Fact-checked by Grok 2 weeks ago

CCL2

CCL2, also known as monocyte chemoattractant protein-1 (MCP-1), is a small cytokine belonging to the CC chemokine family that primarily functions as a chemoattractant for monocytes, memory T lymphocytes, and natural killer (NK) cells by binding to its receptor, C-C chemokine receptor type 2 (CCR2). Encoded by the CCL2 gene located on chromosome 17q12 in humans, CCL2 consists of 76 amino acids with a molecular weight of approximately 13 kDa and features a characteristic structure including a long N-terminal domain, three antiparallel β-strands, and a C-terminal α-helix. As the first discovered human CC chemokine, CCL2 plays a central role in regulating inflammatory responses by promoting the migration and infiltration of immune cells to sites of infection or injury. Beyond its chemotactic properties, CCL2 influences myeloid cell behavior beyond mere attraction, including modulating integrin expression on monocytes to enhance adhesion and survival, which contributes to its broader impact on immune regulation. In pathological contexts, elevated CCL2 levels are implicated in various diseases; for instance, it drives monocyte recruitment in atherosclerosis, exacerbating plaque formation, and facilitates tumor-associated macrophage infiltration in cancers such as glioblastoma and breast cancer, promoting disease progression. Additionally, CCL2/CCR2 signaling has been linked to neuroinflammation in conditions like multiple sclerosis and contributes to fibrosis in organs such as the lungs and kidneys. These multifaceted roles highlight CCL2 as a key mediator in both physiological immunity and chronic inflammatory disorders, making it a promising target for therapeutic interventions.

Overview and History

Definition and Nomenclature

CCL2, also known as C-C motif chemokine ligand 2, is a small cytokine with a molecular weight of approximately 13 kDa (observed on SDS-PAGE) that belongs to the CC chemokine subfamily of the chemokine superfamily. This subfamily is defined by a conserved structural motif featuring two adjacent cysteine residues near the N-terminus, which form disulfide bonds critical to the protein's function. In contrast to other chemokine subfamilies—such as CXC (with one amino acid separating the first two cysteines), XC (lacking the first and third cysteines), and CX3C (with three amino acids between the first two)—the CC configuration distinguishes CCL2 and its relatives in their roles within immune regulation.80165-X) Historically, CCL2 has been referred to by several alternative names, including monocyte chemoattractant protein-1 (MCP-1), small inducible cytokine A2 (SCYA2), and (its murine homolog). These designations arose from early functional and gene cloning studies but led to inconsistencies across and research contexts. To address this, the Chemokine Nomenclature Subcommittee of the International Union of Immunological Societies (IUIS) and the (WHO) established a unified system in 2000, systematically numbering based on their chromosomal locations and phylogenetic relationships.80165-X) Under this framework, CCL2 is positioned as the second in the CC subfamily (denoted as CCL), reflecting its genomic organization on human chromosome 17 alongside other CC members.80165-X) This standardized naming facilitates precise communication in scientific literature and underscores CCL2's classification within the broader chemokine family, which orchestrates leukocyte trafficking through chemotactic gradients.

Discovery

Early research into monocyte chemoattractants provided initial hints of CCL2's existence through studies on tumor-derived factors. In 1983, Bottazzi and colleagues identified chemotactic activity for mononuclear phagocytes in culture supernatants from murine and human tumor cells, observing a correlation between this activity and macrophage content in neoplastic tissues, suggesting a role in regulating tumor-associated macrophage infiltration. These findings indicated that tumor cells could secrete soluble factors attracting monocytes, laying groundwork for later chemokine discoveries. The formal identification of CCL2, initially termed monocyte chemoattractant protein-1 (MCP-1) or monocyte chemotactic and activating factor (MCAF), occurred in 1989 through from conditioned media of stimulated cells. Yoshimura and colleagues purified two chemoattractants from glioma cell line supernatants, demonstrating potent chemotactic activity specific to but not neutrophils using Boyden chamber assays. Concurrently, Rollins and colleagues reported MCP-1 in cytokine-treated fibroblasts and endothelial , highlighting its induction by inflammatory stimuli in these key vascular and . The murine homolog, known as the JE , had been cloned earlier in 1988 by Rollins and colleagues from platelet-derived growth factor-stimulated fibroblasts, with expression in cells confirming its cytokine-like properties and -attracting function. Full-length MCP-1 cDNA was cloned and expressed in COS-1 cells in 1989 by Yoshimura and colleagues, enabling further functional validation of its role as a specific chemoattractant. In the pre-nomenclature era, CCL2 was referred to variably as MCAF, a RANTES-like factor, leading to confusion among early researchers due to overlapping functional descriptions. This ambiguity was resolved through international symposia in the early , culminating in the standardized system that designated it as CCL2 within the CC subfamily.00193-8) Early studies further expanded its profile; in 1996, Doranz and colleagues demonstrated that CCL2 suppresses HIV-1 infection by competing for the receptor on target cells, establishing an antiviral role alongside its chemotactic functions.

Genetics

Gene Structure and Location

The CCL2 is located on the long arm of human 17 at position 17q12, specifically spanning genomic coordinates 34,255,274 to 34,257,208 (GRCh38 assembly), which corresponds to a total length of approximately 1.9 kb. This locus is part of a larger CC gene cluster on 17q11.2-q12, which includes neighboring genes such as CCL1, CCL7, CCL8, CCL11, CCL13, CCL15, and CCL16, arising from tandem gene duplications that expanded the family. The gene consists of three exons separated by two introns, with a compact typical of CC chemokines. Exon 1 primarily encompasses the (UTR), exon 2 encodes the and the N-terminal portion of the protein, and exon 3 contains the coding for the C-terminal region along with the 3' UTR. Transcription of the primary isoform yields a mature mRNA transcript (NM_002982.4) of 741 bp, which translates into a 99- pre-proprotein precursor. Splicing produces at least three transcript variants, though the predominates in most tissues. The is highly conserved across mammals, exhibiting approximately 58% identity with the murine Ccl2 ortholog, reflecting shared functional constraints in immune regulation. The promoter region upstream of the CCL2 transcription start site features multiple regulatory elements that enable rapid, inducible expression in response to inflammatory stimuli. Key binding sites include those for in the distal promoter, activator protein 1 (AP-1), and specificity protein 1 (SP-1) in the proximal region, which collectively drive transcription upon activation by cytokines such as interleukin-1 (IL-1) and tumor necrosis factor alpha (TNF-α). These elements allow for tight control of CCL2 expression, primarily in monocytes, endothelial cells, and fibroblasts during immune responses.

Genetic Variations and Population Genetics

The CCL2 gene harbors several single nucleotide polymorphisms (SNPs) that influence its expression and function. The most studied variant is rs1024611, an A-to-G transition located 2578 base pairs upstream in the promoter region, which disrupts a putative site and enhances binding of the , leading to increased CCL2 transcription in response to inflammatory stimuli. Another notable polymorphism is rs13900, a C-to-T change in the 3' (3' UTR), which modulates mRNA stability by altering the binding affinity of the HuR; the T allele results in greater transcript stability compared to the C allele. These SNPs are often in strong (LD), with rs13900 serving as a proxy for rs1024611 in haplotype analyses. Allele frequencies of these variants exhibit population-specific patterns, reflecting historical migrations and potential selective pressures. For rs1024611, the G allele frequency is approximately 30% in European-ancestry populations, rises to 45-50% in East Asian groups such as and , and is lower at around 20-25% in African-ancestry populations, based on data from the . The rs13900 T allele shows similar stratification, with higher prevalence in Asian cohorts. These variants are embedded in a on 17q11.2, where rs1024611 demonstrates LD with polymorphisms in the nearby CCL11 gene, forming haplotypes that may collectively influence network regulation. Evolutionary analyses indicate that CCL2 polymorphisms have been shaped by , particularly in immune-related contexts. Signals of positive selection on CCL2 have been detected in lineages, suggesting adaptive to counter infectious pressures such as , where higher G allele frequencies in certain endemic regions may modulate recruitment for enhanced clearance. Balancing selection is evident in populations prone to chronic , maintaining diversity that balances immune activation against risks, as inferred from elevated heterozygosity and deviation from neutral drift models in global genomic datasets. Haplotype diversity in the CCL2-CCL11 region traces back to ancient migrations , with reduced variation in non-African groups consistent with founder effects. Functionally, the rs1024611 G drives higher basal and stimulated CCL2 production, amplifying chemoattraction and correlating with elevated levels . The rs13900 T variant enhances post-transcriptional , potentially exacerbating this effect in compound haplotypes. Rare loss-of-function in CCL2, such as predicted frameshifts or nonsense variants, are infrequently reported but have been associated with diminished secretion and reduced immune in cellular models, though their population prevalence remains low (<0.1% in gnomAD). Association studies highlight a tension between neutral drift and adaptive evolution; for instance, the G 's enrichment in malaria-endemic Asian and African subgroups supports immune modulation benefits, while excess in inflammatory cohorts suggests context-dependent fitness trade-offs.

Molecular Biology

Protein Structure

CCL2 is synthesized as a 99-amino acid pre-proprotein in humans, consisting of a 23-amino acid N-terminal signal peptide (residues 1–23) that directs secretion, followed by the mature protein of 76 amino acids (residues 24–99). The calculated molecular weight of the unglycosylated mature is approximately 8.7 kDa. The mature protein adopts the canonical chemokine fold, characterized by a flexible N-terminal domain, a short β0 strand (residues 9–11), an antiparallel three-stranded β-sheet arranged in a Greek key topology (strands β1: 27–31, β2: 40–45, β3: 51–54), and an overlying C-terminal α-helix (residues 58–69). This tertiary structure is stabilized by two conserved intramolecular disulfide bonds between Cys34–Cys59 and Cys35–Cys75 (using full precursor numbering), which link the N-terminal cysteines to those in the 30s and 40s loops, respectively, maintaining the core fold essential for stability. Post-translational modifications of CCL2 include O-linked glycosylation primarily at Thr10 (mature numbering) in the N-terminal region, which can attach GalNAc or extended glycans and modestly reduces chemotactic activity by altering surface charge and conformation. At physiological concentrations, CCL2 predominantly exists as a monomer in solution; however, at high concentrations (>100 μM), it forms homodimers through intermolecular interactions involving the N-terminal loops and β1 strands, resulting in an elongated antiparallel . The solution structure of the CCL2 homodimer was first resolved by NMR spectroscopy in 1997 (PDB ID: 1DOM), revealing a monomeric form at low concentrations but dimeric assembly with a buried surface area of ~800 Ų; subsequent X-ray crystallography studies (e.g., PDB ID: 1DOL, 1.9 Å resolution) confirmed the dimeric state on surfaces, with minimal deviations in the core fold (RMSD <1 Å). While human and murine CCL2 share ~69% sequence identity overall, notable variations occur in the N-terminal region (human: residues 1–10: QPDAINAPVT; murine: QPDSTQSIIS), influencing conformational flexibility and receptor binding affinity to CCR2, with the murine variant exhibiting higher N-terminal dynamics and slightly reduced potency in some chemotaxis assays.

Receptors and Signaling Pathways

CCL2 primarily interacts with the C-C chemokine receptor type 2 (), a seven-transmembrane G protein-coupled receptor (GPCR) expressed predominantly on monocytes, macrophages, and certain other immune cells, with high binding affinity characterized by a dissociation constant (Kd) of approximately 0.26 nM. CCR2 exists in two main isoforms, and , generated by alternative splicing of the C-terminal tail; these isoforms share identical extracellular and transmembrane domains but differ in the length and sequence of their intracellular C-termini (14 amino acids for CCR2A versus 31 for CCR2B), which can influence receptor desensitization, internalization, and downstream signaling specificity. In addition to CCR2, CCL2 exhibits weaker interactions with secondary receptors, including low-affinity binding to and as well as the atypical decoy receptor (also known as ), which scavenges chemokines without triggering signaling; notably, CCL2 does not bind to receptors of the family due to structural incompatibility between CC and CXC chemokines. The binding mechanism of CCL2 to CCR2 involves a two-site interaction model, where the N-terminal domain of CCL2 initially engages the extracellular N-terminus of CCR2 through hydrophobic and hydrogen bond interactions, followed by docking of CCL2's N-terminal residues into the receptor's extracellular loops, particularly the second extracellular loop (ECL2), to stabilize the complex and induce conformational changes for G protein activation. A 2022 cryo-EM structure of the (PDB ID: 7XA3) at 2.9 Å resolution confirms this model, showing CCL2 inserting deeply into the transmembrane domain and highlighting key interactions such as hydrogen bonds between ECL2 residues (e.g., C190, G191, P192) and transmembrane helix 3 (e.g., Y120) with CCL2's N-terminus, while mutations in these sites significantly impair binding and signaling; furthermore, CCL2 can dimerize via its N-terminal region, allowing a single dimer to engage and activate two CCR2 molecules simultaneously, enhancing avidity at low ligand concentrations. Upon CCL2 binding, CCR2 couples to heterotrimeric G proteins of the Gi/o subclass, leading to GDP-GTP exchange on the Gαi/o subunit and dissociation into Gαi/o and Gβγ components; the free Gβγ subunits activate phospholipase C-β (PLC-β), which hydrolyzes phosphatidylinositol 4,5-bisphosphate (PIP2) to produce inositol 1,4,5-trisphosphate (IP3) and diacylglycerol (DAG), culminating in IP3-mediated release of Ca²⁺ from intracellular stores and DAG-dependent activation of protein kinase C (PKC). This G protein activation also inhibits adenylyl cyclase via Gαi/o, reducing cyclic AMP levels, while parallel pathways include recruitment of phosphoinositide 3-kinase (PI3K) to the membrane by Gβγ, leading to Akt phosphorylation and promotion of cell survival and migration; mitogen-activated protein kinase (MAPK)/extracellular signal-regulated kinase (ERK) cascade activation drives transcriptional responses for proliferation and differentiation; and Janus kinase (JAK)/signal transducer and activator of transcription (STAT) pathway engagement facilitates cytokine production and gene expression changes. To prevent prolonged signaling, CCR2 undergoes rapid desensitization following ligand binding, primarily through phosphorylation of its C-terminal tail by G protein-coupled receptor kinases (GRKs), such as GRK2, which recruits β-arrestins (particularly β-arrestin 2) to uncouple the receptor from G proteins and sterically hinder further activation. β-Arrestin binding also promotes clathrin-mediated endocytosis of CCR2 via adaptor protein 2 (AP-2) recruitment, leading to receptor internalization into early endosomes for either recycling or lysosomal degradation, thereby attenuating responsiveness. Additionally, CCR2 signaling exhibits cross-talk with (TLR4), where TLR4 activation induces GRK2 translocation to the plasma membrane, enhancing CCR2 phosphorylation and desensitization while paradoxically augmenting monocyte chemotaxis through balanced regulation of receptor availability.

Physiological Functions

Role in Immune Response

CCL2, also known as monocyte chemoattractant protein-1 (MCP-1), plays a central role in orchestrating innate immune responses by forming chemotactic gradients that direct the mobilization and recruitment of monocytes from the bone marrow to sites of infection. Specifically, it facilitates the egress of Ly6Chi inflammatory monocytes in a CCR2-dependent manner, enabling their rapid infiltration into tissues to combat pathogens such as . For instance, during bacterial like sepsis or Listeria monocytogenes challenge, CCL2-mediated monocyte recruitment is essential for efficient bacterial clearance, as demonstrated in models where CCL2 deficiency impairs pathogen elimination and increases susceptibility. This process not only enhances but also amplifies local inflammatory signals to coordinate broader immune activation. Beyond monocytes, CCL2 contributes to adaptive immunity by attracting memory + T cells and dendritic cells (DCs) to facilitate and immune modulation. It promotes the recruitment of CCR2-expressing conventional DCs to inflammatory sites, where they can present antigens to T cells, thereby supporting effective priming of adaptive responses. Additionally, CCL2 influences the Th1/Th2 balance by recruiting Th2, Th17, and regulatory T cells, which helps fine-tune profiles during immune challenges, as evidenced by altered Th1/Th2 ratios in CCL2-deficient models during parasitic infections. This selective attraction ensures coordinated interactions between innate and adaptive components for optimal defense. In antiviral immunity, CCL2 supports host defense by recruiting natural killer (NK) cells and inflammatory monocytes to viral infection sites, enhancing cytolytic activity and viral containment, as seen in models of herpes simplex virus (HSV-1) infection. CCL2 also aids in resolving immune responses post-pathogen clearance, particularly in wound healing, where it coordinates macrophage polarization toward an anti-inflammatory M2 phenotype to promote tissue repair. By recruiting and priming macrophages via CCR2 signaling, CCL2 facilitates the transition from pro-inflammatory M1 to reparative M2 states, accelerating extracellular matrix remodeling and reducing fibrosis in injury models. In allergic contexts, CCL2 modulates type 2 immunity by preferentially attracting basophils, which amplify eosinophil influx and Th2 cytokine production, thereby supporting humoral responses against allergens without directly chemotaxing eosinophils.

Role in Homeostasis and Development

CCL2 plays a crucial role in maintaining vascular by facilitating the egress of monocytes from the into circulation, which supports endothelial cell integrity and overall vascular stability under physiological conditions. This process involves the CCL2-CCR2 axis, which regulates the steady-state mobilization of inflammatory monocytes, ensuring their availability for patrolling and repairing vascular without inducing . In during , CCL2 contributes to recruitment by attracting monocyte-derived cells that differentiate into , stabilizing nascent blood vessels and promoting proper vascular maturation. In neural development, CCL2 is essential for guiding the and positioning of within the embryonic , where it is secreted by neural progenitor cells to orchestrate microglial colonization from the . This chemokine-mediated recruitment ensures are properly distributed to support neuronal differentiation and during embryogenesis. Additionally, CCL2 helps maintain the blood- barrier integrity by modulating interactions between endothelial cells, , and through the CCL2-CCR2 signaling axis, thereby preserving a selective barrier that protects the developing . CCL2 influences by directing the migration of precursors to sites of , thereby contributing to calcium and skeletal maintenance. In physiological contexts, such as during , CCL2 recruits these precursors to balance bone formation and resorption, preventing imbalances that could disrupt mineral . This targeted migration supports the required for tissue integrity throughout life. In , CCL2 modulates the presence of resident in lean fat depots, where low-level expression maintains an anti-inflammatory, M2-like population essential for metabolic . These , recruited via CCL2-CCR2 interactions, facilitate remodeling, insulin sensitivity, and under normal conditions, preventing excessive and supporting energy balance. During pregnancy, constitutive CCL2 expression at the maternal-fetal interface supports invasion and placental development by attracting macrophages that secrete factors like G-CSF, which enhance and vascular remodeling. This low-level activity ensures proper spiral artery modification and nutrient exchange without triggering immune rejection, contributing to successful embryogenesis and fetal .

Pathological Implications

Involvement in Inflammatory Diseases

CCL2, also known as monocyte chemoattractant protein-1 (MCP-1), plays a central role in the of various inflammatory diseases by facilitating the of and macrophages to sites of chronic inflammation via its interaction with the receptor. In these conditions, elevated CCL2 expression amplifies immune cell infiltration, perpetuating tissue damage and disease progression. In atherosclerosis, CCL2 promotes the recruitment of circulating to arterial plaques through the CCL2-CCR2 axis, where these cells differentiate into that uptake oxidized to form foam cells, thereby accelerating plaque growth and instability. This monocyte infiltration is a key driver of lesion progression, as evidenced by studies showing reduced atherosclerotic burden in CCL2-deficient models. Elevated CCL2 levels in human plaques correlate with increased macrophage content and cardiovascular risk. Rheumatoid arthritis (RA) features synovial overexpression of CCL2, which attracts CCR2-expressing monocytes into the joint space, leading to accumulation that sustains and contributes to and erosion. These infiltrating s release pro-inflammatory cytokines, exacerbating joint destruction, as demonstrated in RA synovial analyses showing high CCL2 correlation with disease severity. Genetic and pharmacological evidence supports CCL2's role in driving this pathogenic loop. In (), CCL2 facilitates the transmigration of perivascular monocytes across the (), promoting and demyelination by enabling immune cell entry into the . This recruitment disrupts BBB integrity and amplifies lesion formation, with CCL2 levels elevated in MS and active plaques. Experimental models confirm that CCL2-CCR2 signaling exacerbates disease progression through monocyte-mediated pathology. Type 2 diabetes involves CCL2-mediated recruitment of CCR2-positive macrophages into adipose tissue, where they polarize toward a pro-inflammatory M1 phenotype, fostering chronic low-grade inflammation that impairs insulin signaling and promotes insulin resistance. This adipose macrophage infiltration is linked to obesity-associated metabolic dysfunction, as CCL2 knockout mice exhibit reduced inflammation and improved glucose homeostasis on high-fat diets. Circulating CCL2 levels in diabetic patients reflect adipose tissue inflammation severity. Recent 2025 research highlights CCL2's involvement in post-COVID long-haul syndrome, where persistent CCL2 elevation drives endothelial injury and chronic inflammation, contributing to ongoing cardiac and systemic symptoms through sustained activation. In (ARDS), including COVID-19-related cases, CCL2 orchestrates lung accumulation, intensifying alveolar inflammation and impairing gas exchange, as shown in models where CCL2-CCR2 blockade reduces immune cell influx. These findings underscore CCL2's emerging role in post-viral inflammatory sequelae.

Role in Cancer and Fibrosis

CCL2 plays a pivotal role in cancer progression by recruiting tumor-associated macrophages (TAMs), which foster an immunosuppressive . These TAMs, primarily polarized to phenotype, suppress antitumor immune responses through of cytokines and promotion of T-cell exhaustion. In various solid tumors, CCL2-mediated TAM infiltration correlates with increased tumor growth and immune evasion. For instance, in esophageal , the CCL2-CCR2 axis recruits M2-like TAMs that upregulate PD-L2 expression, enhancing PD-1 signaling and immune suppression. In addition to immune modulation, CCL2 promotes , particularly in and cancers, by inducing in tumor cells. Autocrine CCL2 signaling activates on cancer cells, triggering downstream pathways like MEK/ERK that enhance and invasion while repressing E-cadherin expression. This process facilitates dissemination to distant sites, with elevated CCL2 levels associated with poorer in these malignancies. CCL2 also drives by increasing and inducing VEGF expression in endothelial cells, supporting nutrient supply to growing tumors; neutralization of CCL2 has been shown to reduce vessel density and tumor volume in preclinical models. In , CCL2 from the recruits immunosuppressive monocytes that accelerate disease progression and resistance to anti-angiogenic therapies. Similarly, in , CCL2 contributes to , the dense fibrotic that shields tumor cells from immune attack and chemotherapy. In fibrosis, CCL2 facilitates monocyte recruitment and differentiation into myofibroblasts across multiple organs, exacerbating deposition and tissue scarring. Through the CCL2- axis, circulating monocytes are drawn to sites, where they undergo macrophage-to-myofibroblast (MMT), adopting a profibrotic phenotype that sustains chronic inflammation. Recent studies highlight this in (IPF), where bone marrow-derived monocytes, activated via CCL2/CCR2 signaling, drive alveolar remodeling and accumulation; inhibition of this axis reduces fibrotic progression in murine models. In renal fibrosis following ischemic , CCL2 from tubular epithelial cells amplifies monocyte infiltration and MMT, leading to persistent matrix buildup and impaired . Hepatic fibrosis in nonalcoholic () similarly involves CCL2-mediated recruitment of inflammatory monocytes, promoting activation and progression to . Elevated CCL2 levels in NASH livers correlate with increased infiltration and fibrotic , creating a feed-forward loop of inflammation and scarring. CCL2's profibrotic effects are amplified through crosstalk with TGF-β, where CCL2 upregulates TGF-β production in fibroblasts, enhancing Smad signaling and differentiation. In cancer, CCL2 interacts with the PD-1/ axis to evade immunity; tumor-derived CCL2 induces PD-L1 expression on and cancer cells, blunting T-cell and supporting metastatic niches.

Clinical Significance

As a Biomarker

CCL2, also known as monocyte chemoattractant protein-1 (MCP-1), is measured primarily through in biological fluids such as , , and , with mean levels in healthy individuals typically ranging from 200 to 370 pg/mL. Quantitative is employed to quantify CCL2 mRNA expression in tissue samples for assessing local production. Cutoff values for diagnostic purposes vary by disease context; for instance, levels exceeding 200 pg/mL have been associated with inflammatory states, while levels below approximately 100 pg/mL have been associated with increased risk of adverse cardiovascular outcomes, such as , in certain conditions. In prognostic applications, elevated circulating CCL2 levels in atherosclerosis indicate plaque instability and predict adverse cardiovascular outcomes, including and long-term mortality. In oncology, high CCL2 correlates with poor overall survival and increased risk in and cancers, serving as an independent predictor of tumor progression. For diagnostics, CCL2 is integrated into multi-biomarker panels, such as those combining it with (CRP) to monitor disease activity and progression in (RA). In neuroinflammatory disorders, CSF CCL2 concentrations distinguish (MS) from other conditions, with elevated levels correlating to disease activity, and also show discriminatory power comparable to neurodegeneration markers in (AD). Advances as of 2025 underscore circulating CCL2 as a predictor of fibrosis progression in (IPF), where it serves as a shared with systemic sclerosis, reflecting common pathogenic mechanisms. Post-stroke, CCL2 elevations indicate activation and systemic inflammatory responses, aiding in outcome prediction. Despite these utilities, CCL2's role as a is constrained by its non-specificity, as levels rise in diverse inflammatory conditions including nephropathy, , and , reducing diagnostic precision. Additionally, genetic variants in the CCL2 and genes can influence circulating levels, complicating interpretation.

Therapeutic Targeting

Therapeutic strategies targeting the CCL2/CCR2 axis primarily focus on inhibiting receptor-ligand interactions to reduce monocyte recruitment and mitigate inflammation in fibrotic and neoplastic conditions. CCR2 antagonists, including small-molecule inhibitors, have been developed to block this pathway. Bindarit, an indazole derivative that selectively inhibits CCL2 synthesis by targeting the CCL2 mRNA-stabilizing protein, has demonstrated anti-inflammatory effects in preclinical models of atherosclerosis and restenosis, reducing monocyte chemotaxis without affecting other chemokines. In clinical trials, bindarit showed tolerability and reduced inflammatory markers in conditions like diabetic nephropathy, though larger cardiovascular outcome studies are ongoing. Similarly, MLN1202, a humanized monoclonal antibody against CCR2, was evaluated in phase II trials for atherosclerosis, where it significantly lowered high-sensitivity C-reactive protein levels and was well-tolerated, indicating proof-of-mechanism for CCR2 blockade in reducing systemic inflammation. Cenicriviroc, a dual CCR2/CCR5 antagonist, has advanced to phase III trials (AURORA study) for nonalcoholic steatohepatitis (NASH) with liver fibrosis, building on phase II data showing improved fibrosis resolution without NASH worsening in twice as many patients compared to placebo after one year of treatment, though full phase III results confirmed safety but limited histological efficacy. Anti-CCL2 antibodies represent another approach to neutralize the directly. Carlumab (CNTO 888), a human monoclonal antibody targeting CCL2, was tested in a phase II trial for metastatic castration-resistant , where it was well-tolerated but failed to demonstrate antitumor activity as a monotherapy, primarily due to rapid rebound hypersecretion of CCL2 upon treatment cessation, which negated sustained blockade. To address such limitations, emerging bispecific antibodies targeting both CCL2 and epitopes have been engineered using structure-guided design, showing promise in preclinical models by enhancing binding affinity and reducing immunosuppressive myeloid cell recruitment in tumors, with preclinical bispecific antibodies targeting CCL2 and continuing to demonstrate enhanced efficacy and initial clinical translation efforts underway as of November 2025. Despite these advances, therapeutic targeting of CCL2/CCR2 faces significant challenges, including functional redundancy with other that bind CCR2, such as CCL7, which can compensate for CCL2 inhibition and sustain in inflammatory settings like gliomas and cancer . This redundancy often leads to incomplete pathway blockade, as evidenced by studies where CCL2 neutralization alone failed to fully impair immune cell infiltration due to CCL7 upregulation. Consequently, combination therapies are increasingly emphasized; for instance, CCR2 antagonists paired with PD-1 inhibitors have enhanced antitumor responses in preclinical tumor models by alleviating myeloid suppression and boosting T-cell infiltration, with a 2025 study demonstrating reduced brain metastases through CCL2 blockade combined with PD-1/P-selectin modulation. Recent 2025 updates highlight structure-based leveraging cryo-EM structures of the CCL2-CCR2-Gi , resolved at high in 2022, which reveal deep insertion into the receptor's and inform the development of allosteric modulators to improve selectivity and overcome redundancy. Insights from the structure have informed of allosteric modulators. inhibitors, such as DMX-200, are in phase 3 trials for renal as of 2025, with preclinical models showing reduced infiltration and decreased deposition in ; the phase 3 ACTION3 trial of DMX-200 remains ongoing, with topline results anticipated in 2026. approaches remain preclinical but promising; CRISPR/Cas9 editing of CCL2 promoter single nucleotide polymorphisms (SNPs) has been explored to downregulate expression in fibrotic tissues, while siRNA delivery via nanoparticles enables local knockdown of CCL2 in tumor microenvironments, reducing in mouse models without systemic toxicity.