Fact-checked by Grok 2 weeks ago

Macrophage colony-stimulating factor

Macrophage colony-stimulating factor (M-CSF), also known as colony-stimulating factor 1 (CSF-1), is a and key hematopoietic that primarily regulates the , , , and of cells in the , including monocytes and macrophages. First identified in 1969, M-CSF is encoded by the * and exists in multiple isoforms produced through and post-translational modifications, including secreted forms (a 35–40 kDa and a 120 kDa ) and membrane-bound variants that can be cleaved to release soluble factors. It exerts its effects by binding to the colony-stimulating factor 1 receptor (CSF1R), a transmembrane receptor encoded by the CSF1R (c-fms) proto-oncogene, which is predominantly expressed on myeloid lineage cells. Upon binding, M-CSF activates downstream signaling pathways such as PI3K/Akt and MAPK/ERK, promoting cell and effector functions. M-CSF plays essential roles in hematopoiesis by driving the commitment of hematopoietic progenitor cells toward the monocyte- lineage and maintaining tissue-resident populations throughout development and . In the , it enhances , cytotoxicity against pathogens and tumor cells, , and production of pro-inflammatory cytokines like IL-6 and G-CSF, while also supporting anti-inflammatory M2 polarization for repair and resolution of . Beyond immunity, M-CSF is critical for differentiation and , trophoblast development during , and in various s. Dysregulation of M-CSF signaling is implicated in pathological conditions, including accelerated due to recruitment, from excessive activity, and cancer progression via tumor-associated macrophages that foster and immune evasion. Clinically, recombinant human M-CSF has been investigated for its potential to accelerate myeloid recovery in patients undergoing for , reducing the duration of and , and improving outcomes in fungal infections by bolstering macrophage-mediated defenses. Targeting the M-CSF/CSF1R axis with inhibitors is an emerging strategy in to reprogram tumor-associated macrophages and enhance antitumor immunity, with ongoing trials in solid tumors like and . Additionally, M-CSF levels serve as a for disease , with elevated concentrations associated with poor outcomes in various malignancies.

Discovery and Molecular Biology

Discovery and History

The concept of factors promoting hematopoietic colony formation emerged in the early through experiments by James E. Till and Ernest A. McCulloch, who demonstrated that irradiated cells formed visible nodules in recipient spleens, indicating the existence of self-renewing stem cells responsive to stimulatory signals in cultures. Their work laid the foundation for identifying colony-stimulating factors (CSFs), including the one specific to macrophages. The macrophage-specific CSF, later termed M-CSF or CSF-1, was first recognized in 1969 by William A. Robinson and colleagues, who identified a soluble activity in mouse lung-conditioned medium that selectively stimulated pure colony growth in vitro from precursors. Purification efforts in the included partial isolation from mouse lung-conditioned medium in 1973-1974 and full purification of mouse M-CSF from L-cell conditioned medium in 1977 (specific activity ~10^8 units/mg protein). Human M-CSF was partially purified from urine in 1971 and further in 1975, confirming its nature. These milestones clarified M-CSF's role as distinct from other CSFs like GM-CSF, leading to its nomenclature as colony-stimulating factor-1 (CSF-1) to reflect its broad mononuclear lineage support, while M-CSF emphasized its specificity; early terms like "osteoclast colony-stimulating factor" were proposed in the 1990s but subsumed under CSF-1/M-CSF as its osteoclastogenic functions were elucidated. Molecular characterization accelerated in the mid- when the CSF1 was cloned in 1985 by Kawasaki et al., revealing a 4-kb cDNA encoding a 256-amino-acid precursor with a 32-residue , enabling recombinant production and confirmation of its single-copy genomic structure spanning 20 kb with 10 exons. was detailed in 1987 by Ladner et al., identifying multiple isoforms including secreted and forms, as well as a membrane-bound variant, which accounted for diverse mRNA species ranging from 1.5 to 4.5 kb. The essential role of M-CSF isoforms was further elucidated in 1990 through the osteopetrotic (op/op) mutant, lacking secreted M-CSF, confirming their functions in . By the late , studies highlighted M-CSF's hormonal regulation in , with Jeffrey W. Pollard and colleagues demonstrating in 1987 its essential role in placental development through elevated uterine expression during in mice.

Gene and Isoforms

The human CSF1 gene, which encodes macrophage colony-stimulating factor (M-CSF), is located on the short arm of chromosome 1 at band p13.2, spanning genomic positions 109,910,242 to 109,930,992 on the forward strand (GRCh38 assembly). In mice, the orthologous Csf1 gene resides on chromosome 3, from positions 107,648,364 to 107,667,785 on the reverse strand. The gene consists of 10 exons and 9 introns, covering approximately 20 kilobases of genomic DNA. The CSF1 gene produces four major transcript variants through , each contributing to distinct protein forms. Transcript variant 1 (NM_000757.6) encodes the primary soluble isoform, while variants 2, 3, and 4 generate isoforms associated with membrane-bound and structures. These transcripts share a common N-terminal but differ in their 3' regions, enabling the production of three principal protein isoforms: a 520-amino-acid precursor (accession NP_000748.1) that is secreted after processing, a 256-amino-acid form with attachment sites, and a cell-surface isoform derived from proteolytic cleavage of longer precursors. The diversity arises from , particularly involving 6 for the , combined with proteolytic cleavage of longer precursors to yield the membrane-bound variant. Recent structural studies (as of 2023) have provided insights into the CSF1-CSF1R complex via cryo-EM, enhancing understanding of isoform-specific signaling. The CSF1 gene and its protein products exhibit strong evolutionary across mammalian , with the core coding sequence showing over 80% between and orthologs, reflecting its essential role in mononuclear biology. This extends to the exon-intron boundaries and key regulatory elements, underscoring the gene's fundamental importance in hematopoiesis.

Structure and Expression

Protein Structure

Macrophage colony-stimulating factor (M-CSF), also known as CSF-1, functions as a disulfide-linked homodimer, with each monomer typically ranging from 18 to 45 kDa in molecular weight, varying by isoform and post-translational modifications. The overall architecture features a four-α-helix bundle motif (helices A–D arranged in an up-up-down-down topology), characteristic of the cytokine family, including an antiparallel β-sheet and loops that stabilize the dimer interface through interchain disulfide bonds. This dimeric structure is essential for its biological activity, as monomeric forms exhibit reduced potency. The primary structure of the mature secreted isoform derives from a 522-amino-acid precursor, following cleavage of an N-terminal 32-amino-acid signal peptide, resulting in a ~190-amino-acid polypeptide chain. Alternative splicing generates multiple isoforms, including shorter soluble variants (e.g., 190–256 amino acids) and longer membrane-bound forms (up to 522 amino acids), all sharing a conserved growth factor domain of ~150 residues central to function. Post-translational modifications significantly influence M-CSF stability and secretion. N-linked glycosylation occurs at two sites in certain isoforms, adding carbohydrate moieties that increase and protect against , while a proteoglycan variant incorporates chains attached via serine residues in the C-terminal domain of membrane-bound isoforms. The soluble homodimeric form is primarily produced via proteolytic processing of the transmembrane precursor by enzymes such as and cathepsins, releasing the bioactive ectodomain. The three-dimensional structure of dimeric human recombinant M-CSF has been elucidated by at 2.5 Å resolution (PDB: 1HMC), revealing the compact four-helix bundle with nine bonds (six intrachain, three interchain) that maintain the active conformation. The domain, encompassing residues ~33–190, contains key hydrophobic cores and receptor-binding sites on helices A and D, underscoring its role in ligand-receptor interactions without involving the variable C-terminal regions of longer isoforms.

Expression Patterns and Regulation

Macrophage colony-stimulating factor (M-CSF), encoded by the CSF1 gene, exhibits a broad tissue distribution with particularly high expression levels in the , , , and lungs. In the and , M-CSF production is elevated during to support and immune modulation at the maternal-fetal interface. tissue, particularly osteoblasts, constitutively produces M-CSF to regulate differentiation and . Lung expression contributes to maintenance, while lower but detectable levels occur in other sites such as the , , and colon. M-CSF is synthesized by diverse cell types, including macrophages, osteoblasts, fibroblasts, and tumor cells, with production varying by isoform. Activated macrophages and monocytes serve as key sources during immune responses, while fibroblasts and osteoblasts provide steady-state levels essential for . Tumor cells often overexpress M-CSF to recruit immunosuppressive macrophages into the microenvironment. The protein exists in soluble and membrane-bound forms, with the soluble isoform predominating in circulation from fibroblasts and endothelial cells, whereas membrane-bound M-CSF is more prominent on osteoblasts and macrophages, facilitating localized cell-cell interactions. Isoform-specific localization influences paracrine versus , though detailed variations are tied to . Expression of M-CSF is tightly regulated at transcriptional and post-transcriptional levels, with additional modulation by hormones and inflammatory cues. Transcriptional control involves factors such as Sp1, which binds to promoter regions to drive basal and induced expression in various cell types; the SWI/SNF-like BAF complex further enhances promoter accessibility for cytokine-responsive transcription. Constitutive expression occurs in fibroblasts and endothelial cells, supporting steady-state survival, whereas in monocytes, M-CSF production is inducible by (LPS) or proinflammatory cytokines like TNF-α, amplifying responses during or . Hormonal influences, particularly during , upregulate M-CSF in uterine and placental tissues to promote and . Post-transcriptional regulation includes microRNAs, though specific impacts on CSF1 transcripts remain under investigation. Serum M-CSF levels display diurnal variation, peaking in the evening and troughing at night, reflecting circadian control potentially linked to systemic immune rhythms.

Receptor and Signaling

CSF1R Receptor

The colony-stimulating factor 1 receptor (CSF1R), also known as c-Fms or macrophage colony-stimulating factor receptor, is a class III transmembrane (RTK) that specifically binds M-CSF to regulate myeloid and function. Encoded by the CSF1R on 5q32 in humans, it belongs to the platelet-derived growth factor receptor family and plays a central role in transducing signals from M-CSF into intracellular responses. As the sole receptor for M-CSF, CSF1R's activation is essential for the , , and differentiation of mononuclear . Structurally, the mature human CSF1R protein comprises 972 , with a predicted molecular weight of approximately 108 . Its extracellular region features five immunoglobulin-like (D1-D5), where D1-D3 primarily mediate interactions and D4-D5 contribute to receptor dimerization and stability. A single transmembrane anchors the receptor to the , while the intracellular portion includes a juxtamembrane and a bilobed interrupted by a kinase insert, enabling autophosphorylation upon activation. Binding of the M-CSF homodimer to CSF1R occurs with high affinity, characterized by a (Kd) of approximately 0.1-0.4 nM on intact cells, though the isolated ectodomain exhibits lower affinity (~20 nM at 37°C) due to the stabilizing influence of the transmembrane region. This interaction induces rapid receptor dimerization, primarily through homotypic contacts in the D4 domain, leading to conformational changes that position the intracellular domains for trans-autophosphorylation. In addition to M-CSF, interleukin-34 (IL-34) binds CSF1R as an alternative with comparable affinity, engaging primarily the D1-D3 domains via hydrophobic interactions despite structural and sequence differences from M-CSF, thus eliciting similar dimerization. CSF1R expression is largely restricted to myeloid lineage cells, with high levels on circulating monocytes, tissue-resident macrophages, osteoclasts, and , where it supports their and responsiveness to . Lower expression occurs on other cells such as myeloid dendritic cells and certain neural progenitors. The receptor exists in multiple isoforms, primarily from post-translational modifications. A soluble form of CSF1R, generated by proteolytic cleavage of the membrane-bound receptor, circulates at low levels and functions as a by sequestering M-CSF and IL-34 to regulate ligand availability and attenuate signaling in inflammatory contexts.

Signaling Pathways

Upon binding of macrophage colony-stimulating factor (M-CSF) to the colony-stimulating factor 1 receptor (CSF1R), the receptor undergoes dimerization, which triggers autophosphorylation at specific intracellular residues, including Y723, Y807, and Y921. These phosphorylation events create docking sites for various signaling molecules, initiating intracellular essential for myeloid cell responses. The primary downstream pathways activated include the PI3K/AKT pathway, which promotes cell survival and proliferation through phosphorylation at Y723; the MAPK/ERK pathway, driving differentiation; and the , facilitating gene expression changes. Src family kinases are recruited early via sites like Y559 (juxtamembrane), contributing to amplification of these signals and cytoskeletal reorganization. Downstream, these cascades activate transcription factors such as PU.1, which regulates myeloid differentiation, and c-Fos, involved in proliferative responses, while suppressors of cytokine signaling (SOCS) proteins provide to attenuate pathway activity. In macrophages, M-CSF-induced signaling leads to cytoskeletal rearrangements, including actin polymerization and membrane ruffling, which support through Rac2 and WAVE2-Abi1 complexes. The duration of signaling is critical: sustained activation of ERK and AKT pathways from endosomal CSF1R supports long-term survival, whereas transient signaling promotes motility and short-term responses.

Biological Functions

Role in Hematopoiesis and Immunity

Macrophage colony-stimulating factor (M-CSF), also known as CSF1, plays a central role in hematopoiesis by promoting the and of monocyte-macrophage progenitors derived from hematopoietic stem cells. It acts as a key that supports the commitment of myeloid precursors toward the monocytic lineage, facilitating their maturation into monocytes and macrophages. This process is essential for steady-state , ensuring the continuous production of these cells under normal physiological conditions. In the context of immunity, M-CSF enhances the survival of macrophages by inhibiting through activation of pathways such as , thereby prolonging their functional lifespan in tissues. It also augments key macrophage functions, including of pathogens and debris. In certain activated states, M-CSF can enhance production of proinflammatory cytokines such as IL-1 and TNF. Additionally, M-CSF induces in macrophages via phosphatidylinositol 3-kinase (PI3K) signaling, enabling their recruitment to sites of or . These activities collectively bolster innate immunity by maintaining a robust population of active macrophages. M-CSF further contributes to immune through its involvement in osteoclastogenesis, where it drives the of precursors into osteoclasts, supporting as part of the skeletal immune interface. It also activates for enhanced , priming them to recognize and eliminate malignant cells via direct contact or secreted factors. Studies in animal models underscore these roles: Csf1 mice, such as the osteopetrotic (op/op) strain, exhibit severe deficiencies in and due to impaired , leading to reduced cellularity and immunological deficits; administration of recombinant M-CSF rescues these defects by restoring numbers and function to near-normal levels.

Roles in Other Physiological Processes

Macrophage colony-stimulating factor (M-CSF), also known as CSF-1, plays essential roles in developmental processes beyond hematopoiesis, particularly in reproductive tissues. In placental development, M-CSF is critical for the and invasion of cells, which are vital for proper implantation and placental formation. It stimulates the growth and differentiation of placental s, enhancing their secretion of and supporting overall placental expansion. Studies have shown that CSF-1 signaling directly promotes the of placenta-derived cells, underscoring its necessity for embryonic support during early . Similarly, during , M-CSF facilitates mammary gland branching by regulating ductal elongation and alveolar development. In mice lacking functional CSF-1, such as those with targeted disruptions, there is significantly reduced ductal growth and branching in the s, highlighting M-CSF's indispensable role in preparing the gland for . In tissue homeostasis, M-CSF contributes to the maintenance and function of specialized cell populations across various organs. In the , M-CSF regulates proliferation and activation, promoting their phagocytic activity and providing by modulating inflammatory responses in the . For instance, local delivery of M-CSF enhances microglial density, reduces sizes following , and supports functional , demonstrating its protective effects on neural tissue. Recent studies from the 2020s have further elucidated M-CSF's involvement in microglial-mediated during neural development, where CSF-1/CSF1R signaling ensures proper circuit refinement by yolk sac-derived , preventing abnormal connectivity. In , M-CSF mediates hyperplasia and differentiation, influencing the physiological expansion of fat depots and ; themselves produce M-CSF, which in turn supports recruitment and tissue remodeling. M-CSF also plays a role in cholesterol homeostasis by promoting cholesterol efflux in macrophages via ABCA1 expression. Regarding , its upregulation by in the female reproductive tract enhances endometrial receptivity and supports . Additional physiological effects of M-CSF include its promotion of through recruitment and . M-CSF accelerates tissue repair by increasing infiltration at wound sites, favoring an M2-like that resolves and promotes and extracellular matrix deposition. This is evident in models where M-CSF supplementation enhances density and shifts their profile toward wound-healing functions. The importance of M-CSF in is further illustrated by studies on osteopetrotic (op/op) mice, which carry a in the Csf1 leading to CSF-1 deficiency; these mutants exhibit severe alongside due to impaired placental development and reduced function, with pregnancies failing despite viable embryos. In female reproduction, directly upregulates M-CSF secretion from granulosa cells and uterine tissues, integrating hormonal cues with immune modulation to optimize .

Clinical Relevance

Involvement in Diseases

Macrophage colony-stimulating factor (M-CSF), also known as CSF1, plays a pathological role in various inflammatory diseases by promoting activation and survival. In , M-CSF is upregulated in vascular lesions and facilitates the differentiation of into foam cells, which accumulate lipids and contribute to plaque formation and progression. Elevated M-CSF levels enhance recruitment to arterial walls, exacerbating and lesion development in hyperlipidemic models. Similarly, in (RA), M-CSF is overexpressed in and tissues, driving the activation and proliferation of synovial that sustain chronic joint and tissue destruction. This activation supports the differentiation of into osteoclast-like cells, amplifying erosive damage in affected joints. In cancer, M-CSF critically influences tumor-associated macrophages (TAMs) by polarizing them toward a pro-tumor phenotype, which suppresses anti-tumor immunity and promotes angiogenesis, invasion, and . In , high M-CSF expression correlates with increased TAM infiltration and reduced patient survival, as it enhances macrophage-mediated support for tumor cell growth and dissemination. In , M-CSF overexpression drives TAM recruitment and polarization, facilitating transcoelomic through feedback loops involving like CCL18. Recent 2024 studies highlight M-CSF's role in modulating TAM functions in these malignancies, showing that its inhibition reduces pro-tumor macrophage activity and tumor progression in preclinical models. Beyond inflammation and cancer, M-CSF contributes to pathology in other conditions. In acute and chronic kidney injury, M-CSF mediates monocyte infiltration into renal tissues, promoting macrophage accumulation that amplifies tubular damage and impairs repair processes. In osteoporosis, excess M-CSF signaling enhances osteoclast differentiation and activity, leading to imbalanced bone resorption and accelerated bone loss, particularly in estrogen-deficient states. In neurodegenerative diseases, dysregulated M-CSF/CSF1R signaling alters microglial homeostasis, contributing to neuroinflammation and neuronal damage through aberrant microglial proliferation and activation. Specific clinical correlations underscore M-CSF's disease involvement. Serum M-CSF levels are significantly elevated in patients compared to healthy controls, correlating with disease severity and complications such as . Additionally, genetic variants near the CSF1 gene, such as rs11102024, are associated with increased risk, including , by elevating M-CSF expression and .

Biomarkers and Diagnostics

Macrophage colony-stimulating factor (M-CSF), also known as CSF-1, serves as a biomarker in various diseases through measurement of its serum or plasma levels, primarily via enzyme-linked immunosorbent assay (ELISA). In oncology, elevated M-CSF concentrations in serum are associated with cancer progression and poor prognosis; for instance, in epithelial ovarian cancer, median levels reach approximately 633 pg/mL compared to 299 pg/mL in healthy controls. These elevations correlate with tumor burden and are used prognostically, particularly when combined with markers like CA-125 and HE4 for improved diagnostic sensitivity in early-stage detection. In cardiovascular disease, M-CSF acts as a prognostic indicator for atherosclerotic plaque instability, with higher plasma levels predicting adverse events such as by reflecting macrophage-driven in plaques. Similarly, in renal failure, circulating M-CSF levels correlate with the decline in estimated (eGFR), serving as a marker of progressive and associated cardiovascular risks in patients. Specific assays for M-CSF quantification include commercial ELISA kits, such as the Quantikine Human M-CSF kit from R&D Systems, which detect levels in the range of 78.1–5,000 pg/mL with high sensitivity for and samples. Additionally, M-CSF levels often correlate with CSF1R (the M-CSF receptor) expression in tumor tissues, assessed via (IHC), where co-expression in tumor and stromal cells indicates higher histological grades and infiltration. Recent advances in the have integrated M-CSF-related biomarkers with imaging modalities for assessing tumor-associated s (TAMs) in , such as () tracers targeting CSF1R to visualize macrophage density and activity in solid tumors like and cancers. However, limitations include M-CSF's non-specificity, as elevated levels can arise from general unrelated to , potentially reducing diagnostic specificity in non-oncologic contexts.

Therapeutic Applications

As a Therapeutic Agent

Recombinant macrophage colony-stimulating factor (M-CSF), also known as colony-stimulating factor 1 (CSF-1), has been investigated as a primarily for its role in promoting and differentiation and function. In clinical settings, it has been investigated for enhancing protection against opportunistic infections following high-dose and (HSCT) by stimulating the proliferation and maturation of mononuclear essential for immune responses. M-CSF has shown promise in enhancing protection against opportunistic infections, particularly bacterial and fungal pathogens, in immunocompromised patients such as those undergoing HSCT. Preclinical and early clinical studies demonstrate that M-CSF administration increases mature myeloid cell production, improving survival in models of lethal infections post-HSCT. Additionally, M-CSF enhances responses by activating macrophages, which boosts and immune effector functions, potentially augmenting adaptive immunity in strategies. In , topical application of recombinant M-CSF accelerates skin repair by promoting M2-like polarization and regeneration. Experimental evidence indicates that M-CSF treatment on cutaneous wounds increases vascularization, collagen deposition, and re-epithelialization, with obstruction of M-CSF signaling impairing these processes. Theoretical applications include support for function in conditions like to improve bone density, though clinical investigations remain limited to preliminary studies in related myeloid disorders. Pharmacokinetically, recombinant M-CSF exhibits nonlinear elimination, with an initial distribution of approximately 1.9 to 4.1 hours following intravenous , influencing dosing regimens such as 4-8 μg/kg in supportive care protocols. Common side effects are mild and include fever, chills, , and , typically resolving without intervention and occurring in a minority of patients. As of November 2025, developments in M-CSF biosimilars continue to be explored for potential utility in combinations, though no new approvals have been reported beyond investigational uses.

As a Drug Target

Macrophage colony-stimulating factor (M-CSF, also known as CSF-1) and its receptor CSF1R represent a key therapeutic target in diseases driven by dysregulated macrophage activity, particularly through inhibitors that block ligand-receptor interactions or downstream signaling to deplete or reprogram tumor-associated macrophages (TAMs) and reduce pro-inflammatory responses. Targeting this pathway aims to disrupt the supportive role of M2-like macrophages in tumor progression and chronic inflammation without directly administering M-CSF. Inhibitors of the M-CSF/CSF1R axis include monoclonal antibodies against CSF-1, such as lacnotuzumab (MCS110), a high-affinity that blocks CSF-1 binding to CSF1R, leading to depletion in preclinical models of solid tumors. Clinical trials have evaluated MCS110 in combination with , including a phase II randomized study in advanced where it was combined with and , demonstrating target engagement but comparable antitumor activity to alone, with manageable safety including and . Small-molecule CSF1R kinase inhibitors, such as pexidartinib (PLX3397), selectively inhibit CSF1R , depleting and showing efficacy in phase III trials for , where it achieved a 39% objective response rate (ORR) compared to 0% with ; long-term data as of 2025 confirm sustained clinical benefits with improved ORR. Another example, ARRY-382, an oral CSF1R inhibitor, has been tested in phase Ib/II trials combined with for advanced solid tumors, including , resulting in partial responses in 20% of patients and stable disease in 40%, with evidence of reduced counts indicating pathway blockade. In cancer therapy, M-CSF/CSF1R inhibition prevents macrophage polarization, enhances antitumor immunity, and synergizes with immune checkpoint inhibitors like PD-1 blockers to improve outcomes in preclinical models of and . For instance, pexidartinib combined with in phase I/II trials for advanced solid tumors, including , showed an ORR of approximately 30-40% in select cohorts, attributed to increased T-cell infiltration. In autoimmune diseases, such as and experimental autoimmune encephalomyelitis (a model for ), CSF1R inhibitors like JNJ-40346527 and Ki20227 reduce macrophage-driven inflammation and activity, with phase II data in RA demonstrating reduced disease activity scores when combined with disease-modifying antirheumatic drugs. Despite these advances, challenges include on-target toxicities such as monocytopenia and elevated liver enzymes due to broad myeloid cell depletion, observed in up to 20% of patients on CSF1R inhibitors like pexidartinib, necessitating dose adjustments. Preclinical successes, however, highlight reduced in mouse models of through CSF-1 blockade at tumor sites, supporting ongoing efforts to optimize combinations for clinical . In February 2025, the US Food and Drug Administration approved vimseltinib, another CSF1R inhibitor, for based on phase 3 trial data demonstrating significant antitumor responses. As of November 2025, dozens of clinical trials are exploring CSF1/CSF1R inhibitors, primarily in , with emerging data in autoimmune indications.

Molecular Interactions

Protein-Protein Interactions

Macrophage colony-stimulating factor (M-CSF, also known as CSF-1) primarily binds to its cognate receptor, colony-stimulating factor 1 receptor (CSF1R, also called CD115 or c-Fms), a receptor expressed on myeloid cells. This interaction occurs with high confidence, as indicated by a STRING database score of 900, reflecting robust experimental from co-immunoprecipitation and structural studies. The binding of dimeric M-CSF to the extracellular domains D1-D3 of CSF1R induces receptor dimerization and autophosphorylation, initiating downstream signaling, though M-CSF itself does not form higher-order multimers beyond its native dimeric structure. In addition to CSF1R, M-CSF associates indirectly with , particularly αvβ3, to facilitate in macrophages and osteoclasts. Upon M-CSF stimulation, CSF1R recruits αvβ3 to adhesion sites, stabilizing contacts and promoting cytoskeletal reorganization, as demonstrated by co-immunoprecipitation and assays. CSF1R, in turn, interacts with several intracellular proteins to propagate signals. at 721 (Y721) on CSF1R enables direct binding to the p85β regulatory subunit of (PIK3R2), confirmed through co-immunoprecipitation in macrophages, which activates the PI3K pathway briefly for motility. Similarly, 697 (Y697) recruits GRB2, an adaptor protein that links to the /Ras/MEK/ERK cascade, as shown in binding assays with phosphopeptides. SHP-1 (PTPN6), a , associates with CSF1R upon ligand stimulation, undergoing and modulating receptor activity, evidenced by co-immunoprecipitation in microglial cells. Other interactions include soluble forms of CSF1R, which act as decoy receptors by binding M-CSF and sequestering it from membrane-bound CSF1R, thereby attenuating signaling; this has been utilized in overexpression studies to block M-CSF activity in models.

Genetic and Pathway Interactions

Macrophage colony-stimulating factor (M-CSF), encoded by the , participates in complex genetic and pathway networks that regulate and . Genetic variants in CSF1 and its receptor gene CSF1R have been implicated in several diseases. For example, mutations in CSF1R, particularly dominant-negative variants in the domain, cause adult-onset leukoencephalopathy with axonal spheroids and pigmented (ALSP), a rare autosomal dominant disorder characterized by progressive degeneration and microglial dysfunction. These mutations disrupt CSF1R signaling, leading to impaired microglial and neurodegeneration, with onset typically in the 40s to 60s. At the pathway level, M-CSF integrates with the IL-34/CSF1R axis, where both ligands bind the same receptor to drive microglial development and maintenance, though with distinct spatial and temporal requirements. IL-34 and M-CSF activate overlapping downstream signals via CSF1R, including PI3K-AKT and MAPK pathways, but IL-34 additionally engages PTPζ in the , influencing neuronal support. Cross-talk exists with the GM-CSF pathway, where M-CSF and GM-CSF receptors differentially polarize monocytes toward versus pro-inflammatory macrophages, respectively, modulating tumor microenvironments and inflammatory responses. In the cytokine- receptor interaction pathway (hsa04060), M-CSF signaling contributes to broader cytokine networks, facilitating immune cell differentiation and activation through JAK-STAT and other cascades. Regulatory interactions further fine-tune M-CSF expression and activity. MicroRNAs such as miR-21 indirectly regulate CSF1 by targeting PTEN, thereby upregulating M-CSF secretion in cancer cells and promoting recruitment. Upstream, hypoxia-inducible factor-1α (HIF-1α) directly binds the CSF1 promoter under hypoxic conditions, enhancing M-CSF transcription to support survival and in inflamed tissues. Recent 2020s studies have shown that CSF1R inhibition shifts toward a phagocytic state, reducing amyloid-beta plaques but potentially exacerbating in models. These interactions underscore M-CSF's role in balancing immune and .

References

  1. [1]
    Functions of macrophage colony-stimulating factor (CSF1 ... - PubMed
    Nov 4, 2021 · CSF1 is the primary growth factor required for the control of monocyte and macrophage differentiation, survival, proliferation and renewal.
  2. [2]
    Macrophage colony-stimulating factor - Holland-Frei Cancer Medicine
    M-CSF stimulates differentiation of progenitor cells to mature monocytes, and prolongs the survival of monocytes. It enhances cytotoxicity, superoxide ...
  3. [3]
    Regulation and function of macrophage colony-stimulating factor ...
    Macrophage colony-stimulating factor (CSF1) controls the proliferation, differentiation and survival of cells of the macrophage lineage in mammals and birds ( ...
  4. [4]
    Macrophage colony-stimulating factor and its role in the tumor ...
    Apr 3, 2024 · This study delves into the biological properties and functions of M-CSF in regulating tumor-associated macrophages (TAMs) and its role in modulating host ...Introduction · Regulatory role of M-CSF on... · Clinical applications of M-CSF...<|control11|><|separator|>
  5. [5]
    The origins of the identification and isolation of hematopoietic stem ...
    In 1961, Till and McCulloch published the first of their breakthrough series of experiments that indicated that (1) hematopoiesis could be studied as a ...
  6. [6]
    Ernest Armstrong McCulloch. 21 April 1926—20 January 2011
    Aug 30, 2017 · The discovery of stem cells. McCulloch and Till are best known for their work in the 1960s on identifying haematopoietic stem cells (HSCs).
  7. [7]
    The Nature and Action of Granulocyte-Macrophage Colony ...
    Granulocyte-macrophage colony stimulating factor (GM-CSF) stimulates the in vitro proliferation and differentiation of granulocytic and macrophage cells.
  8. [8]
    CSF-1 Receptor Signaling in Myeloid Cells - PMC - PubMed Central
    The glycoprotein, colony-stimulating factor-1 (CSF-1), also known as macrophage-CSF (M-CSF), was the first of the CSFs to be purified (Stanley and Heard 1977) ...
  9. [9]
    Molecular Cloning of a Complementary DNA Encoding ... - Science
    Complementary DNA (cDNA) clones encoding human macrophage-specific colony-stimulating factor (CSF-1) were isolated. One cDNA clone codes for a mature ...
  10. [10]
    Human CSF-1: gene structure and alternative splicing of mRNA ...
    Characterization of the human CSF-1 gene shows that it contains 10 exons and 9 introns, which span 20 kb. Analysis of multiple CSF-1 transcripts demonstrates ...
  11. [11]
    Apparent role of the macrophage growth factor, CSF-1, in ... - Nature
    Dec 3, 1987 · These findings indicate that CSF-1, under hormonal influence, plays a role in placental development and function.
  12. [12]
    1435 - Gene ResultCSF1 colony stimulating factor 1 [ (human)] - NCBI
    Aug 19, 2025 · CSF1 is expressed by the intestinal epithelial cells to regulate Mphi macrophages and maintain epithelial homeostasis and is downregulated in ...Missing: chromosome | Show results with:chromosome
  13. [13]
    Csf1 colony stimulating factor 1 (macrophage) [ (house mouse)] - NCBI
    Sep 24, 2025 · CSF1 was expressed in astrocytes and neurons in normal mouse spinal cord. CSF1 upregulation in CNS may play an important role in the ...Missing: chromosome | Show results with:chromosome
  14. [14]
    Entry - *120420 - COLONY-STIMULATING FACTOR 1; CSF1 - OMIM
    CSF1 is a cytokine required for differentiation of monocyte lineage cells (eg, tissue macrophages, osteoclasts, and microglia) during development.
  15. [15]
    Human CSF-1: gene structure and alternative splicing of ... - PubMed
    Characterization of the human CSF-1 gene shows that it contains 10 exons and 9 introns, which span 20 kb.
  16. [16]
    CSF1 - Macrophage colony-stimulating factor 1 | UniProtKB - UniProt
    Cytokine that plays an essential role in the regulation of survival, proliferation and differentiation of hematopoietic precursor cells.<|control11|><|separator|>
  17. [17]
    Colony Stimulating Factor 1 - an overview | ScienceDirect Topics
    Macrophage-colony stimulating factor (M-CSF), also known as colony stimulating factor-1 (CSF-1), was the first CSF to be isolated after purification from the ...Missing: recognized | Show results with:recognized
  18. [18]
    Functional evolution of the colony-stimulating factor 1 receptor ...
    Sep 5, 2019 · The contact amino acids in both CSF1 and CSF1R diverged among avian species. Ligand-binding domain swaps between chicken and zebra finch CSF1 ...
  19. [19]
    Proteoglycan form of macrophage colony-stimulating factor binds ...
    We recently isolated a proteoglycan form of macrophage colony-stimulating factor (PG-M-CSF) that carries a chondroitin sulfate glycosaminoglycan chain.
  20. [20]
    Three-dimensional structure of dimeric human recombinant ...
    The structure of a recombinant human M-CSF dimer, determined at 2.5 angstroms by x-ray crystallography, contains two bundles of four alpha helices laid end-to- ...Missing: crystal PDB 3EII
  21. [21]
    CSF1 Gene - GeneCards | CSF1 Protein | CSF1 Antibody
    The protein encoded by this gene is a cytokine that controls the production, differentiation, and function of macrophages. The active form of the protein is ...
  22. [22]
    Local M-CSF (Macrophage Colony-Stimulating Factor) Expression ...
    Oct 22, 2020 · Our studies show that CSF1 is required for the expansion of macrophages in lesions, promoting both survival and growth. Our experiments, using ...
  23. [23]
    Macrophages and CSF-1: Implications for development and beyond
    This review summarizes the key functions of macrophages and their primary regulator, colony-stimulating factor (CSF)-1, during development.Classification Of Macrophage... · Macrophages In Organogenesis · Csf-1 And Macrophage...
  24. [24]
    Macrophage colony-stimulating factor and its role in the tumor ...
    Apr 4, 2024 · This study delves into the biological properties and functions of M-CSF in regulating tumor-associated macrophages (TAMs) and its role in modulating host ...
  25. [25]
    Diverse in vivo effects of soluble and membrane-bound M-CSF ... - NIH
    Targeting tumor-infiltrating macrophages decreases tumor-initiating cells, relieves immunosuppression, and improves chemotherapeutic responses. Cancer Res.Missing: osteoblasts fibroblasts
  26. [26]
    Effects of Macrophage Colony-Stimulating Factor (M-CSF) on ...
    The priming effect of MCSF on LPS-induced cytokine production was found to require 3-day preincubation, and reached a maximum at the concentration of 104 U/ml.
  27. [27]
    CSF1R Gene - Colony Stimulating Factor 1 Receptor - GeneCards
    Protein attributes for CSF1R Gene. Size: 972 amino acids; Molecular mass: 107984 Da. Protein existence level: PE1. Quaternary ...
  28. [28]
  29. [29]
    CSF1R - Macrophage colony-stimulating factor 1 receptor - UniProt
    Tyrosine-protein kinase that acts as a cell-surface receptor for CSF1 and IL34 and plays an essential role in the regulation of survival, proliferation and ...
  30. [30]
  31. [31]
  32. [32]
    The M-CSF receptor in osteoclasts and beyond - PMC - NIH
    CSF1R protein expression is restricted to myeloid cells and is much higher in tissue resident macrophages than in blood monocytes. CSF1R signal transduction ...
  33. [33]
    A novel soluble form of the CSF-1 receptor inhibits proliferation of ...
    These soluble receptors result from alternative splicing of the mRNA species, which leads to the production of a truncated protein that has the ...Missing: isoform decoy
  34. [34]
    The M-CSF receptor in osteoclasts and beyond - Nature
    Aug 17, 2020 · CSF1. CSF1 (also known as macrophage colony-stimulating factor (M-CSF)) is a ligand for CSF1R and a growth factor that regulates the survival, ...
  35. [35]
    Signaling by CSF1 (M-CSF) in myeloid cells - Reactome
    Colony stimulating factor-1 (CSF1, CSF-1, also called macrophage colony stimulating factor, M-CSF) is a disulfide-linked dimer that stimulates the ...Missing: via | Show results with:via
  36. [36]
    Regulation of myelopoiesis by proinflammatory cytokines in ...
    ... M-CSFR [93]. M-CSF is broadly expressed and detected in the blood circulation and regulates steady-state myelopoiesis. M-CSFR is predominantly expressed by ...
  37. [37]
    M-CSF Induces Monocyte Survival by Activating NF-κB p65 ... - NIH
    Dec 22, 2011 · Macrophage colony-stimulating factor (M-CSF) promotes mononuclear phagocyte survival and proliferation. The transcription factor Nuclear ...
  38. [38]
    M-CSF increases proliferation and phagocytosis while modulating ...
    We show here that the cytokine M-CSF dramatically influences the phenotype of adult human microglia. These results pave the way for future investigation.
  39. [39]
    Macrophage colony-stimulating factor and its receptor signaling ...
    M-CSF alone did not induce cytokine release; however, costimulation with M-CSF and GA for 24 h produced a significant increase in the release of TNF-□ and IL-1β ...<|control11|><|separator|>
  40. [40]
    Phosphorylation of CSF-1R Y721 mediates its association with PI3K ...
    The CSF-1 receptor (CSF-1R) transduces these pleiotropic signals through autophosphorylation of eight intracellular tyrosine residues. We have used a novel bone ...
  41. [41]
    Macrophages can recognize and kill tumor cells bearing ... - PubMed
    This study indicates that mM-CSF provides the necessary binding and triggering molecules through which macrophages can initiate direct tumor cell cytotoxicity.
  42. [42]
    Effects of Macrophage Colony-Stimulating Factor on ... - PubMed - NIH
    The former three populations expanded to normal levels in op/op mice after daily M-CSF administration, indicating that they are developed and differentiated due ...
  43. [43]
    Local macrophage colony-stimulating factor expression regulates ...
    Previous studies have shown that deficiency of macrophage colony-stimulating factor (M-CSF or CSF1) dramatically reduces atherosclerosis in hyperlipidemic mice.
  44. [44]
    Arterial colony stimulating factor-1 influences atherosclerotic lesions ...
    The data also suggest that the decreased atherosclerosis observed in CSF-1 deficient mice is not attributable to fewer circulating monocytes. CSF-1 ...
  45. [45]
    Managing Macrophages in Rheumatoid Arthritis by Reform or ...
    Expression of GM-CSF and M-CSF is increased in synovial fluid from RA patients, and they form a cytokine network between myeloid cells and neighboring cells, ...
  46. [46]
    Rheumatoid arthritis synovial macrophage-osteoclast differentiation ...
    ... M-CSF are the only humoral factors required for RA synovial macrophage-osteoclast differentiation. OPG was found to inhibit osteoclast formation by RA ...
  47. [47]
    Functional Relationship between Tumor-Associated Macrophages ...
    The critical role of M-CSF in the turn-over of TAM is reflected in the drastic reduction in macrophages in the primary tumor at different stages of tumor ...
  48. [48]
    Tumor-associated macrophages induced spheroid formation ... - NIH
    Dec 25, 2021 · Tumor-associated macrophages induced spheroid formation by CCL18-ZEB1-M-CSF feedback loop to promote transcoelomic metastasis of ovarian cancer ...Result · Tams Produced Ccl18 To... · Zeb1 In Ovca Cells Is...
  49. [49]
    CSF-1 signaling mediates recovery from acute kidney injury - PMC
    Nov 12, 2012 · In response to DT injection, renal Csf1 mRNA levels were increased as early as 2 days after DT injection, and immunostaining determined that ...
  50. [50]
    Estrogen blocks M-CSF gene expression and osteoclast formation ...
    Estrogen replacement fails to block M-CSF mRNA expression and osteoclast formation in ovariectomized mice lacking Egr-1.
  51. [51]
    Colony stimulating factor 1: friend or foe of neurons? - PMC
    In the homeostatic brain, a baseline level of CSF1 helps to maintain microglial ... CSF1/CSF1R system is neuroprotective in many neurodegenerative models.
  52. [52]
    Association between serum macrophage colony-stimulating factor ...
    Serum M-CSF levels are significantly higher in neonates with sepsis. High serum M-CSF levels may have a possible role in the pathogenesis of thrombocytopenia ...
  53. [53]
    Genetic Association and Expression Correlation between Colony ...
    Feb 14, 2020 · Our data suggest that genetic variants near CSF1 are associated with AOSD and the rs11102024 T allele links to higher M-CSF levels and systemic ...
  54. [54]
    IL-6, M-CSF and IAP Cytokines in Ovarian Cancer - Karger Publishers
    Oct 14, 1999 · All cytokines were found to have scattered levels: M-CSF levels ranged from 3 to 5,696 pg/ml (median 1,230 pg/ml) and were skewed toward the ...
  55. [55]
    Diagnostic Power of Selected Cytokines, MMPs and TIMPs in ...
    The SE value in the ovarian cancer group was the highest for HE4 (78.0%) and MMP-7 (78.0%). The diagnostic SP was the highest for M-CSF, VEGF and MMP-9 (95.2%; ...
  56. [56]
    M-CSF in a new biomarker panel with HE4 and CA 125 in the ...
    May 3, 2015 · Our results have demonstrated significant differences in the concentration levels of M-CSF, CA 125 and HE4 between the groups of ovarian cancer ...
  57. [57]
    Potential Clinical Utility of Macrophage Colony-stimulating Factor ...
    Dec 23, 2019 · It is important that these biomarkers can distinguish patients with stable, inactive plaques from those with unstable, active plaques, and also ...
  58. [58]
    Association of plasma macrophage colony-stimulating factor with ...
    Aug 16, 2019 · A five-year follow-up study found that M-CSF is significantly associated with annual decline in estimated glomerular filtration rate (eGFR) [16] ...
  59. [59]
    Human M-CSF ELISA - Quantikine DMC00B - R&D Systems
    $$45 deliveryThe Quantikine Human M-CSF Immunoassay is a 4.5 hour solid phase ELISA designed to measure human M-CSF in cell culture supernates, serum, plasma, saliva, ...Missing: commercial | Show results with:commercial
  60. [60]
    Expression of M-CSF and CSF-1R is correlated with histological ...
    Conclusion: The expression of M-CSF and CSF-1R in tumor cell areas and adjacent stromal areas correlate with the histological grade of soft tissue tumors.Missing: levels | Show results with:levels
  61. [61]
    Candidate Tracers for Imaging Colony-Stimulating Factor 1 ...
    All of these observations suggest that CSF1R is a viable target for imaging inflammation generally and neuroinflammation specifically. PET imaging of CSF1R ...
  62. [62]
    Macrophages in cardiovascular diseases: molecular mechanisms ...
    May 31, 2024 · M4 macrophages, mostly found in unstable plaques, highly express chemokines, such as CCL2 and CXCL4, and proteases, such as MMP-12, which ...
  63. [63]
    Clinical applications of hematopoietic growth factors - PubMed
    Current clinical uses of hematopoietic growth factors include decreasing cytopenias associated with chemotherapy, those due to congenital or acquired bone ...
  64. [64]
    M-CSF improves protection against bacterial and fungal infections ...
    Oct 10, 2016 · These results encourage potential clinical applications of M-CSF to prevent severe infections after HS/PC transplantation. Subjects ...Missing: post- | Show results with:post-
  65. [65]
    Macrophage colony-stimulating factor (M-CSF) enhances ... - PubMed
    These data indicate that M-CSF enhances the capacity of mature macrophages to release oxygen reduction products, and they are consistent with reports that CSF ...
  66. [66]
    Accelerating skin wound healing by M-CSF through generating ...
    Jul 1, 2016 · Our study further revealed that topical application of M-CSF protein on skin wounds accelerates healing while obstruction of M-CSF by a ...
  67. [67]
    Nonlinear pharmacokinetics of recombinant human macrophage ...
    At 10 mg/kg, two half-lives were initially observed, but after about 20 hr the plasma M-CSF suddenly declined with a steep slope.
  68. [68]
    Macrophage Colony-Stimulating Factor - ScienceDirect.com
    Constitutional symptoms (malaise, fatigue, insomnia, headache, nausea, fever, and chills) sometimes occurred. There was no evidence of long-term toxicity during ...Missing: myalgia | Show results with:myalgia
  69. [69]
    Full article: Antibodies to watch in 2025 - Taylor & Francis Online
    Here, we discuss key events in antibody therapeutics development that occurred during 2024 and forecast key events related to the late-stage clinical pipeline ...<|separator|>
  70. [70]
    Dual roles and therapeutic targeting of tumor-associated ... - Nature
    Aug 25, 2025 · The investigation of TAMs has propelled the consideration of macrophages as targets for cancer therapy, particularly in clinical trials ...
  71. [71]
    Colony-stimulating factor 1 receptor (CSF1R) inhibitors in cancer ...
    Jul 18, 2017 · Early clinical data suggest good tolerability of CSF1/CSF1R-targeting compounds; however, available efficacy data are still limited, with the ...
  72. [72]
    Phase Ib/II Study of Lacnotuzumab in Combination with ...
    Lacnotuzumab (MCS110) is a high-affinity, humanized monoclonal antibody directed against human CSF-1 and it blocks the interaction of CSF-1 with CSF-1R on TAMs.
  73. [73]
    A Randomized Phase II Study of Anti-CSF1 Monoclonal Antibody ...
    Jan 1, 2022 · Despite successful target engagement and anticipated pharmacokinetic profile, lacnotuzumab + gem-carbo showed comparable antitumor activity ...
  74. [74]
    Pexidartinib, a Novel Small Molecule CSF-1R Inhibitor in Use ... - NIH
    May 4, 2020 · Other than TGCT, pexidartinib has been studied pre-clinically and clinically in gastrointestinal stromal tumors, colorectal cancer, pancreatic ...
  75. [75]
    ARRY-382 in Combination with Pembrolizumab in Patients ... - NIH
    This Phase 1b/2 study evaluated the selective CSF-1 receptor (CSF-1R) inhibitor, ARRY-382, plus the anti–PD-1 agent, pembrolizumab, in patients with advanced ...
  76. [76]
    CSF1/CSF1R Signaling Inhibitor Pexidartinib (PLX3397 ...
    Preclinical studies have shown that CSF1R blockade may inhibit tumor growth in several cancer models through the elimination or polarization of TAMs (14, 15).
  77. [77]
    NCT02452424 | A Combination Clinical Study of PLX3397 and ...
    The goal of this clinical research study is to learn how PLX3397 and pembrolizumab work together to affect cancer cells.
  78. [78]
    Results from a Phase IIA Parallel Group Study of JNJ-40346527, an ...
    Oct 1, 2015 · JNJ-40346527, a selective CSF-1R kinase inhibitor, and traditional DMARD may have complementary effects in reducing RA inflammation.
  79. [79]
    The selective M-CSF receptor tyrosine kinase inhibitor Ki20227 ...
    Recombinant CSF-1, antibodies against the ligand and the ... This review examines the potential clinical uses of modulators of the CSF-1/CSF-1R system.
  80. [80]
    Challenges and prospects of CSF1R targeting for advanced ...
    CSF1R expression modulates tumor-associated macrophages, making CSF1R blockade an appealing immune-modulating therapeutic target.
  81. [81]
    Targeting CSF-1 signaling between tumor cells and macrophages at ...
    Jul 11, 2025 · Targeting CSF-1 signaling between tumor cells and macrophages at TMEM doorways inhibits breast cancer dissemination ... ovarian, breast, and ...
  82. [82]
    Clinical evaluation of colony-stimulating factor 1 receptor inhibitors
    Signaling through colony-stimulating factor 1 receptor (CSF1R) regulates the development, differentiation, and activation of mononuclear phagocytic cells.<|control11|><|separator|>
  83. [83]
    CSF1 protein (human) - STRING interaction network
    CSF1 CSF1 CSF1R CSF1R IL34 IL34 CSF2 CSF2 EGF EGF ... Minimum required interaction score: highest confidence (0.900), high confidence (0.700), medium ...
  84. [84]
    Three-Dimensional Structure of Dimeric Human Recombinant ...
    The structure of a recombinant human M-CSF dimer, determined at 2.5 angstroms by x-ray crystallography, contains two bundles of four α helices laid end-to-end.Missing: multimerization | Show results with:multimerization
  85. [85]
    M-CSF induces the stable interaction of cFms with α V β 3 integrin in ...
    In addition, M-CSF stimulation initiated recruitment of αVβ3 integrin to adhesion contacts and also induced the association of αVβ3 integrin with signaling ...
  86. [86]
    Convergence of α v β 3 Integrin–And Macrophage Colony ...
    In addition, we found that in Src-deficient cells, M-CSF initiated the recruitment of αvβ3 integrin and PLC-γ to adhesion contacts. M-CSF also induced the ...
  87. [87]
    Colony-stimulating Factor-1 Receptor Utilizes Multiple Signaling ...
    We have investigated the signaling mechanism of CSF-1-induced D2 expression. Analyses of CSF-1 receptor autophosphorylation mutants show that, although certain ...Missing: Y921 | Show results with:Y921
  88. [88]
    A Novel Role for Protein Tyrosine Phosphatase SHP1 in Controlling ...
    Feb 1, 2001 · Stimulation of microglial cells with CSF-1 induced increase in tyrosine phosphorylation of SHP1 and CSF-1-R, reaching a maximum after 30 min, ...
  89. [89]
    Alleviation of neuropathic pain by over-expressing a soluble colony ...
    Sep 9, 2020 · Such findings indicate that blocking CSF1 signalling may be an effective approach in treating nerve injury-induced neuropathic pain. Indeed, it ...
  90. [90]
    Clinical and genetic characterization of adult‐onset ... - NIH
    CSF1R mutations account for 3.5% (5/149) of the adult‐onset leukoencephalopathy in Taiwan. CSF1R mutations outside the tyrosine kinase domain may also disturb ...
  91. [91]
    ALSP - Adult onset leukoencephalopathy with axonal spheroids and ...
    CSF1R related leukoencephalopathy is caused by mutations in the CSF1R gene. People usually begin experiencing symptoms in their 40's-60's although this may ...
  92. [92]
    Analysis of the human monocyte-derived macrophage transcriptome ...
    Mar 6, 2017 · We used these data to re-examine the hundreds of loci associated with susceptibility to inflammatory bowel disease (IBD) in genome-wide association studies.
  93. [93]
    The twin cytokines interleukin-34 and CSF-1: masterful conductors ...
    Jan 1, 2021 · CSF-1 and IL-34 share a common receptor, the macrophage-colony stimulating factor receptor (CSF-1R), which is activated in a similar way by both ...
  94. [94]
    CSF1R Ligands IL-34 and CSF1 Are Differentially Required for ...
    We show that while CSF1 is required to establish microglia in the developing embryo, both CSF1 and IL-34 are required beginning in early postnatal development.
  95. [95]
    M-CSF and GM-CSF Receptor Signaling Differentially Regulate ...
    M-CSF and GM-CSF receptor signaling differentially regulate monocyte maturation and macrophage polarization in the tumor microenvironment.
  96. [96]
    hsa04060 - KEGG PATHWAY
    Cytokines are released by various cells in the body, usually in response to an activating stimulus, and they induce responses through binding to specific ...Missing: CSF | Show results with:CSF
  97. [97]
    miR-21 is targeted by omega-3 polyunsaturated fatty acid to regulate ...
    miR-21 targets PTEN to regulate CSF-1 expression in breast cancer cells We have shown above that PTEN and miR-21 independently regulate expression of CSF-1 ( ...
  98. [98]
    Hypoxia-inducible factor-dependent signaling between triple ... - NIH
    Taken together, the ChIP data demonstrate that the human CSF1 and CCR5 genes are directly regulated by both HIF-1 and HIF-2. CCL5 and CSF1 Stimulate Macrophage ...
  99. [99]
    Switch to phagocytic microglia by CSFR1 inhibition drives amyloid ...
    Aug 23, 2024 · Colony stimulating factor 1-receptor (CSF-1R) is predominantly expressed on microglia and its levels are significantly increased in ...Missing: 2020s | Show results with:2020s
  100. [100]
    A multimodal approach of microglial CSF1R inhibition and GENUS ...
    Jul 16, 2025 · The CSF1R inhibitor PLX3397, an FDA-approved treatment for a rare cancer, has been shown to reduce microglia count, lower inflammation, and ...Missing: 2020s | Show results with:2020s