2 signaling is implicated in numerous pathologies, including promoting tumor progression and metastasis in cancers such as hepatocellular carcinoma and breast cancer via modulation of the tumor microenvironment, as well as exacerbating chronic inflammatory conditions like atherosclerosis, obesity-associated inflammation, and inflammatory bowel disease.[1][2] Additionally, CXCL2 can form heterodimers with related chemokines like CXCL1, enhancing its chemoattractant potency in bacterial infection models.[3]Due to its central role in neutrophil biology and disease, CXCL2 has emerged as a potential biomarker for prognosis in inflammatory and oncologic disorders, with ongoing research exploring CXCR2 antagonists as therapeutic targets to mitigate excessive inflammation or tumor-associated immune evasion.[2]
Discovery and Nomenclature
Historical Discovery
CXCL2 was first identified as part of the growth-regulated oncogene (GRO) family in the late 1980s, emerging from studies on genes overexpressed in transformed cells. The founding member of the family, GROα (also known as CXCL1), was cloned in 1987 from transformed Chinese hamster fibroblasts and human breast epithelial cells, where its expression was found to correlate with cellular growth regulation. This discovery laid the groundwork for recognizing the GRO genes as encoding cytokines with potential roles in inflammation and oncogenesis.In 1988, further characterization linked GRO expression to inflammatory stimuli, as demonstrated by Haskill et al., who showed that adherence of human monocytes to extracellular matrix selectively induced mRNA for GRO and other mediators, suggesting its involvement in monocyte activation during inflammation. Concurrently, the murine homolog of CXCL2, known as macrophage inflammatory protein-2 (MIP-2), was isolated and characterized by Wolpe et al. from endotoxin-stimulated rabbit macrophages; MIP-2 was identified as a heparin-binding protein with potent chemotactic activity for human neutrophils, marking it as a key inflammatorychemokine in rodents.[4][5]The specific identification of human CXCL2, termed GROβ or GRO2, occurred in 1990 when Haskill et al. cloned three related human GRO genes, revealing that GROβ shares approximately 90% amino acid identity with GROα and is part of the CXC chemokine subfamily. That same year, Iida and Grotendorst reported the cloning and sequencing of a novel GRO transcript (GROβ) from activated human monocytes, confirming its expression in leukocytes and wound tissue, and establishing its chemotactic properties for neutrophils through in vitro assays.[6]Genetic mapping efforts in 1990 localized the GRO gene cluster, including CXCL2, to humanchromosome 4q21 using a GROα cDNA probe that hybridized to all three genes in somatic cell hybrids and fluorescence in situ hybridization, providing early insights into its genomic organization within the chemokine superfamily.[6]
Alternative Names and Classification
CXCL2 is known by several alternative names, including growth-regulated oncogene 2 (GRO2), GRO-beta, melanoma growth stimulatory activity beta (MGSA-beta), and macrophage inflammatory protein 2-alpha (MIP-2α) in humans, while its murine homolog is also designated MIP-2α.[7][1][8]Originally identified as part of the growth-related oncogene (GRO) family, CXCL2 is classified within the CXC subfamily of chemokines, defined by a conserved motif featuring two cysteines separated by one amino acid (CXC).[9][7]This CXC subfamily differs from other chemokine classes, such as CC (adjacent cysteines), C (one cysteine), and CX3C (three amino acids between cysteines), based on the arrangement of their conserved cysteine residues.[9]CXCL2 specifically belongs to the ELR+ subset of CXC chemokines, distinguished by the glutamic acid-leucine-arginine (ELR) motif positioned immediately N-terminal to the CXC sequence.[9]It exhibits 90% amino acidsequence identity with CXCL1 and 86% identity with CXCL3.[9]
Gene and Protein Structure
Genomic Organization
The CXCL2 gene, with official Gene ID 2920, is located on the long arm of human chromosome 4 at cytogenetic band 4q13.3, spanning genomic coordinates 74,097,040 to 74,099,196 on the reverse strand (GRCh38 assembly).[1] This positions it within a compact chemokinegene cluster on chromosome 4q, alongside closely related CXC motif chemokines such as CXCL1 (encoding GROα) and CXCL3 (encoding GROγ), which share structural and functional similarities in inflammatory signaling.[10] The cluster arrangement facilitates coordinated regulation of these genes during immune responses.The gene itself encompasses approximately 2,157 base pairs and comprises 4 exons, with the mature mRNA featuring a 3' untranslated region of about 700 bp that terminates at the polyadenylation site.[9] Upstream of the coding region, the promoter contains conserved NF-κB binding sites, including a consensus sequence (GGAA GTTCCC) around position -640 relative to the transcription start site, which is essential for transcriptional activation in response to proinflammatory cytokines like IL-1β.[11] Mutation of this NF-κB site significantly impairs IL-1β-induced expression, highlighting its role in inflammatory gene control.[11]Several single nucleotide polymorphisms (SNPs) have been identified within the CXCL2 locus that influence gene expression levels and are associated with variations in inflammatory responses, such as altered white blood cell counts in genome-wide association studies.[12] These variants contribute to inter-individual differences in chemokine production during infection or tissue injury.
Protein Sequence and Structure
The human CXCL2 protein is synthesized as a precursor of 107 amino acids, which undergoes cleavage of a 34-amino-acid N-terminal signal peptide to yield the mature form consisting of 73 amino acids.[7][2]A hallmark feature of the mature CXCL2 is the ELR motif (Glu-Leu-Arg) located at positions 9-11, which is conserved among ELR-positive CXC chemokines and contributes to receptor specificity.[9] The protein also contains four conserved cysteine residues that form two intramolecular disulfide bonds: Cys7-Cys34 and Cys10-Cys36 (numbered relative to the mature sequence), stabilizing the overall fold.[7][13]The three-dimensional structure of CXCL2 has been determined for its murine ortholog MIP-2 (which shares high sequence similarity), featuring both monomeric and dimeric forms. The monomer adopts a compact fold characteristic of CXC chemokines, comprising an N-terminal unstructured loop, a three-stranded antiparallel Greek key β-sheet (strands connected by loops), and a C-terminal α-helix that packs against the β-sheet.[13][14] In the dimer, the interface is mediated primarily by the N-terminal loops and helices from each monomer, facilitating interactions relevant to its localization.[13] More recently, a cryo-EM structure of the human CXCL2 in complex with its receptor CXCR2 (PDB: 8XVU, 2025) reveals the ligand-receptor interaction, confirming the conserved chemokine fold and highlighting the role of the ELR motif in binding.[15]CXCL2 possesses glycosaminoglycan (GAG) binding sites, primarily involving basic residues in the N-terminal region and the C-terminal helix, enabling immobilization on the extracellular matrix and formation of chemokine gradients.[16] CXCL2 exhibits approximately 90% amino acid sequence identity to CXCL1, reflecting their close evolutionary relationship within the ELR+ CXC subfamily.[17]
Expression and Regulation
Cellular Sources
CXCL2 is primarily produced by activated monocytes and macrophages, which serve as key sources during inflammatory responses.[18] These immune cells secrete CXCL2 in response to stimuli such as lipopolysaccharide (LPS) and tumor necrosis factor (TNF), contributing to neutrophilrecruitment at sites of infection or injury.[19] Neutrophils themselves also act as a significant cellular source, particularly in TNF-stimulated tissues, where they produce CXCL2 to facilitate their own transendothelial migration.[20]In addition to hematopoietic cells, non-immune cells including endothelial cells, fibroblasts, and epithelial cells express CXCL2 upon stimulation. Endothelial cells release CXCL2 during inflammatory conditions, aiding in leukocyte adhesion and extravasation.[2] Fibroblasts and epithelial cells, such as those in the lung or skin, upregulate CXCL2 production in response to cytokines like TNF or interleukin-1 (IL-1), supporting local immune responses.[21] Single-cell RNA sequencing data indicate enhanced expression in mononuclear phagocytes, endothelial cells, fibroblasts, and various epithelial subtypes under inflammatory contexts.[21]Basal expression of CXCL2 is low in most immune cells but becomes markedly upregulated in inflamed tissues, including the lung, liver, and adipose tissue. In the liver, CXCL2 shows the highest constitutive expression among tissues (RPKM 52.2), while in adipose tissue, it contributes to obesity-associated inflammation.[1] Upregulation occurs at inflammation sites, such as alveolar macrophages in the lung during infection.[22]In murine models, the ortholog MIP-2 (CXCL2) is primarily produced by macrophages and monocytes, with additional sources in epithelial cells and hepatocytes following injury or infection.[23] This species-specific pattern highlights macrophages as a conserved primary source across mammals. Cytokine stimulation, such as by TNF or IL-1, briefly induces CXCL2 in these cells.[20]
Regulatory Mechanisms
The expression of CXCL2 is tightly controlled at the transcriptional level, primarily through activation by pro-inflammatory cytokines such as interleukin-1β (IL-1β) and tumor necrosis factor-α (TNF-α). These cytokines induce CXCL2 transcription by promoting the nuclear translocation and DNA binding of NF-κB subunits p65 and p50 to consensus κB sites in the CXCL2 promoter, leading to rapid upregulation of mRNA levels, often peaking within 3 hours of stimulation.[11]STAT1, particularly when serine-phosphorylated at position 727, cooperates with NF-κB to enhance this process, as evidenced by reduced CXCL2 expression upon STAT1 inhibition.[11] Additionally, the AP-1 transcription factor complex contributes to CXCL2 induction, often in synergy with NF-κB, although it can also participate in negative regulation under certain anti-inflammatory contexts, such as pregnane X receptor (PXR)-mediated suppression.[24][25]Post-transcriptional mechanisms further modulate CXCL2 levels, influencing mRNA stability and translation. The RNA-binding protein HuR associates with the 3' untranslated region (UTR) of CXCL2 mRNA in cytokine-stimulated airway epithelial cells, potentially facilitating its stabilization, though direct enhancement of stability has not been consistently observed for CXCL2 specifically.[26] In contrast, microRNAs such as miR-146a act as negative regulators; knockout of miR-146a in mouse models results in elevated CXCL2 expression and exacerbated inflammation, indicating miR-146a suppresses CXCL2 translation or stability during immune responses.[27]Environmental stimuli, including bacterial lipopolysaccharide (LPS), hypoxia, and oxidative stress, serve as potent inducers of CXCL2 expression, often converging on the NF-κB pathway. LPS, recognized by Toll-like receptor 4 (TLR4), triggers robust CXCL2 production in renal tubular cells and macrophages via downstream activation of NF-κB, with sirtuin 2 (SIRT2) modulating this response to prevent excessive inflammation.[28]Hypoxia upregulates CXCL2 in various cell types, such as prostate cancer cells and tumor-associated macrophages, through hypoxia-inducible factor (HIF)-1 and HIF-2 binding to hypoxia response elements (HREs) in the promoter, though effects can vary by cell context (e.g., downregulation in some melanoma lines).[29]Oxidative stress, including exposure to oxidized phospholipids, cooperatively induces CXCL2 alongside cytokines like TNF-α, promoting chemokine release in inflammatory settings.[30]Feedback loops provide dynamic autoregulation of CXCL2 activity, particularly involving neutrophils. Neutrophils produce CXCL2 in an autocrine manner upon immune complex stimulation, amplifying their own recruitment and effector functions (e.g., reactive oxygen species production) via CXCR2 signaling, but high CXCL2 levels trigger negative feedback by downregulating CXCR2 surface expression to limit excessive inflammation.[31] Additionally, cis-acting long non-coding RNA lnc-Cxcl2, induced in lung epithelial cells during viralinfection, restrains CXCL2 expression by binding the promoter and recruiting ribonucleoprotein La to reduce chromatin accessibility, thereby curbing neutrophil-mediated lungdamage in a feedback manner.[32]
Biological Functions
Receptor Interactions
CXCL2 primarily binds to the chemokine receptor CXCR2, a seven-transmembrane G-protein-coupled receptor predominantly expressed on neutrophils, with high affinity characterized by a dissociation constant (Kd) of approximately 1-10 nM.[33] This interaction is crucial for CXCL2's role in neutrophil chemotaxis and activation, as CXCR2 serves as the main signaling receptor for ELR+ CXC chemokines like CXCL2.[34]In addition to CXCR2, CXCL2 exhibits lower affinity binding to CXCR1, another G-protein-coupled receptor on neutrophils, with a Kd typically in the range of 10-100 nM, making CXCR1 a secondary receptor.[35] CXCL2 is also recognized by the atypical chemokine receptor ACKR1 (also known as DARC), which functions as a scavenger without initiating signaling, thereby regulating CXCL2 levels by internalizing and degrading it to prevent excessive inflammation.[36]The binding mechanism of CXCL2 to CXCR2 relies on the N-terminal ELR (Glu-Leu-Arg) motif, which is essential for receptor activation and distinguishes ELR+ chemokines from non-activating variants. Recent cryo-EM structures (as of 2023) have elucidated the atomic details of CXCR2 activation by ELR+ chemokines like CXCL2, revealing key interactions in the receptor's orthosteric pocket.[37][38] Furthermore, CXCL2 interacts with glycosaminoglycans (GAGs) on the endothelial surface, enhancing its haptotactic presentation to CXCR2 and facilitating localized gradient formation for efficient receptor engagement.[39]Upon binding to CXCR2, CXCL2 induces dissociation of the heterotrimeric G-protein, specifically coupling to Gαi subunits, which inhibits adenylyl cyclase and mobilizes intracellular calcium.[40] This initial event triggers downstream activation of phosphoinositide 3-kinase (PI3K) and mitogen-activated protein kinase (MAPK) pathways, initiating rapid cellular responses.[41]
Cellular Effects
CXCL2 primarily exerts its cellular effects through binding to the CXCR2 receptor, inducing directed migration of neutrophils in response to concentration gradients typically ranging from 1 to 10 nM.[42] This chemotactic activity enables neutrophils to sense and follow haptotactic or soluble gradients formed by CXCL2, facilitating rapid recruitment to sites of inflammation.[36] At these concentrations, CXCL2 triggers intracellular signaling cascades, including calcium mobilization and actin polymerization, which drive the polarization and directional motility essential for neutrophil traversal of endothelial barriers.[43]In hematopoietic cells, CXCL2 signaling via CXCR2 suppresses the proliferation of myeloid progenitor cells, thereby maintaining steady-state hematopoiesis in the bone marrow. This inhibitory effect helps regulate the balance between progenitor expansion and differentiation, preventing excessive myeloid output under homeostatic conditions.[44] Conversely, CXCL2 promotes the mobilization of hematopoietic stem and progenitor cells into the peripheral blood, enhancing their release from bone marrow niches through disruption of retention signals and synergy with other mobilizing agents.[45]On endothelial cells, CXCL2 activates CXCR2 to upregulate adhesion molecules such as ICAM-1, promoting neutrophil firm adhesion and transendothelial migration while also stimulating endothelial proliferation and sprouting for angiogenic responses.[46] Effects on other immune cells are minimal; CXCL2 elicits weak chemotaxis in T cells and negligible responses in eosinophils due to low CXCR2 expression on these populations.[47]The functional dichotomy between CXCL2 monomers and dimers further modulates these effects: dimers exhibit higher affinity for glycosaminoglycans (GAGs) on cell surfaces and extracellular matrix, enabling immobilization and formation of stable gradients for haptotaxis, whereas monomers predominate in soluble forms that directly activate CXCR2 for transient receptor signaling and chemotaxis.[42] This monomer-dimer equilibrium ensures precise spatiotemporal control of neutrophil guidance without overactivation.[48]
Physiological Roles
Immune Response
CXCL2, also known as macrophage inflammatory protein-2 (MIP-2), plays a pivotal role in innate immunity by facilitating the recruitment of neutrophils to sites of bacterial infection, thereby enhancing pathogen clearance. Upon detection of bacterial components such as lipopolysaccharide (LPS), macrophages and other resident cells produce CXCL2, which binds to the CXCR2 receptor on neutrophils, inducing chemotaxis and directed migration to the infected tissue.[49] In murine models of pneumonia caused by Legionella pneumophila, disruption of CXCR2 signaling, including responses to CXCL2, impairs neutrophil accumulation in the lungs, leading to reduced bacterial clearance and increased mortality.[50] Similarly, during LPS-induced peritonitis, early neutrophil influx driven by CXCL2 from mast cells and macrophages is essential for limiting bacterial dissemination and promoting host survival.[51] This process ensures rapid deployment of neutrophils, whose antimicrobial granules and oxidative bursts effectively combat invading pathogens in healthy immune responses.[52]In acute inflammation, CXCL2 amplifies monocyte and macrophage responses by contributing to the orchestrated influx of these cells, sustaining the inflammatory cascade necessary for tissue defense. Although primarily recognized for neutrophil attraction, CXCL2 also engages CXCR2 on monocytes, promoting their recruitment to inflammatory foci where they differentiate into macrophages capable of phagocytosis and cytokine production.[53] Secreted by activated monocytes and macrophages themselves, CXCL2 forms a positive feedback loop that enhances the overall leukocyte response during early inflammatory phases, as observed in models of sterile and infectious acute lung injury.[54] This amplification supports the transition from innate neutrophil-dominated clearance to broader mononuclear phagocyte involvement, optimizing pathogen containment without excessive tissue damage in physiological contexts.[55]CXCL2 contributes to hematopoietic regulation by helping maintain stem cell quiescence amid immune challenges, ensuring a balanced supply of leukocytes during stress. Through interactions with CXCR2 on hematopoietic stem and progenitor cells (HSPCs), CXCL2 ligands support the quiescent state of these cells within the bone marrow niche, preventing premature exhaustion during inflammatory demands.[56] In the CXCR2 signaling network, CXCL2 aids in HSPC survival and self-renewal, particularly under conditions of immune activation where rapid leukocyte production is required, thus preserving long-term hematopoietic integrity.[57]CXCL2 integrates with other chemokines, such as CXCL1 and CXCL8 (IL-8), to synergistically drive robust leukocyte influx at infection sites, enhancing the efficiency of immune orchestration. These ELR+CXC chemokines share the CXCR2 receptor and exhibit complementary expression patterns, with CXCL2 accentuating the chemotactic gradient established by CXCL1 and CXCL8 to promote sequential neutrophil waves.[58] In experimental models of inflammation, combined action of CXCL1, CXCL2, and CXCL8 results in amplified neutrophil and monocyte migration compared to individual chemokines, underscoring their cooperative role in mounting a coordinated innate response.[36] This synergy ensures precise spatiotemporal control of leukocyte trafficking, vital for effective antimicrobial defense.[59]
Wound Healing and Angiogenesis
CXCL2 plays a pivotal role in wound healing by facilitating the recruitment of neutrophils to the injury site, primarily through its interaction with the CXCR2 receptor on these cells. This recruitment occurs during the early inflammatory phase, where neutrophils perform essential functions such as phagocytosis of cellular debris and pathogens to prevent infection and prepare the wound bed for subsequent repair processes.[60] In experimental models, sustained CXCL2 expression, as observed in diabetic wounds, prolongs neutrophil presence, highlighting its influence on the temporal dynamics of inflammation resolution.[60]Beyond neutrophil mobilization, CXCL2 indirectly supports fibroblast involvement in wound healing by contributing to the inflammatory milieu that promotes granulation tissue formation. Neutrophils recruited by CXCL2 release additional mediators that enhance fibroblast migration and proliferation, aiding in extracellular matrix deposition and remodeling during the proliferative phase.[60] This process ensures effective tissue reconstruction, with studies demonstrating impaired woundclosure in contexts where CXCR2 signaling is disrupted, underscoring CXCL2's necessity for coordinated cellular responses.[61]In angiogenesis, CXCL2 acts as a potent pro-angiogenic factor by binding to CXCR2 on endothelial cells, inducing their chemotaxis, proliferation, and tube formation to support new vessel development. This mechanism is particularly evident in repair contexts, where CXCR2 neutralization reduces vascularization and delays healing.[62] Although CXCL2's effects can operate independently of vascular endothelial growth factor (VEGF), it enhances VEGF-regulated angiogenesis in certain physiological settings, amplifying endothelial activation and vascular remodeling.[63]The pro-angiogenic activity of CXCL2 stems from its ELR+ motif, a structural feature shared by other CXC chemokines (such as CXCL1 and CXCL8) that enables CXCR2-mediated promotion of vascular growth. In contrast, ELR- CXC chemokines like CXCL4 exhibit anti-angiogenic properties by inhibiting endothelial proliferation and migration, thereby maintaining a balance in angiogenic responses during tissue repair.[62] This dichotomy ensures that CXCL2-driven angiogenesis is contextually regulated to support healing without excessive vascularization.[64]
Pathological Implications
Role in Inflammation and Infection
CXCL2 plays a pivotal role in acute infections by facilitating neutrophil recruitment to sites of bacterial invasion, thereby enhancing host defense against pathogens such as those causing pneumonia and sepsis. In lipopolysaccharide (LPS)-induced lung inflammation models, CXCL2 expression is markedly upregulated in the lungs, promoting the influx of polymorphonuclear leukocytes (PMNs) via interaction with CXCR2 receptors on endothelial cells.[65] Similarly, in murine models of Klebsiella pneumoniae infection and septic peritonitis, CXCL2 is essential for effective neutrophilmigration to infected tissues, where its absence leads to exacerbated bacterial dissemination and higher mortality rates.[66]In chronic inflammatory conditions like rheumatoid arthritis (RA), CXCL2 levels are elevated in synovial fluids, particularly in anti-citrullinated protein antibody (ACPA)-positive patients, contributing to sustained neutrophil accumulation in affected joints. This chemokine, primarily secreted by activated neutrophils within the synovium, amplifies further recruitment through autocrine and paracrine signaling, exacerbating joint damage and inflammation.[67][68]CXCL2 is also upregulated in systemic lupus erythematosus (SLE), where it drives aberrant neutrophil activation via the PI3K/AKT/NF-κB signaling pathway, as demonstrated in recent transcriptomic analyses of patient-derived neutrophils. This axis promotes proinflammatory cytokine production and neutrophil extracellular trap formation, intensifying autoimmune-mediated tissue damage in SLE.[69]In experimental infection models, CXCL2 is critical for pathogen control during Escherichia coli challenges, with its expression upregulated in response to bacterial virulence factors to orchestrate early neutrophil responses in renal and systemic sites.[70] In neonatal murine sepsis induced by E. coli, CXCL2 upregulation supports chemokine networks that sustain neutrophil-mediated bacterial clearance, highlighting its protective role in vulnerable populations.[71] For viral infections, such as influenza, regulatory elements like lnc-Cxcl2 modulate CXCL2 levels in lung epithelial cells to fine-tune neutrophil infiltration and prevent excessive inflammation while maintaining antiviral defenses.[32]
Involvement in Cancer
CXCL2 exhibits pro-tumor effects primarily through the recruitment of tumor-associated neutrophils (TANs), which facilitate metastasis in various cancers. In hepatocellular carcinoma (HCC), tumor-infiltrating monocytes produce CXCL2, promoting neutrophil recruitment into the tumor microenvironment and enhancing metastatic potential via the CXCL2-CXCR2 axis. Similarly, in breast cancer, elevated CXCL2 levels contribute to TAN infiltration, supporting tumor progression and lung metastasis by creating an immunosuppressive niche. In colon cancer, CXCL2 mediates peritoneal metastasis by driving neutrophilchemotaxis and activation through CXCR2, thereby aiding tumor cell invasion and dissemination.[72][73]The ELR+ motif of CXCL2 plays a key role in promoting angiogenesis and tumor growth. This structural feature enables CXCL2 to bind CXCR2 on endothelial cells, inducing vascularization that supports nutrient supply to hypoxic tumors. In pancreatic cancer, CXCL2 is upregulated in the tumor microenvironment, contributing to aberrant angiogenesis and serving as a potential biomarker for disease progression, as evidenced by recent multiomics analyses linking its expression to NETosis and poor outcomes.[53][74]Tumor cell-derived CXCL2 engages in crosstalk with neutrophils, polarizing them toward a pro-tumor (N2) phenotype via the PI3K/AKT pathway, which enhances immune suppression in the tumor microenvironment. This polarization fosters TAN-mediated inhibition of cytotoxic T cells and promotes extracellular matrix remodeling, further aiding tumor evasion and growth.[75]High serum levels of CXCL2 correlate with poor survival across multiple cancers, including HCC, colon, and ovarian cancers, where elevated circulating CXCL2 reflects increased TAN activity and metastatic burden, serving as an independent prognostic indicator.[53][18]
Associations with Other Diseases
CXCL2 plays a significant role in cardiovascular diseases, particularly atherosclerosis, where it promotes the adhesion of neutrophils to the vascular endothelium, thereby exacerbating plaque formation and chronic inflammation. In this process, elevated CXCL2 expression in the aortic arch, often driven by pathways such as IL-17A/IL-17RA and NF-κB activation, facilitates the recruitment and firm adhesion of neutrophils to endothelial cells, contributing to endothelial dysfunction and lesion progression. Studies in human and murine models have demonstrated that increased CXCL2 levels correlate with enhanced neutrophilchemotaxis and monocyte infiltration in atherosclerotic plaques, underscoring its pro-atherogenic effects independent of acute infectious responses.[76][2]In metabolic disorders, CXCL2 is upregulated in adipose tissue during obesity, where it drives neutrophil recruitment and sustains low-grade inflammation that impairs insulin signaling and promotes insulin resistance. Human adipose tissue from obese individuals shows elevated CXCL2 expression, which correlates with increased neutrophil activation and endothelial inflammation, linking adiposity to systemic metabolic dysfunction. This chemokine's role in adipose inflammation is evidenced by its ability to influence endothelial cell function, fostering a pro-inflammatory microenvironment that hinders glucose homeostasis without direct involvement in oncogenic processes.[76][2]CXCL2 expression is elevated in the retinal pigment epithelium (RPE) of patients with age-related macular degeneration (AMD), contributing to the degenerative changes observed in this ocular condition. Genome-wide analyses of RPE-choroid complexes from AMD-affected eyes reveal consistent upregulation of CXCL2 across all disease phenotypes, alongside other chemokines, promoting localized inflammation and glial activation that accelerate RPE degeneration and photoreceptor loss. This elevation supports a chemokine-driven inflammatory response in the retina, distinct from broader systemic inflammatory cascades.[77]In pulmonary diseases, CXCL2 is implicated in chronic obstructive pulmonary disease (COPD) and asthma exacerbations, where it contributes to airway remodeling through sustained neutrophil infiltration and structural alterations. In COPD, bronchoalveolar lavage fluid shows increased CXCL2 levels, which correlate with disease severity and promote neutrophil accumulation, leading to mucus hypersecretion and epithelial damage that perpetuate airway fibrosis. Similarly, in severe asthma, CXCL2 facilitates neutrophil-specific chemotaxis and smooth muscle cell migration, enhancing subepithelial fibrosis and bronchial hyperresponsiveness during exacerbations. These effects highlight CXCL2's involvement in chronic airway pathology, mediated via CXCR2 signaling.[78][79]
Clinical Applications
Stem Cell Mobilization
CXCL2, also known as growth-regulated oncogene beta (GROβ), plays a key role in hematopoietic stem cell (HSC) mobilization through its interaction with the CXCR2 receptor on neutrophils, leading to the release of matrix metalloproteinase-9 (MMP-9). This protease disrupts the retention of HSCs in the bone marrow by cleaving stromal cell-derived factor-1 (SDF-1) and other adhesion molecules, facilitating HSC egress into the peripheral blood. When combined with plerixafor, a CXCR4antagonist that blocks SDF-1 binding to HSCs, CXCL2 exhibits synergistic effects, enhancing mobilization efficiency without relying on granulocyte colony-stimulating factor (G-CSF).[80]Clinical evidence for CXCL2-based mobilization comes from trials using MGTA-145, a bioengineered, protease-resistant variant of CXCL2 (GROβT), developed by Magenta Therapeutics. In a Phase 1 trial in healthy donors, a single dose of MGTA-145 combined with plerixafor mobilized a median of 4 × 10⁶ CD34+ cells/kg from one apheresis session in 92% of participants, with peak circulating CD34+ cells reaching 40/μL.[81] A Phase 2 trial (NCT04552743, initiated in 2020) in patients with multiple myeloma demonstrated efficacy, where 88% of participants achieved at least 2 × 10⁶ CD34+ cells/kg in 1-2 apheresis days, and 68% met this threshold in a single day. However, development of MGTA-145 was halted by Magenta Therapeutics in February 2023 following a patientdeath in a separate trial and a strategic review, with no further clinical advancement reported as of November 2025.[82][83]This regimen offered advantages over traditional G-CSF-based methods, including same-day dosing and collection, which reduces the mobilization timeline from 4-5 days to hours and eliminates the need for multi-day G-CSF injections. Post-transplant recovery is accelerated, with median neutrophil engraftment occurring at day 12, compared to longer durations with G-CSF. Additionally, the mobilized grafts showed superior engraftment potential, with 23-fold higher short-term repopulating activity in preclinical models and reduced graft-versus-host disease risk due to lower CD8+ T-cell content.[82][81]Outcomes include significantly increased yields of CD34+ cells, particularly primitive HSCs (CD90+CD45RA-), comprising up to 35.8% of the graft versus 6.9% with G-CSF alone. The safety profile is favorable, characterized by mild, transient adverse events such as grade 1 bone pain in 44% of cases, resolving within 30 minutes, and modest elevations in neutrophils without serious complications. No grade 3 or 4 events were reported in the trials.[81][82]
Therapeutic Targeting
Therapeutic targeting of CXCL2 primarily involves modulating its receptor, CXCR2, or directly neutralizing the chemokine to mitigate excessive neutrophil recruitment in inflammatory and cancerous conditions. CXCR2 inhibitors have advanced to clinical trials, demonstrating potential in reducing tumor-associated inflammation and enhancing immunotherapy responses. For instance, AZD5069, a selective CXCR2 antagonist, is being evaluated in a phase I/II trial combined with durvalumab, an anti-PD-L1 immune checkpointinhibitor, for patients with advanced hepatocellular carcinoma (HCC), with the study ongoing as of 2025 to assess safety, dosing, and efficacy in modulating the tumor microenvironment.[84] Similarly, reparixin, another CXCR2 inhibitor, has shown promise in phase Ib and II trials for breast cancer, where it reduced cancer stem cell markers and inflammatory cytokines when combined with paclitaxel, and is currently under investigation in phase II for myelofibrosis to block IL-8-mediated signaling and inhibit disease progression.[85][86] These inhibitors target the CXCL2-CXCR2 axis to limit pro-tumorigenic neutrophil infiltration, with preclinical data supporting their role in suppressing metastasis in lung and pancreatic cancers.[87]In models of systemic lupus erythematosus (SLE), elevated CXCL2 levels correlate with neutrophil hyperactivation via the PI3K/AKT/NF-κB pathway, contributing to inflammation.[88] Direct neutralization of CXCL2 using antibodies represents an emerging preclinical strategy to curb neutrophil-driven inflammation in autoimmune diseases. Although specific trials for age-related macular degeneration (AMD) are lacking, analogous preclinical studies in inflammatory skin models show that anti-CXCL2 antibodies effectively reduce neutrophil infiltration and tissue damage, suggesting potential broader applicability for ocular inflammatory conditions involving similar pathways.[89]CXCL2 also serves as a prognostic biomarker in oncology, particularly in pancreatic cancertrials. Data from the 2025 ASCO Annual Meeting on the phase IItrial of elraglusib (9-ING-41), a GSK-3β inhibitor combined with gemcitabine/nab-paclitaxel, identified high baseline CXCL2 levels as a predictive marker of improved overall survival, reversing its unfavorable prognostic role in untreated patients and highlighting its utility in stratifying responders to targeted therapies.[90][91] This biomarker insight supports personalized approaches, where CXCL2 expression guides combination regimens to optimize outcomes in metastatic pancreatic ductal adenocarcinoma.Despite these advances, therapeutic targeting of CXCL2 and CXCR2 faces challenges due to the chemokine's dual pro- and anti-inflammatory roles, which can promote tumor suppression in some contexts while driving progression in others, necessitating careful dosing to avoid immunosuppression. Combination therapies, such as pairing CXCR2 inhibitors with checkpoint inhibitors or chemotherapy, are being explored to enhance efficacy and mitigate resistance, as seen in ongoing trials where such synergies improve antitumor immunity without excessive toxicity.[2] Balancing these effects remains critical for translating preclinical success into durable clinical benefits.