Glycosylation: Critical for structural integrity and function .
His-tag: Facilitates affinity purification (e.g., Ni-NTA columns) .
Activity: Binds colony-stimulating factor 1 receptor (CSF-1R), stimulating monocyte/macrophage differentiation and survival .
IL-34 is a pleiotropic cytokine with roles in:
Myeloid Cell Regulation: Promotes monocyte survival, macrophage differentiation, and osteoclast development via CSF-1R and alternative receptors like PTP-ζ and syndecan-1 .
Immunomodulation: Enhances pro-inflammatory cytokine production (e.g., TNF-α, IL-6) while inducing regulatory T-cell (Treg) expansion under certain conditions .
Neuroprotection: Maintains microglial populations in the brain and hippocampus, influencing neuroinflammation and neurodegeneration .
IL34 Human, Sf9 reduces macrophage uptake of Aβ42 (a neurotoxic amyloid-β isoform) and downregulates MMP-9, an extracellular Aβ-degrading enzyme .
| Parameter | M-CSF-Treated Macrophages | IL34-Treated Macrophages |
|---|---|---|
| Aβ42 Uptake Efficiency | High | Reduced |
| MMP-9 Expression | High | Decreased |
| Survival Post-Aβ Exposure | Stable | Reduced |
Data adapted from murine bone marrow-derived macrophages (BMMO) .
IL34 exacerbates cardiac remodeling via NF-κB signaling, increasing CCL2-driven macrophage infiltration .
| Effect | IL34 Knockout Mice | Wild-Type Mice |
|---|---|---|
| Infarct Size | Reduced | Increased |
| CCL2 Expression | ↓ (NF-κB-dependent) | ↑ |
| Fibrosis | Attenuated | Severe |
Data from murine models of ischemia-reperfusion injury .
IL34-deficient mice exhibit skeletal hypomineralization and osteoclast accumulation, indicating its role in osteoblast-osteoclast balance .
IL34 signals through:
CSF-1R: Activates ERK1/2 phosphorylation, driving monocyte/macrophage proliferation .
Syndecan-1: Modulates cellular migration and glioma cell function .
| Receptor | Function | Tissue-Specific Role |
|---|---|---|
| CSF-1R | Macrophage differentiation | Bone marrow, spleen, brain |
| PTP-ζ | Microglial maintenance | Hippocampus, cortex |
| Syndecan-1 | Tumor microenvironment modulation | Gliomas, immune cells |
Adapted from structural and functional studies .
Data from murine and human studies .
IL34 Human, Sf9 serves as a tool for studying:
Interleukin-34, IL-34, IL34, C16orf77, Chromosome 16 Open Reading Frame.
Sf9, Insect cells.
ADPNEPLEMW PLTQNEECTV TGFLRDKLQY RSRLQYMKHY FPINYKISVP YEGVFRIANV TRLQRAQVSE RELRYLWVLV SLSATESVQD VLLEGHPSWK YLQEVQTLLL NVQQGLTDVE VSPKVESVLS LLNAPGPNLK LVRPKALLDN CFRVMELLYC SCCKQSSVLN WQDCEVPSPQ SCSPEPSLQY AATQLYPPPP WSPSSPPHST GSVRPVRAQG EGLLPHHHHH H.
IL-34 (Interleukin-34) is a cytokine functionally similar to CSF-1 (Colony Stimulating Factor 1), playing a key role in the development and function of mononuclear lineage cells. Despite sharing the CSF1R receptor with CSF-1, IL-34 has distinct tissue-specific functions, particularly in the development and maintenance of Langerhans cells in the skin and microglia in the brain . IL-34 is a glycosylated polypeptide containing 231 amino acids (residues 21-242) with a molecular mass of approximately 26.3 kDa, though it may appear as 28-40 kDa on SDS-PAGE due to glycosylation . Unlike many cytokines with broader expression patterns, IL-34 demonstrates remarkable tissue selectivity, making it particularly valuable for targeted research applications .
IL-34 expression in the nervous system is regulated by multiple factors including developmental stage, neuronal activity, and neuronal subtype. Research indicates that IL-34 expression increases developmentally, with robust expression observed in regions like the anterior cingulate cortex (ACC) by postnatal day 15 . At the cellular level, IL-34 expression correlates with neuronal activity markers such as Fos, with significantly higher expression in Fos+ (active) compared to Fos- (inactive) neurons . This activity-dependent regulation appears particularly pronounced in glutamatergic (Gad2-) neurons rather than GABAergic (Gad2+) neurons, suggesting cell-type specificity in IL-34 production within the brain .
Recombinant human IL-34 produced in Sf9 insect cells requires specific handling conditions to maintain its biological activity. For short-term use (within 2-4 weeks), store at 4°C. For longer-term storage, maintain at -20°C and avoid repeated freeze-thaw cycles . When preparing for long-term storage, addition of a carrier protein (0.1% HSA or BSA) is recommended to enhance stability . The protein is typically supplied as a sterile filtered colorless solution with concentration of 0.25 mg/ml in a buffer containing 20 mM Tris-HCl (pH 8.5), 1 mM DTT, 0.15M NaCl, 0.1 mM PMSF, 1 mM EDTA, and 30% glycerol . For shipping and transportation, the protein should be maintained with ice packs to preserve stability and activity .
To measure IL-34-induced signaling, researchers should focus on detecting the phosphorylation of CSF1R and downstream mediators including ERK1/2 and AKT. Western blotting after stimulation with IL-34 (50-100 ng/mL) for short time periods (5-10 minutes) can effectively demonstrate pathway activation . When comparing IL-34 signaling with CSF-1, parallel stimulation conditions should be established using equivalent concentrations of both cytokines. Microglial cell lines such as N13 provide suitable models for these experiments, as they express functional CSF1R . For quantitative analysis, phosphorylation levels should be normalized to total protein levels and compared to unstimulated controls. Researchers should also consider examining temporal dynamics of signaling by collecting samples at multiple time points to capture both immediate (5-10 min) and sustained signaling responses.
To investigate IL-34's role in microglial development, researchers should consider complementary approaches targeting both IL-34 expression and function. Developmental time-course studies should examine critical periods such as postnatal days 8-15, when IL-34 expression significantly increases in regions like the anterior cingulate cortex . For phenotypic analysis, combine markers of microglial number (Iba1), maturation (TMEM119), and function (CD68 for phagocytic capacity) . Additionally, morphological assessment using 2D Sholl analysis to quantify ramification can provide insights into microglial maturation state .
For functional approaches, IL-34 knockout models or neutralizing antibodies can be employed. When using antibodies, multiple antibody types (monoclonal, polyclonal) with validated neutralizing capacity should be tested, and IC50 values compared (in the range of 0.4-2.0 nM) . When analyzing microglial protein expression in the context of varying cell numbers, implement analytical approaches that correct for cell density differences, such as creating binary masks of microglial markers (Iba1) to properly quantify other protein signals (e.g., TMEM119) on a per-cell basis .
IL-34 demonstrates remarkable tissue selectivity compared to CSF-1, despite both cytokines signaling through CSF1R. When designing experiments to distinguish their effects, researchers should examine multiple tissue compartments simultaneously. In the central nervous system, IL-34 is crucial for microglial development and maintenance, with IL-34 knockout models showing significant reductions in microglial numbers and maturation . Importantly, IL-34 inhibition shows minimal effects on peripheral macrophage populations in healthy mice, in contrast to broader CSF1R inhibition which affects multiple tissue macrophage populations .
To investigate this differential regulation experimentally, researchers should design studies that compare forebrain regions (IL-34 dependent) with brainstem and cerebellum (CSF-1 dependent) . Analysis should include quantification of microglial densities, morphological assessment, and expression of maturation markers like TMEM119. RNA-sequencing of isolated microglia from different brain regions of wild-type and IL-34 knockout mice can further elucidate region-specific transcriptional programs regulated by IL-34 versus CSF-1 . For functional studies, selective IL-34 neutralization can be compared with CSF1R inhibition to distinguish tissue-specific versus global effects on myeloid populations .
IL-34 plays a significant role in microglial proliferation, a hallmark of many neurodegenerative conditions including Alzheimer's disease and prion disease . For researchers investigating therapeutic applications, selective IL-34 inhibition offers advantages over broader CSF1R inhibition strategies. In the ME7 prion disease model, IL-34 neutralization reduces microglial proliferation while avoiding systemic effects on peripheral macrophage populations .
Methodologically, researchers should consider comparing IL-34 inhibition with CSF1R inhibition in terms of microglial numbers, morphology, and inflammatory profiles. Flow cytometry and immunohistochemistry approaches should quantify not only microglial density but also activation states using markers for homeostatic (TMEM119, P2RY12) versus reactive phenotypes (CD68, MHC-II) . For functional assessment, researchers should evaluate cognitive performance, synaptic function, and neuropathological hallmarks specific to the disease model. Importantly, the timing of intervention should be carefully considered, as IL-34's effects on microglia appear to be developmentally regulated, with intervention after key developmental windows potentially offering more selective effects .
IL-34 has been implicated in autoimmune conditions, with elevated serum levels observed in patients with systemic lupus erythematosus (SLE) that correlate with disease severity . When designing studies to investigate IL-34's role in autoimmunity, researchers should consider several methodological approaches.
For clinical samples, paired analysis of serum IL-34 levels with disease activity scores provides correlative evidence, but mechanistic studies require more sophisticated approaches . In experimental systems, researchers should examine how IL-34 influences regulatory T cell subsets, particularly Foxp3+CD8+CD45RClo Tregs which have been implicated in transplant models . The impact of IL-34 on macrophage migration into affected tissues should be assessed using adoptive transfer approaches with labeled cells and intravital imaging .
In autoimmune disease models, researchers should evaluate how IL-34 overexpression or inhibition affects the production of autoantibodies, autoreactive T cell responses, and tissue inflammation . Importantly, measurement of IL-34 in both circulation and affected tissues is necessary, as local production may have different effects than systemic levels. Flow cytometric analysis of immune cell populations in multiple compartments (circulation, lymphoid organs, affected tissues) should be conducted to comprehensively evaluate IL-34's immunomodulatory effects.
Recombinant IL-34 can be produced in various expression systems, including Sf9 insect cells as described in the search results . When comparing IL-34 from different sources, researchers should consider several technical aspects that may influence experimental outcomes.
Post-translational modifications, particularly glycosylation patterns, differ significantly between expression systems. Human IL-34 produced in Sf9 cells has a molecular weight of approximately 26.3 kDa but appears as 28-40 kDa on SDS-PAGE due to insect cell-specific glycosylation . These differences can affect protein stability, receptor binding affinity, and biological activity. Researchers should therefore validate activity using functional assays such as CSF1R phosphorylation or cellular proliferation before comparing results across studies using different protein sources.
Additionally, tags used for purification (such as the His-tag described in the recombinant IL-34 ) may influence protein folding or receptor interaction. Control experiments comparing tagged versus untagged proteins or proteins with different tag positions (N-terminal versus C-terminal) are advisable for critical experiments. Purification methods also influence contaminant profiles and protein conformational states, necessitating careful quality control through analytical techniques like SEC-MALS or dynamic light scattering to assess homogeneity prior to biological testing.
Distinguishing between IL-34 and CSF-1 mediated effects presents a significant challenge since both cytokines signal through the same receptor (CSF1R). Researchers can employ several complementary approaches to address this challenge.
Selective neutralization using validated antibodies against either IL-34 or CSF-1 allows for pathway-specific inhibition . When using neutralizing antibodies, validation of their specificity and efficacy is crucial - they should block the target cytokine without cross-reactivity . Genetic approaches using conditional knockout models for either cytokine offer more definitive distinction but may have developmental confounders.
Spatiotemporal analysis provides another approach, as IL-34 and CSF-1 have distinct expression patterns in the brain and other tissues. For instance, microglia in forebrain regions depend more heavily on IL-34, while those in brainstem and cerebellum are maintained by CSF-1 . Analyzing region-specific responses can therefore help distinguish the cytokines' respective contributions.
For signaling studies, while both cytokines activate CSF1R, they may induce quantitative or qualitative differences in downstream pathway activation. Detailed phosphoproteomic analysis or kinetic studies of ERK1/2 and AKT activation may reveal subtle differences in signaling signatures . Combining these approaches with appropriate controls and validation experiments will provide more robust differentiation between IL-34 and CSF-1 mediated effects.
Neuron-microglia communication represents a frontier in neuroimmunology research, with IL-34 emerging as a key mediator of this interaction. The finding that neuronal activity regulates IL-34 expression suggests a mechanism by which neurons signal to microglia. Researchers interested in this field should consider several cutting-edge approaches.
Co-culture systems combining primary neurons and microglia offer controlled environments to study direct communication. For activity-dependent IL-34 production, researchers can employ optogenetic stimulation of neurons followed by multiplex cytokine analysis or proximity ligation assays to detect IL-34 secretion and receptor binding. Single-cell RNA sequencing of neuron-microglia co-cultures under various stimulation conditions can reveal cell type-specific responses and signaling cascades.
For in vivo studies, conditional IL-34 knockout in specific neuronal populations (using CRE-driver lines for glutamatergic versus GABAergic neurons) would help delineate cell type-specific contributions . Combined with two-photon imaging of fluorescently labeled microglia, researchers can assess real-time microglial responses to neuronal activity in the presence or absence of IL-34 signaling. Spatial transcriptomics approaches can map IL-34 expression patterns alongside microglial distribution and activation states across brain regions during development or disease progression.
The selective role of IL-34 in microglial development and maintenance makes it an attractive target for modulating neuroinflammation without broadly affecting peripheral immune function . Researchers exploring its therapeutic potential should consider several methodological approaches.
Preclinical evaluation should compare IL-34 neutralization with established CSF1R inhibitors across multiple disease models, including Alzheimer's disease, multiple sclerosis, and prion disease . Beyond measuring microglial numbers, comprehensive phenotypic characterization should assess microglial activation states, phagocytic capacity, and inflammatory cytokine production. Single-cell RNA sequencing of microglia from treated animals can reveal shifts in disease-associated microglial signatures toward homeostatic states.
For translational potential, researchers should develop humanized antibodies or small molecule inhibitors with favorable CNS penetration profiles. Blood-brain barrier penetration can be assessed using in vitro models or in vivo microdialysis. Central versus peripheral effects should be distinguished using methods such as intracerebroventricular delivery versus systemic administration. Importantly, developmental timing must be considered, as IL-34's effects on microglia differ between developmental stages and adulthood . Therefore, therapeutic interventions should be evaluated across different age groups to identify optimal treatment windows.
Interleukin 34 (IL-34) is a cytokine that plays a crucial role in the immune system by acting as a ligand for the colony-stimulating factor-1 receptor (CSF1R). It was first identified in 2008 through comprehensive proteomic analysis and has since been recognized for its significant functions in various biological processes .
IL-34 is a four-helical homodimeric protein that does not belong to any previously identified cytokine family . It is highly conserved across vertebrates, including fish, amphibians, birds, and mammals . The protein is expressed in several tissues, particularly in response to inflammatory stimuli . Notably, IL-34 shares structural homology with macrophage colony-stimulating factor (M-CSF) and signals via the CSF1R .
IL-34 primarily stimulates the proliferation of human peripheral blood monocytes and the formation of macrophage colonies . It promotes the survival of monocytes and the proliferation of macrophages, playing a hematopoietic differentiation role in the bone marrow . Additionally, IL-34 induces the differentiation, adhesion, and proliferation of osteoclasts, contributing to bone homeostasis .
Recombinant IL-34 is produced using various expression systems, including the Sf9 insect cell line. This recombinant protein is optimized for use in cell culture, differentiation studies, and functional assays . It is commonly used in in vitro studies of monocyte, macrophage, and dendritic cell differentiation, as well as in the investigation of CSF1R signaling .
IL-34 has been extensively studied for its role in the development of myeloid compartments, including dendritic cells, monocytes, Langerhans cells, and microglia . Its ability to induce myeloid cell expansion and differentiation makes it a valuable tool in research focused on immune responses and hematopoiesis .