Recombinant Human Granulocyte colony-stimulating factor protein (CSF3), partial (Active)

Shipped with Ice Packs
In Stock

Description

Biological Activity and Mechanisms

rhG-CSF binds to its transmembrane receptor (CSF3R), activating JAK/STAT, MAPK, and PI3K/Akt pathways . Key effects include:

  • Neutrophil Production: Stimulates proliferation and differentiation of myeloid progenitors .

  • Stem Cell Mobilization: Enhances hematopoietic stem cell release into peripheral blood .

  • Anti-Apoptotic Effects: Upregulates BCL2 expression in blastocysts, reducing apoptosis .

In vitro studies demonstrate a dose-dependent increase in neutrophil counts, with optimal activity at 10–100 ng/mL .

Therapeutic Uses

  • Chemotherapy-Induced Neutropenia: Reduces infection risk by accelerating neutrophil recovery .

  • Stem Cell Transplantation: Mobilizes CD34+ hematopoietic stem cells for collection .

Experimental Applications

  • Embryonic Development: Enhances blastocyst formation rates in porcine IVF/SCNT models (33% increase at 10 ng/mL) .

  • Neuroprotection: Investigated for mitigating neuronal apoptosis in cerebral ischemia .

Comparative Analysis of Recombinant Variants

ParameterFilgrastim (E. coli)Lenograstim (CHO cells)
Amino Acids175 (non-glycosylated)174 (glycosylated)
Half-Life3.5–3.8 hours4–5 hours
BioactivityEquivalentEquivalent

PEGylated variants (e.g., pegfilgrastim) extend half-life to 15–80 hours .

Research Findings

  • In Vitro Models: rhG-CSF increases POU5F1 (pluripotency) and PCNA (proliferation) gene expression in blastocysts .

  • Preclinical Studies: Reduces amyloid-beta burden in Alzheimer’s mouse models .

  • Clinical Trials: Phase IIb trials ongoing for stroke neuroprotection .

Product Specs

Buffer
Lyophilized from a 0.2 µm filtered 10mM sodium acetate buffer, containing 5% trehalose, pH 4.0
Form
Lyophilized powder
Lead Time
5-10 business days
Notes
Repeated freezing and thawing is not recommended. Store working aliquots at 4°C for up to one week.
Reconstitution
We recommend that this vial be briefly centrifuged prior to opening to bring the contents to the bottom. Please reconstitute protein in deionized sterile water to a concentration of 0.1-1.0 mg/mL. We recommend adding 5-50% of glycerol (final concentration) and aliquoting for long-term storage at -20°C/-80°C. Our default final concentration of glycerol is 50%. Customers may use this as a reference.
Shelf Life
The shelf life is influenced by several factors, including storage conditions, buffer ingredients, storage temperature, and the inherent stability of the protein. Generally, the shelf life of the liquid form is 6 months at -20°C/-80°C. The shelf life of the lyophilized form is 12 months at -20°C/-80°C.
Storage Condition
Store at -20°C/-80°C upon receipt. Aliquoting is necessary for multiple uses. Avoid repeated freeze-thaw cycles.
Tag Info
Tag-Free
Synonyms
C17orf33; Colony stimulating factor 3 (granulocyte); Colony stimulating factor 3; CSF 3; CSF beta; CSF3; CSF3_HUMAN; CSF3OS; Csfg; Filgrastim; G-CSF; GCSA; GCSF; Granulocyte colony stimulating factor; Granulocyte colony-stimulating factor; Lenograstim; Macrophage granulocyte inducer 2; MGC45931; MGI 2; Pluripoietin
Datasheet & Coa
Please contact us to get it.
Expression Region
31-204aa
Mol. Weight
18.7 kDa
Protein Length
Partial of Isoform Short
Purity
>98% as determined by SDS-PAGE.
Research Area
Immunology
Source
E.coli
Species
Homo sapiens (Human)
Target Names
Uniprot No.

Target Background

Function
Granulocyte/macrophage colony-stimulating factors are cytokines that play a crucial role in hematopoiesis. They regulate the production, differentiation, and function of two related white blood cell populations: granulocytes and monocytes-macrophages. This specific CSF induces the production of granulocytes.
Gene References Into Functions
  1. G-CSF and IL-6 levels may serve as predictive indicators of the onset of neonatal Leukemoid reaction. PMID: 29459579
  2. This research describes the construction and characterization of three G-CSF dimeric proteins generated using different linker peptides. GCSF-Lalpha exhibited the most favorable performance in terms of purity and in vitro activity. PMID: 28721592
  3. This study demonstrates that cortisol inhibits CSF3 expression through DNA methylation and suppresses invasion in first-trimester trophoblast cells. PMID: 28846166
  4. This research proposes that in aggressive pancreatic ductal adenocarcinoma, elevated G-CSF contributes to tumor progression by promoting an increase in infiltration of neutrophil-like cells with high immunosuppressive activity. This mechanism provides a potential avenue for a neoadjuvant therapeutic approach to treat this challenging disease. PMID: 28775207
  5. In conclusion, this study identified an elevated lipolysis condition within the follicular fluid (FF) of Polycystic Ovary Syndrome (PCOS) patients with metabolic syndrome (MS). The TNF-alpha and G-CSF levels in FF were correlated with the percentage of top-quality embryos. PMID: 28082237
  6. This research identified a set of IL-17A-regulated genes in keratinocytes that recapitulate typical psoriasis genes exemplified by DEFB4A, S100A7, IL19, and CSF3. These genes were identified based on the differences in the expression profiles of cells stimulated with six cytokines versus cells stimulated with only five cytokines lacking IL-17A. PMID: 26944069
  7. The results indicated that the addition of recombinant IL-1 significantly increased G-CSF expression in fibroblasts. However, IL-1 receptor antagonist only partially abrogated KCM-stimulated G-CSF expression, suggesting the involvement of additional keratinocyte-releasable factors. PMID: 27340768
  8. The reviewed data establish that any damage to brain tissue tends to cause an increase in G-CSF and/or GM-CSF (G(M)-CSF) synthesized by the brain. Glioblastoma cells themselves also synthesize G(M)-CSF. G(M)-CSF synthesized by the brain due to damage by a growing tumor and by the tumor itself stimulates bone marrow to shift hematopoiesis towards granulocytic lineages away from lymphocytic lineages. PMID: 28459367
  9. CD146/MCAM is identified as the functional galectin-3-binding ligand on endothelial cell surfaces responsible for galectin-3-induced secretion of metastasis-promoting cytokines. PMID: 28364041
  10. These findings suggest that G-CSF could decrease the Th1/Th2 ratio in the context of immune thrombocytopenic purpura, providing insights into the direct and indirect immunomodulatory mechanisms underlying G-CSF functions in Th1/Th2 cells. PMID: 27815970
  11. GCSF impairs CD8(+) T cell functionality by interfering with central activation elements. PMID: 26990855
  12. High expression of G-CSF is associated with Tongue squamous cell carcinoma. PMID: 27316348
  13. This study demonstrates that haplotypes consisting of single nucleotide polymorphisms within the PSMD3, CSF3, and MED24 genes are associated with asthma in Slovenian patients. PMID: 27163155
  14. Cancer cells can produce IL-18, which is involved in the processes of angiogenesis, invasion, and metastasis. A decrease in the production of IL-6 and GCSF by peripheral blood cells could serve as an indicator of malignant progression in invasive ductal breast carcinoma. PMID: 27021370
  15. G-CSF showed favorable effects only on the migration of HUVECs, and no direct influence was observed on osteoblasts. PMID: 27006951
  16. Tumor G-CSF expression is an indicator of an extremely poor prognosis in cervical cancer patients treated with chemotherapy. PMID: 26666576
  17. G-CSF stimulates beta1 integrin expression and Swan 71 cell migration by activating PI3K and MAPK signaling pathways. PMID: 26992288
  18. These data suggest that G-CSF may contribute to tumor growth and decrease the antitumor effect of radiotherapy, potentially by promoting vascularization in cancer lesions. PMID: 25976379
  19. In vitro chemotaxis assays and an in vivo transplantation model for chemoattraction confirmed that UCX((R)) are chemotactic to CD34(-)/CD45(-) BM-MSCs through a cell-specific mobilization mechanism mediated by G-CSF. PMID: 24480602
  20. Case Report: undifferentiated colon carcinoma producing G-CSF. PMID: 25400792
  21. G-CSF increases MMP-2 activity and VEGF secretion in trophoblasts through activation of PI3K/Akt and Erk signaling pathways. PMID: 25249155
  22. Elevated IL-8 and G-CSF may be involved in the pathophysiology of narcolepsy. PMID: 24994458
  23. GM-CSF, through its stimulatory function on macrophages, may promote aneurysm progression. PMID: 25389911
  24. G-CSF restricts cancer growth and progression by supporting the survival of sympathetic nerve fibers in 6-hydroxydopamine-sympathectomized mice. PMID: 24975135
  25. Transgenic poultry with a gene of human granulocyte colony-stimulating factor (gcsf) was developed through artificial insemination with the transfected sperm. PMID: 25510103
  26. Data suggest that the long-term protein secondary structural stability/unfolding of GCSF can be modeled from short-term physicochemical phenomena assessed through spectroscopic measurements. PMID: 24421157
  27. SCF+G-CSF treatment in chronic stroke remodels neural circuits in the aged brain. PMID: 23750212
  28. The results demonstrated that 3DHSA-G-CSF has the ability to increase the peripheral white blood cell (WBC) counts of neutropenia model mice, and the half-life of 3DHSA-G-CSF is longer than that of native G-CSF. PMID: 24151579
  29. HNF1A gene was associated with C-reactive protein, and the region including PSMD3 and CSF3 genes was associated with white blood cell count. PMID: 22788528
  30. Administration of G-CSF in a dosage regimen commonly used for bone marrow donors is well tolerated and safe, and provides a signal of positive change in a task of cognitive performance in 8 patients with mild to moderate stage Alzheimer's disease. PMID: 22751169
  31. Activation of the RAS/MEK/ERK pathway regulates G-CSF expression through the Ets transcription factor. PMID: 23530240
  32. These data suggest that GCSF, which is elevated in patient serum, may play a significant role in exacerbating disease in ANCA vasculitis. PMID: 23087180
  33. CEACAM1 inhibits both G-CSF production by myeloid cells and G-CSF-stimulated tumor angiogenesis. PMID: 23319418
  34. G-CSF level in sera of patients with advanced stages of breast cancer was elevated compared to early stages. PMID: 23244154
  35. G-CSF and VEGF levels in sera might be associated with an early phase of brain protection after birth in severe asphyxia treated with head cooling. PMID: 22944463
  36. Granulocyte-colony stimulating factor contributes to glioma progression that may be linked to glioma genesis and recurrence. PMID: 22313638
  37. Plasma G-CSF is elevated after injury and is greater in patients with shock. The rise in G-CSF is associated with prolonged mobilization of hematopoietic progenitor cells. PMID: 23063381
  38. Treatment of 6-week-old bone marrow stromal cells with GCSF significantly improves their proliferation activity and growth factor production and recovers therapeutic effects in the injured brain. PMID: 21981141
  39. Alteration of Dickkopf-1 and receptor activator of nuclear factor-kappaB ligand during PBSC mobilization in healthy donors by G-CSF. PMID: 22120987
  40. These findings suggest that IL-1beta, IL-1Ra, and granulocyte colony-stimulating factor are functional markers of EV71-related cardiac dysfunction. PMID: 22829643
  41. G-CSF administration at 10 mug/kg/day is safe for patients with worsening symptoms of compression myelopathy and may be effective for their neurological improvement. PMID: 21935680
  42. Non-small cell lung cancer specimens with G-CSF expression exhibited poor differentiation, remarkable atypia, prominent necrosis, and infiltration of tumor mass by neutrophils or emperipolesis. PMID: 22336152
  43. Decreased soluble TGF-beta1, Tie-2, and angiopoietins serum levels in bone marrow after treating healthy donors with granulocyte colony-stimulating factor. PMID: 22465760
  44. Elevated plasma GCSF concentration was positively correlated with the severity of ischemic stroke. PMID: 22440005
  45. Induction of Bv8 expression by granulocyte colony-stimulating factor in CD11b+Gr1+ cells: key role of Stat3 signaling. PMID: 22528488
  46. G-CSF can ameliorate cardiac diastolic dysfunction and morphological damage, particularly fibrosis of the myocardium, in Otsuka Long-Evans Tokushima fatty rats with diabetic cardiomyopathy. PMID: 21999467
  47. Human recombinant G-CSF enhances angiogenesis following indirect bypass surgery, a combined therapy that is a safe and convenient treatment method. PMID: 21273924
  48. Case Report: diagnosis of G-CSF-producing ascending colon cancer. PMID: 22443081
  49. Prdx4 inhibits G-CSF-induced signaling and proliferation in myeloid progenitors. PMID: 22045733
  50. Data indicate that serum G-CSF levels were lower in JAK2 V617F-positive versus negative erythrocytosis. PMID: 21645282

Show More

Hide All

Database Links

HGNC: 2438

OMIM: 138970

KEGG: hsa:1440

STRING: 9606.ENSP00000225474

UniGene: Hs.2233

Protein Families
IL-6 superfamily
Subcellular Location
Secreted.

Q&A

What is Recombinant Human Granulocyte Colony-Stimulating Factor (CSF3) and how does it function?

Recombinant Human Granulocyte Colony-Stimulating Factor (CSF3) is a hematopoietic cytokine that primarily stimulates neutrophil production and hematopoietic stem cell mobilization. The protein functions by binding to its cognate receptor (G-CSFR) on target cells and initiating receptor dimerization, which activates downstream signaling pathways. This interaction triggers various cellular processes involved in proliferation, differentiation, and functional activation of neutrophil precursors and mature neutrophils. CSF3 is structurally distinct from GM-CSF (Granulocyte-Macrophage Colony-Stimulating Factor), which has broader effects on multiple myeloid lineages. The biological effects of CSF3 are mediated via binding to receptors expressed specifically on target cells including myeloid progenitors and mature neutrophils .

What are the key structural domains of CSF3 and their significance in receptor interaction?

CSF3 contains several functional domains that are critical for its biological activity. The protein comprises extracellular, transmembrane, and intracellular domains, each serving specific functions in receptor-ligand interactions and signal transduction. The extracellular domain contains the ligand-binding cytokine receptor homology (CRH) domain, which includes conserved motifs such as the di-proline hinge that is essential for maintaining the correct architecture of the ligand/receptor complex. Mutations in this region, such as the P229H mutation, can disrupt receptor function by altering the receptor architecture and limiting G-CSF binding and dimerization. The transmembrane domain is critical for receptor stability within the cell membrane, while the intracellular domain contains multiple regions involved in signal transduction, including binding sites for JAK kinases and other signaling molecules .

How do CSF3R mutations affect neutrophil production and related pathologies?

CSF3R mutations lead to a spectrum of myeloid disorders with seemingly contradictory phenotypes. Mutations can be classified into three main categories: extracellular domain mutations, transmembrane proximal mutations, and intracellular truncation mutations. Extracellular domain mutations, commonly found in Severe Congenital Neutropenia (SCN) patients, often act in a dominant-negative manner by disrupting ligand binding and receptor architecture, leading to decreased neutrophil production. Conversely, transmembrane and intracellular truncation mutations can cause hyperresponsiveness to CSF3, resulting in excessive neutrophil production as seen in Chronic Neutrophilic Leukemia (CNL) and atypical Chronic Myeloid Leukemia (aCML). The paradoxical manifestations may result from disruptions in negative feedback mechanisms, altered receptor recycling or degradation, and changes in the activation of downstream signaling pathways such as JAK/STAT .

What are the optimal experimental systems for studying CSF3 signaling mechanisms?

The optimal experimental systems for studying CSF3 signaling mechanisms include both in vitro and in vivo models that recapitulate different aspects of CSF3 biology. For in vitro studies, researchers commonly use primary myeloid progenitor cells, neutrophil precursors, and cell lines expressing CSF3R. These systems allow investigation of direct cellular responses to CSF3 stimulation, including proliferation, differentiation, and functional activation. For signaling studies, researchers should consider monitoring the activation of JAK/STAT pathways, which are central to CSF3 signal transduction. In vivo models, including genetically modified mice with CSF3R mutations or knockouts, provide valuable insights into the physiological roles of CSF3 in hematopoiesis and immune function. When designing experiments, researchers should carefully consider the specific aspects of CSF3 biology they aim to investigate and select appropriate experimental systems accordingly .

How can researchers effectively analyze contradictions in CSF3-related research data?

Researchers encountering contradictory findings in CSF3-related research should employ a systematic approach to resolve these discrepancies. Contradictions often stem from context-dependent differences, such as cell-type specificity, experimental conditions, or the presence of confounding factors. When analyzing contradictory data, researchers should first examine the specific experimental contexts, including cell types, species, concentrations of CSF3, and time points of analysis. Additionally, apparent contradictions may result from incomplete information about regulatory mechanisms, such as the activities of specific phosphatases (e.g., SHIPs, SHPs) or processes affecting receptor recycling or degradation. In literature-derived knowledge graphs, contradictions have been estimated at a rate of 2.6%, with most apparent contradictions resulting from missing contextual information rather than true scientific contradictions .

What techniques are recommended for measuring CSF3-induced signaling events?

For measuring CSF3-induced signaling events, researchers should employ a combination of techniques that capture both immediate signaling events and downstream biological responses. To monitor receptor activation and early signaling, phospho-specific western blotting or phospho-flow cytometry can detect the activation of JAK kinases and STAT transcription factors. For more comprehensive analysis, phosphoproteomics can identify the full spectrum of phosphorylation events triggered by CSF3 stimulation. Reporter gene assays using STAT-responsive elements can measure transcriptional activation downstream of CSF3 signaling. For long-term effects, proliferation assays (e.g., tritiated thymidine incorporation, Ki-67 staining), differentiation assays (morphological assessment, surface marker analysis), and functional assays (e.g., respiratory burst, phagocytosis) are valuable. Researchers should also consider the temporal dynamics of signaling, as CSF3 induces both immediate and delayed responses through distinct signaling mechanisms .

What molecular mechanisms explain the divergent phenotypes in CSF3R-related disorders?

The divergent phenotypes observed in CSF3R-related disorders—ranging from severe neutropenia in SCN to neutrophilia in CNL—remain incompletely understood, but several molecular mechanisms have been proposed. One possibility involves disruption of negative feedback mechanisms in G-CSFR signaling. In normal conditions, G-CSFR signaling is tightly regulated by negative regulators including SOCS proteins, phosphatases (SHPs, SHIPs), and receptor internalization/degradation processes. Different mutations might differentially affect these regulatory mechanisms. Extracellular domain mutations typically impair ligand binding and receptor activation, leading to reduced neutrophil production. In contrast, transmembrane proximal mutations or intracellular truncation mutations may disrupt negative regulation, resulting in hyperactive signaling and excessive neutrophil production. The specific connection between SCN and CNL/aCML remains unresolved, but possible explanations include disruption of negative feedback loops through loss of phosphatase activity or alterations in receptor recycling/degradation mechanisms that normally control signal duration and intensity .

What considerations should researchers account for when designing clinical studies involving CSF3?

When designing clinical studies involving CSF3, researchers should consider several important factors to ensure robust and clinically relevant outcomes. First, patient selection criteria should account for the specific disease context, as responses to CSF3 vary significantly across different neutropenic conditions. Dosing regimens should be carefully established, as both underdosing and overdosing can lead to suboptimal outcomes or adverse effects. Monitoring parameters should include not only neutrophil counts but also functional assessments of neutrophil activity and long-term outcomes such as infection rates. Additionally, genetic screening for CSF3R mutations before and during treatment is critical, as certain mutations may predict treatment response or risk of leukemic transformation. Researchers should also implement long-term follow-up protocols, as complications such as leukemic progression may develop years after initiation of CSF3 therapy. Finally, studies should incorporate mechanistic investigations to better understand the molecular basis of treatment responses and complications, potentially identifying biomarkers for personalized treatment approaches .

What are the challenges in analyzing context-dependent effects of CSF3 signaling?

Analyzing the context-dependent effects of CSF3 signaling presents several methodological challenges that researchers must address. One primary challenge is the integration of diverse contextual factors that influence CSF3 signaling outcomes, including cell type, tissue microenvironment, concurrent signaling inputs, and temporal dynamics. Extraction of contextual information from the literature requires sophisticated natural language processing techniques capable of identifying relevant experimental conditions and biological contexts. When apparent contradictions arise in CSF3 research findings, they often reflect missing contextual information rather than true biological contradictions. For instance, a study estimated that 2.6% of apparent contradictions in a PubMed-scale knowledge graph required qualification by information such as population group, species, or dosage. Additionally, the evolution of scientific knowledge poses challenges, as early strong associations between CSF3 signaling and biological outcomes may be refined or contradicted by subsequent studies. Researchers should develop methodologies that explicitly account for these contextual dependencies and data evolution to generate more accurate and nuanced models of CSF3 signaling .

How do mutations in different domains of CSF3R differentially affect signaling pathways?

Mutations in different domains of CSF3R have distinct effects on signaling pathways, leading to diverse pathophysiological outcomes. This differential impact can be systematically analyzed through domain-specific functional studies. The table below summarizes the effects of mutations in different receptor domains on signaling pathways and associated clinical phenotypes:

DomainExample MutationsEffect on SignalingAssociated DisordersMechanistic Impact
ExtracellularP229HReduced ligand binding, impaired receptor dimerizationSCN, CINDisrupts CRH domain architecture, acts in dominant-negative manner
TransmembraneT618I, T640NConstitutive receptor activationCNL, aCMLLigand-independent activation of JAK/STAT pathways
IntracellularTruncation mutationsLoss of negative regulatory elementsSCN with risk of AML/MDSProlonged STAT activation, impaired receptor internalization

Extracellular domain mutations typically disrupt the ligand-binding capacity of the receptor, resulting in reduced signaling and neutropenia. Transmembrane domain mutations often induce ligand-independent activation of the receptor, leading to constitutive signaling and neutrophilia. Intracellular domain mutations, particularly truncations, frequently remove negative regulatory elements, resulting in prolonged signal duration after ligand stimulation. These domain-specific effects highlight the importance of precise molecular characterization of CSF3R mutations for understanding disease mechanisms and developing targeted therapeutic approaches .

What are emerging approaches for targeting CSF3-dependent malignancies?

Emerging approaches for targeting CSF3-dependent malignancies focus on exploiting the molecular mechanisms underlying aberrant CSF3 signaling. As research has revealed the specific roles of CSF3R mutations in diseases like CNL and SCN with progression to AML, targeted therapeutic strategies have been developed. These include inhibitors targeting specific components of the CSF3 signaling cascade, particularly JAK kinases, which are often hyperactivated in CSF3R-mutated malignancies. Future approaches may include combination therapies that simultaneously target multiple components of the signaling pathway to prevent resistance development. Additionally, emerging technologies such as CRISPR-Cas9 gene editing hold promise for correcting specific CSF3R mutations in hematopoietic stem cells, potentially offering curative options for genetic forms of neutropenia or CSF3R-driven malignancies. Researchers are also exploring immunotherapy approaches that leverage the immunomodulatory effects of CSF3 to enhance anti-tumor immunity. These diverse strategies reflect the growing understanding of CSF3 biology and the potential for translating this knowledge into novel therapeutic interventions .

How can integration of multi-omics data advance our understanding of CSF3 biology?

Integration of multi-omics data represents a powerful approach for gaining comprehensive insights into CSF3 biology. By combining genomics, transcriptomics, proteomics, metabolomics, and functional assays, researchers can develop holistic models of CSF3 signaling networks and their perturbations in disease states. Genomic analysis can identify genetic variants in CSF3R and related genes that influence signaling outcomes. Transcriptomic profiling reveals the gene expression programs activated by CSF3 stimulation in different cell types and conditions. Proteomic and phosphoproteomic approaches can map the signaling cascades triggered by CSF3 receptor activation, including both canonical and non-canonical pathways. Metabolomic analysis can identify metabolic shifts associated with CSF3 stimulation, providing insights into the energetic and biosynthetic demands of neutrophil production and function. Integration of these diverse data types requires sophisticated computational methods, including machine learning approaches that can identify patterns and relationships across multiple molecular layers. This integrated approach promises to resolve apparent contradictions in CSF3 research by revealing context-specific mechanisms and identifying novel therapeutic targets .

What paradoxes in CSF3 research remain unresolved and require further investigation?

Despite significant advances in CSF3 research, several paradoxes remain unresolved and warrant further investigation. One major paradox is the contrasting phenotypes associated with different CSF3R mutations—severe neutropenia in SCN versus neutrophilia in CNL/aCML—despite affecting the same receptor. While various hypotheses have been proposed, including differential effects on negative feedback mechanisms or receptor trafficking, the precise molecular explanations remain incompletely understood. Another paradox involves the dual role of CSF3 therapy in SCN: while it effectively treats neutropenia, it may also contribute to leukemic progression through selection of cells with CSF3R mutations. The mechanisms underlying this transition from therapeutic benefit to malignant transformation require further elucidation. Additionally, contradictions in the literature regarding CSF3 effects on different cell populations may reflect context-dependent actions that need systematic investigation. Resolving these paradoxes will require innovative experimental approaches, including single-cell analyses to capture heterogeneity in cellular responses, longitudinal studies of molecular changes during disease progression, and sophisticated mathematical modeling of signaling dynamics under various conditions .

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.