M CSF Human

Macrophage-Colony Stimulating Factor Human Recombinant
Shipped with Ice Packs
In Stock

Description

Molecular Structure and Isoforms

M-CSF exists in multiple isoforms generated through alternative splicing and post-translational modifications :

IsoformMolecular WeightModificationsCellular Localization
Full-length transmembrane140 kDa (dimer)Covalent dimer, type I membraneCell surface
Secreted glycoprotein86 kDa (dimer)N- and O-glycosylationExtracellular matrix
Proteoglycan-modified200 kDa (subunit)Chondroitin sulfate, glycosylationCirculating or immobilized
Truncated secreted44 kDa (dimer)N-glycosylationExtracellular fluid

The N-terminal 150 amino acids are essential for binding to the CSF-1 receptor (CSF-1R/c-Fms) . Human M-CSF shares 74–88% amino acid identity with mammalian homologs but exhibits species-specific activity (e.g., mouse M-CSF is inactive in humans) .

Biological Functions

M-CSF regulates myeloid and non-hematopoietic systems through CSF-1R signaling :

Key Roles in Immunity and Homeostasis

  • Macrophage Development: Drives proliferation and differentiation of monocyte progenitors into mature macrophages .

  • Osteoclastogenesis: Supports survival and differentiation of osteoclast precursors .

  • Microglial Activation: Enhances phagocytosis of amyloid-β in adult human microglia .

  • Pregnancy: Elevated levels during gestation promote decidual and placental growth .

Functional Modulation

  • Immune Response: Increases macrophage tumoricidal activity, microbial killing, and cytokine production (e.g., IL-6, TNF-α) .

  • Metabolic Regulation: Influences lipid metabolism and atherosclerosis progression .

In Vivo Studies

  • Humanized Mice: M-CSF expression rescued monocyte/macrophage development blocked at the promonocyte stage, enhancing protection against influenza and Mycobacterium infections .

  • Microglia: M-CSF treatment increased microglial proliferation (2.5-fold), phagocytosis of Aβ (3-fold), and reduced HLA-DP/DQ/DR expression .

Clinical Observations

ConditionM-CSF RoleOutcome
Acute Myeloid LeukemiaPost-chemotherapy administrationReduced neutropenia duration
AtherosclerosisParadoxical lipid-lowering vs. pro-inflammatoryContradictory disease modulation
Alzheimer’s DiseaseMicroglial activationEnhanced Aβ clearance

Clinical Applications

  • Therapeutic Use: Recombinant human M-CSF (rhM-CSF) shortens recovery periods post-chemotherapy and improves infection resistance .

  • Biotechnological Production: Expressed in E. coli or insect cells, with bioactivity validated via cell proliferation assays (e.g., ED₅₀ ≤10 ng/mL for NFS-60 cells) .

Pathological Associations

  • Cancer: High M-CSF levels correlate with tumor angiogenesis (via VEGF induction) and poor prognosis .

  • Autoimmunity: Linked to pulmonary fibrosis and rheumatoid arthritis through macrophage hyperactivation .

  • Reproductive Health: Deficiency implicated in recurrent spontaneous abortions .

Receptor and Signaling

CSF-1R (c-Fms) is a tyrosine kinase receptor shared with IL-34. Ligand binding induces receptor dimerization, internalization, and activation of downstream pathways (e.g., PI3K/AKT, MAPK) . Cross-species specificity allows human M-CSF to activate mouse receptors, facilitating preclinical studies .

Product Specs

Introduction
Granulocyte/Macrophage Colony-Stimulating Factors (GM-CSFs) are cytokines that play a crucial role in hematopoiesis, the process of blood cell formation. They regulate the production, differentiation, and function of granulocytes and monocytes-macrophages, two types of white blood cells. MCSF specifically stimulates the production and development of cells belonging to the monocyte/macrophage lineage. MCSF is involved in various physiological processes, including immune responses, bone metabolism, the removal of lipoproteins from the bloodstream, fertility, and pregnancy.
Description

Recombinant Human Macrophage Colony Stimulating Factor, produced in E. coli, is a non-glycosylated, disulfide-linked homodimer. Each polypeptide chain consists of 159 amino acids, resulting in a total molecular mass of 37.1 kDa. The purification process of MCSF involves proprietary chromatographic techniques.

Physical Appearance
The product appears as a sterile, white powder that has been lyophilized (freeze-dried).
Formulation
The MCSF protein was lyophilized in a buffer consisting of 10mM sodium phosphate at pH 8.0 and 50mM NaCl.
Solubility
To reconstitute the lyophilized MCSF, it is recommended to dissolve it in sterile 18M-cm H2O at a concentration of at least 100 µg/ml. This solution can then be further diluted as needed in other aqueous solutions.
Stability
Lyophilized MCSF remains stable at room temperature for up to 3 weeks. However, for long-term storage, it is recommended to store the desiccated product below -18°C. Once reconstituted, MCSF should be stored at 4°C for a period of 2-7 days. For extended storage, it is advisable to store the reconstituted MCSF below -18°C. Repeated freeze-thaw cycles should be avoided to maintain product integrity.
Purity

The purity of the MCSF is determined by SDS-PAGE analysis and is found to be greater than 95.0%.

Biological Activity

The biological activity of MCSF is determined by its ability to stimulate the proliferation of murine M-NFS-60 indicator cells. The ED50 value, which represents the concentration of MCSF required to achieve half-maximal stimulation, is 1.15 ng/ml. This corresponds to a specific activity of 8.7 x 105 Units/mg.

Synonyms
Macrophage Colony Stimulating Factor, CSF-1, Lanimostim, MCSF, MGC31930, M-CSF.
Source
Escherichia Coli.
Amino Acid Sequence
MEEVSEYCSH MIGSGHLQSL QRLIDSQMET SCQITFEFVD QEQLKDPVCY LKKAFLLVQD IMEDTMRFRD NTPNAIAIVQ LQELSLRLKS CFTKDYEEHD KACVRTFYET PLQLLEKVKN VFNETKNLLD KDWNIFSKNC NNSFAECSSQ GHERQSEGS.

Q&A

What is human M-CSF and what are its primary biological functions?

Human Macrophage Colony-Stimulating Factor (M-CSF) is a hematopoietic growth factor that plays essential roles in the differentiation, proliferation, and survival of monocytes and macrophages. It is constitutively expressed in most tissues and readily detected in circulation . M-CSF signals through the c-FMS receptor (CD115), which is a single-pass transmembrane protein tyrosine kinase member of the PDGF family . This cytokine is involved in various biological processes including inflammation, immunity, bone metabolism, and tissue repair.

M-CSF is particularly important in regulating macrophage phenotype and function. Research has shown that M-CSF-derived macrophages are better prepared to transition to a program of tissue repair and display growth-promoting, angiogenic characteristics following stimulation . This contrasts with GM-CSF-derived macrophages, which typically maintain a more inflammatory phenotype.

How do M-CSF-derived macrophages differ from GM-CSF-derived macrophages?

M-CSF and GM-CSF drive fundamentally different macrophage phenotypes with distinct functional capabilities:

CharacteristicM-CSF-Derived MacrophagesGM-CSF-Derived Macrophages
Phenotypic transitionReadily transition to growth-promoting, angiogenic phenotype after stimulationRetain inflammatory phenotype after stimulation
Tissue distributionFrom constitutively expressed M-CSF (most tissues)From GM-CSF (primarily lung, induced during inflammation)
Receptor signalingThrough CD115 (c-FMS)Through CD116 (α/β heterodimeric receptor)
Response to regulatory stimuliMore responsive to pro-resolving mediatorsLess responsive to pro-resolving mediators
Functional biasTissue repair and homeostasisInflammatory response

These differences make M-CSF-derived macrophages better adapted for tissue repair functions, while GM-CSF macrophages undergo more profound activation during inflammatory responses . Understanding these distinctions is crucial when designing experiments to model specific macrophage populations in disease states.

What are the downstream signaling pathways activated by human M-CSF?

M-CSF initiates signaling through binding to its receptor c-FMS (CD115), activating multiple downstream pathways that collectively regulate macrophage development and function. While the search results don't detail all pathways, research has established that M-CSF signaling activates several key cascades, including PI3K/Akt, MAPK/ERK, and JAK/STAT pathways. These signaling mechanisms orchestrate cellular responses including survival, proliferation, differentiation, and cytokine production.

The specific combination and intensity of pathway activation contribute to the distinctive properties of M-CSF-derived macrophages compared to those generated with other growth factors like GM-CSF . These signaling differences ultimately translate to functional differences observed in macrophage populations in various tissue contexts.

What are the optimal conditions for using recombinant human M-CSF in cell culture experiments?

When designing experiments using recombinant human M-CSF, researchers should consider several critical parameters:

  • Storage conditions: Store recombinant human M-CSF at ≤-70°C to maintain protein integrity . Avoid multiple freeze-thaw cycles as this can compromise activity.

  • Concentration range: For monocyte-to-macrophage differentiation, concentrations typically range from 10-100 ng/ml, with 50 ng/ml being commonly used in standard protocols.

  • Administration schedule: For optimal results in differentiation protocols, fresh M-CSF should be added every 2-3 days, as the protein has a limited half-life in culture.

  • Quality assessment: Batch testing for endotoxin contamination is essential, particularly for immunological studies where trace endotoxin can confound results.

  • Co-stimulatory factors: Consider whether additional factors might be required, as continuous M-CSF application in the absence of co-stimulatory signals (e.g., RANKL) might overstimulate hematopoietic lineage development in certain contexts .

Human recombinant M-CSF expressed in E. coli systems is commonly used for research applications , but researchers should verify the source and purification method based on their specific experimental requirements.

How should researchers prepare and analyze samples for M-CSF detection in human specimens?

Sample preparation is critical for accurate M-CSF detection in human specimens:

Following these methodological guidelines ensures more reliable and reproducible M-CSF measurements across different experimental contexts.

What are the most reliable methodological approaches for quantifying human M-CSF in research samples?

Several methodological approaches exist for quantifying human M-CSF, each with specific advantages:

  • Sandwich immunoassays: The MSD 96-Well MULTI-ARRAY and MULTI-SPOT Human M-CSF Ultrasensitive Assay represents a highly sensitive approach, utilizing capture antibodies coated in patterned arrays and detection antibodies labeled with SULFO-TAG reagent . This format offers excellent sensitivity and specificity for serum and plasma samples.

  • Multiplex cytokine analysis: For experimental designs requiring simultaneous measurement of multiple cytokines, bead-based multiplex platforms can quantify M-CSF alongside other analytes of interest.

  • Sample handling optimization: Regardless of detection method, proper sample preparation is critical. For plasma samples, removing additional clotted material by centrifugation after thawing is essential, particularly for heparin-anticoagulated specimens .

  • Assay customization: The typical calibration range for M-CSF assays spans from 0 to 10,000 pg/mL, but researchers may adjust this depending on expected concentrations in their specific samples .

  • Combined detection approaches: For challenging samples, verification with multiple methodologies may be necessary to confirm accurate quantification.

Each quantification method has specific technical requirements and limitations that should be considered in experimental design.

How does M-CSF contribute to bone healing and what methodological considerations are important in fracture models?

M-CSF plays a complex role in bone healing, with several key mechanisms and methodological considerations:

  • Dual functions in bone metabolism: M-CSF critically influences bony healing by recruiting stem cells to fracture sites and impacting hard callus formation through stimulation of osteoclastogenesis .

  • Unexpected anabolic effects: In murine femoral osteotomy models, systemic M-CSF application demonstrated anabolic effects, resulting in significantly larger calluses and increased trabecular thickness compared to wild-type controls without M-CSF treatment .

  • Paradoxical outcomes: Despite increasing callus size and trabecular thickness, M-CSF application did not significantly improve biomechanical properties in murine models . This suggests complex feedback mechanisms between bone formation and resorption that require careful experimental design to elucidate.

  • Hypothesis for mechanism: Continuous M-CSF application in the absence of co-stimulatory signals (e.g., RANKL) might overstimulate the hematopoietic lineage in favor of tissue macrophages rather than osteoclasts . This could potentially explain the unexpected findings in fracture models.

  • Methodological approach: When studying M-CSF in fracture healing, researchers should consider implementing both μCT analysis, histological assessment, and biomechanical testing to fully characterize the effects on bone structure and function .

These findings underscore the importance of comprehensive experimental design when investigating M-CSF's role in bone healing, as effects may vary depending on dose, timing, and presence of co-stimulatory factors.

What are the current challenges in targeting the M-CSF/CSFR1 axis for therapeutic purposes in neurological disorders?

The therapeutic potential of modulating the M-CSF/CSFR1 axis in neurological disorders faces several methodological and translational challenges:

  • Disease-specific dynamics: Low levels of M-CSF were measured in patients with presymptomatic Alzheimer's disease (AD) or mild cognitive impairment (MCI), predicting rapid disease progression toward dementia 2-6 years later . Similarly, decreased mCSF/CSFR1 expression was observed in multiple sclerosis lesions despite increased macrophage/microglial presence . These findings suggest disease-specific roles that must be carefully characterized.

  • Temporal considerations: The effects of M-CSF may vary significantly depending on disease stage, requiring precise timing of therapeutic intervention. This necessitates time-course studies in preclinical models to identify optimal treatment windows .

  • Model selection limitations: Different animal models show varying responses to M-CSF intervention, complicating translation to human applications. Researchers must carefully select models that best recapitulate human disease mechanisms .

  • Age-dependent effects: Findings demonstrate physiological differences between young and elderly subjects, likely reflecting distinct microglial characteristics across age groups . Age-matched controls and age-specific dosing strategies may be necessary in both preclinical and clinical studies.

  • Clinical trial design: While many clinical trials have used M-CSF inhibitors or anti-CSFR1 antibodies, most have focused on cancer or rheumatoid arthritis rather than neurological conditions . Neurological applications require specific trial designs addressing the unique challenges of CNS drug delivery and biomarker assessment.

Addressing these challenges requires integrative approaches combining conditional gene deletion models, time-course studies, and careful translation from preclinical to clinical settings.

How can single-cell RNA sequencing enhance our understanding of M-CSF effects on diverse cell populations?

Single-cell RNA sequencing (scRNA-seq) offers powerful insights into the heterogeneous effects of M-CSF across cell populations:

  • Resolving cellular heterogeneity: scRNA-seq has revealed that M-CSF-derived human macrophages undergo a programmed transition to a growth-promoting, angiogenic phenotype following stimulation, while GM-CSF-derived macrophages retain an inflammatory phenotype . This technology allows researchers to identify distinct macrophage subpopulations with differential responses to M-CSF.

  • Methodological approach: Analysis pipelines for scRNA-seq typically include normalization, variable feature selection, data scaling, neighbor/cluster searches, and differential expression analysis across conditions and samples . For example, a study analyzing M-CSF and GM-CSF macrophage responses evaluated over 17,000 cells with an average of 4,327 cells per experimental condition .

  • Temporal dynamics: scRNA-seq enables tracking of gene expression changes over time, allowing researchers to characterize the temporal sequence of M-CSF-induced phenotypic transitions. This approach can identify early response genes versus later effector programs.

  • Regulatory network identification: By integrating scRNA-seq data with pathway analysis tools, researchers can construct regulatory networks that mediate M-CSF responses, identifying key transcription factors and signaling nodes that drive macrophage phenotype specification.

  • Integration with functional assays: For comprehensive characterization, scRNA-seq findings should be validated with functional assays to confirm that transcriptional changes translate to altered cellular behaviors relevant to disease contexts.

This cutting-edge approach provides unprecedented resolution of M-CSF effects, enabling more precise targeting of specific cellular subsets in therapeutic applications.

How should researchers interpret conflicting data regarding M-CSF effects in different experimental models?

When encountering conflicting data on M-CSF effects, researchers should systematically evaluate several key factors:

  • Context-dependent effects: The mCSF/CSFR1 axis may have different or even opposing effects depending on the disease context . For example, while low mCSF levels predict faster progression in Alzheimer's disease, the role of this pathway may differ in other neurological conditions or in bone healing contexts .

  • Timing considerations: The stage of disease or repair process when M-CSF is administered significantly impacts outcomes. In fracture healing, the temporal sequence of inflammatory and repair phases means M-CSF may have different effects depending on administration timing .

  • Dosage dependencies: M-CSF effects often show non-linear dose-response relationships. In bone healing studies, continuous M-CSF application showed unexpected effects that differed from physiological patterns . Comprehensive dose-response experiments are essential to resolve apparent contradictions.

  • Co-factor requirements: The absence or presence of co-stimulatory signals critically determines M-CSF outcomes. Without RANKL, continuous M-CSF application may overstimulate hematopoietic lineage development toward tissue macrophages rather than osteoclasts .

  • Model system variations: Results from op/op mice (M-CSF deficient) may differ from those using CSFR1 conditional gene deletion or antibody-based inhibition approaches . Differences between murine and human systems add further complexity requiring careful cross-species validation.

Resolving conflicting data requires systematic experimental design with appropriate controls and careful documentation of all variables that might influence M-CSF activity.

What are common technical pitfalls when working with human M-CSF in experimental settings?

Researchers working with human M-CSF should be aware of several technical challenges:

  • Sample stability issues: M-CSF is sensitive to degradation during freeze/thaw cycles. Some analytes may show decreased detectability after the first round of thawing . Implement single-use aliquoting strategies to minimize this issue.

  • Matrix interference effects: In complex biological samples, matrix components can interfere with M-CSF detection. Perform spike and recovery studies to verify assay performance in each specific sample type .

  • Sample processing artifacts: For plasma samples, additional clotting can occur after thawing, particularly in heparin-anticoagulated specimens. Remove clotted material by centrifugation before analysis to prevent inaccurate measurements .

  • Detection antibody competition: In some sandwich immunoassay formats using polyclonal detection antibodies, the detection antibody may include antibodies that compete with immobilized capture antibody . Evaluate whether sequential or simultaneous incubation protocols are optimal for your specific assay.

  • Calibration range limitations: Standard calibration curves typically range from 0-10,000 pg/mL . Samples with extremely high M-CSF concentrations may require additional dilution to fall within the validated range of detection.

  • Recombinant protein variability: Different sources of recombinant human M-CSF (e.g., E.coli-expressed vs. mammalian-expressed) may have different specific activities and glycosylation patterns that affect biological function .

Addressing these technical challenges through careful methodological design is essential for generating reliable and reproducible data on M-CSF biology.

How can researchers differentiate between direct effects of M-CSF and its indirect effects via macrophage modulation?

Differentiating direct versus indirect effects of M-CSF requires sophisticated experimental approaches:

  • Conditional knockout models: CSFR1 conditional gene deletion mice allow tissue-specific or temporal control of M-CSF signaling . By selectively ablating the receptor in specific cell types, researchers can distinguish direct effects on target cells from indirect effects mediated by macrophages.

  • Co-culture systems: Transwell or conditioned media experiments can separate direct M-CSF signaling from paracrine effects mediated by M-CSF-stimulated macrophages. Compare outcomes from direct M-CSF treatment versus exposure to secreted factors from M-CSF-treated macrophages.

  • Temporal analysis: Implement time-course experiments to distinguish immediate direct effects (occurring within minutes to hours of M-CSF exposure) from delayed indirect effects mediated by secondary cytokine production or cellular recruitment.

  • Pathway inhibitor studies: Selective inhibition of downstream M-CSF signaling components can help identify which cellular responses are directly coupled to receptor activation versus those requiring intermediate steps.

  • Single-cell analysis: Techniques like scRNA-seq allow simultaneous evaluation of multiple cell populations, enabling researchers to map the cascade of cellular interactions following M-CSF signaling . This approach can identify cell-type-specific responses and characterize the sequence of events following M-CSF exposure.

By combining these methodological approaches, researchers can develop a more nuanced understanding of how M-CSF orchestrates complex cellular responses in diverse physiological and pathological contexts.

Product Science Overview

Structure and Function

M-CSF is a four α-helical bundle cytokine . It is involved in diverse biological processes, including the regulation of inflammatory responses, bone resorption, atherosclerosis, and brain and placental development . The primary function of M-CSF is to regulate the proliferation, differentiation, and survival of monocytes and macrophages . Additionally, M-CSF plays a significant role in immunological defense, bone metabolism, fertility, and pregnancy .

Production and Recombinant Form

Recombinant human M-CSF is produced using various expression systems, including E. coli and human 293 cells . The recombinant form is used extensively in research to study the differentiation of macrophages from peripheral blood monocytes and the differentiation of osteoclasts from CD14+ monocytes . The recombinant protein is also used in survival studies and apoptosis assays .

Applications

Human recombinant M-CSF has several applications in scientific research, including:

  • Survival studies and apoptosis assays: For example, using peripheral blood monocytes .
  • Differentiation studies: Differentiation of macrophages from peripheral blood monocytes and differentiation of osteoclasts from CD14+ monocytes .
  • Functional assays: Used in various cell culture and functional assays to study the biological activity of M-CSF .
Biological Activity

The biological activity of recombinant human M-CSF is determined by its ability to regulate the proliferation of NFS-60 cells . The activity is measured in International Units (IU) per milligram, with research-grade M-CSF having an activity of ≥ 1×10^7 IU/mg and premium-grade M-CSF having an activity of ≥ 2×10^7 IU/mg .

Storage and Stability

Recombinant human M-CSF is typically stored in a lyophilized state at -20°C for long-term stability . Once reconstituted, it is stable at 4°C for up to two months and at -20°C for up to six months . It is essential to avoid repeated freeze-thaw cycles to maintain the protein’s biological activity .

Quick Inquiry

Personal Email Detected
Please use an institutional or corporate email address for inquiries. Personal email accounts ( such as Gmail, Yahoo, and Outlook) are not accepted. *
© Copyright 2024 Thebiotek. All Rights Reserved.