SPI1 Human

Spi-1 Proto-Oncogene Human Recombinant
Shipped with Ice Packs
In Stock

Description

Hematological Malignancies

  • T-cell Acute Lymphoblastic Leukemia (T-ALL): SPI1 fusion proteins (e.g., TCF7-SPI1) drive oncogenic signaling, promoting NRAS and Wnt/β-catenin pathways .

  • Acute Erythroleukemia (AEL): Elevated SPI1 expression blocks erythroid differentiation, leading to leukemia progression .

Alzheimer’s Disease

SPI1 ExpressionEffectReferences
KnockdownIncreases Aβ aggregation, amyloid plaques, and gliosis in mouse models.
OverexpressionReduces insoluble Aβ40/42, plaque burden, and dystrophic neurites.

SPI1 regulates microglial phagocytosis of amyloid-β and modulates neuroinflammatory pathways. Dysregulated SPI1 levels correlate with Alzheimer’s disease risk in genetic studies .

Fibrotic Diseases

SPI1 induces pro-fibrotic fibroblast polarization by upregulating extracellular matrix genes. Its downregulation reverses fibrotic phenotypes, highlighting its therapeutic potential in fibrosis .

5. Research Findings and Implications
Recent studies underscore SPI1’s dual role in disease:

  • Alzheimer’s Disease: SPI1 overexpression in 5XFAD mice reduced Aβ plaque burden by 45–60% and decreased gliosis, suggesting its potential as a therapeutic target .

  • Erythroleukemia: SPI1 binds active enhancers in erythroid progenitors, recruiting HDAC1 to repress differentiation genes. Inhibiting this axis may restore erythroid maturation .

  • Immune Regulation: SPI1 modulates microglial Aβ clearance and immune responses, with implications for neurodegenerative disease therapies .

Product Specs

Introduction
The SPI1 gene encodes an ETS-domain transcription factor known as Spi-1 Proto-Oncogene. This factor plays a crucial role in activating gene expression during the development of myeloid and B lymphoid cells. SPI1 binds to a purine-rich DNA sequence called the PU-box, typically found near the promoters of its target genes, thereby regulating their expression. Additionally, SPI1 can bind to RNA and influence pre-mRNA splicing, including the alternative splicing of specific target genes.
Description
Recombinant human SPI1, expressed in E. coli, is a single polypeptide chain with a molecular weight of 33.6 kDa. It comprises 294 amino acids, with residues 1-271 corresponding to the SPI1 protein. A 23 amino acid His-tag is fused to the N-terminus to facilitate purification, which is achieved through proprietary chromatographic techniques.
Physical Appearance
A clear solution that has been sterilized through filtration.
Formulation
The SPI1 solution is provided at a concentration of 0.25 mg/ml in a buffer consisting of 20 mM Tris-HCl (pH 8.0) and 10% glycerol.
Stability
For short-term storage (up to 2-4 weeks), the SPI1 solution should be kept at 4°C. For extended storage, it is recommended to freeze the solution at -20°C. To ensure optimal stability during long-term storage, adding a carrier protein such as HSA or BSA (0.1%) is advisable. It's important to avoid repeated cycles of freezing and thawing.
Purity
The purity of SPI1 is determined by SDS-PAGE analysis to be greater than 85.0%.
Synonyms
Transcription factor PU.1, 31 kDa-transforming protein, Transcription factor PU.1 isoform 1, SPI1, OF, PU.1, SFPI1, SPI-1, SPI-A.
Source
Escherichia Coli.
Amino Acid Sequence
MGSSHHHHHH SSGLVPRGSH MGSMLQACKM EGFPLVPPQP SEDLVPYDTD LYQRQTHEYY PYLSSDGESH SDHYWDFHPH HVHSEFESFA ENNFTELQSV QPPQLQQLYR HMELEQMHVL DTPMVPPHPS LGHQVSYLPR MCLQYPSLSP AQPSSDEEEG ERQSPPLEVS DGEADGLEPG PGLLPGETGS KKKIRLYQFL LDLLRSGDMK DSIWWVDKDK GTFQFSSKHK EALAHRWGIQ KGNRKKMTYQ KMARALRNYG KTGEVKKVKK KLTYQFSGEV LGRGGLAERR HPPH.

Q&A

What is SPI1 and what are its primary functions in human biology?

SPI1 is a transcription factor belonging to the ETS family that plays pivotal roles in multiple biological processes. It functions primarily in hematopoiesis (blood cell formation), particularly during the endothelial-to-hematopoietic transition (EHT) where it regulates lineage commitment . SPI1 is also involved in cell differentiation, proliferation, and survival in various contexts.

Research has expanded SPI1's known functions beyond traditional hematopoietic roles to include:

  • Regulation of myeloid and lymphoid lineage commitment through distinct molecular pathways

  • Involvement in neurodegenerative diseases, particularly Alzheimer's disease

  • Contribution to oncogenic processes in certain cancers, including gliomas

  • Modulation of cell cycle progression and apoptotic pathways

The protein's expression follows specific temporal patterns during development, with regulatory actions that are highly context-dependent and dosage-sensitive.

How is SPI1 expression accurately measured in laboratory settings?

Multiple complementary techniques allow for precise measurement of SPI1 expression:

Quantitative Real-Time PCR (qRT-PCR):

  • Standard method for measuring SPI1 mRNA levels

  • Typical primers include:

    • Forward: 5′-GCGACCATTACTGGGACTTCC-3′

    • Reverse: 5′-GGGTATCGAGGACGTGCAT-3′

  • GAPDH commonly serves as internal control with primers:

    • Forward: 5′-GACTCATGACCACAGTCCATGC-3′

    • Reverse: 5′-AGAGGCAGGGATGATGTTCTG-3′

RNA Sequencing:

  • Provides comprehensive transcriptome profiling including SPI1

  • Single-cell RNA-seq offers additional resolution of SPI1 expression at cellular level

  • Has been used to map SPI1 expression during human endothelial-to-hematopoietic transition both in vivo and in vitro

Protein Detection Methods:

  • Western blotting for semi-quantitative protein measurement

  • Immunohistochemistry for spatial localization in tissues

  • Flow cytometry for quantification at single-cell level

When analyzing SPI1 expression data, researchers should consider:

  • Cell-type specificity of expression patterns

  • Temporal dynamics during developmental processes

  • Technical variations between different measurement platforms

  • Appropriate normalization strategies for the chosen method

What downstream targets are regulated by SPI1 and how can they be identified?

SPI1 regulates numerous downstream genes through direct binding to their promoter regions. Key targets include:

In hematopoietic development:

  • KLF1 (Krüppel-like factor 1): Directs erythroid/myeloid lineage development

  • LYL1 (Lymphoblastic Leukemia-Associated Hematopoiesis Regulator 1): Guides lymphoid lineage development

In cancer progression:

  • PAICS (Phosphoribosylaminoimidazole Carboxylase): Promotes proliferation and migration of glioma cells when upregulated by SPI1

Methodologies for identifying SPI1 targets include:

Chromatin Immunoprecipitation (ChIP):

  • ChIP followed by qPCR or sequencing (ChIP-seq)

  • Identifies direct binding sites of SPI1 throughout the genome

  • Has confirmed SPI1 binding to promoters of targets like PAICS

Transcriptomic Analysis After SPI1 Modulation:

  • RNA-seq following SPI1 knockdown or overexpression

  • Reveals genes whose expression changes in response to SPI1 levels

  • Helps distinguish direct from indirect targets

Reporter Assays:

  • Dual luciferase assays using promoter regions of potential targets

  • Confirms functional impact of SPI1 binding on transcriptional activity

  • Has demonstrated SPI1's ability to enhance PAICS expression

Rescue Experiments:

  • Overexpression of putative targets in SPI1-knockdown backgrounds

  • Studies show that KLF1 or LYL1 overexpression partially rescues defects caused by SPI1 knockdown

How can researchers effectively modulate SPI1 expression in experimental settings?

Several approaches allow for precise control of SPI1 expression:

RNA Interference:

  • siRNA for transient knockdown:

    • Achieves 70-90% reduction in expression

    • Useful for short-term experiments (3-5 days)

    • Has been employed in glioma studies to assess SPI1's role in cell migration

  • shRNA for stable knockdown:

    • Lentiviral delivery enables long-term suppression

    • Allows selection of stable SPI1-knockdown cell lines

    • Particularly useful for sustained phenotypic studies

Overexpression Systems:

  • Plasmid-based transient overexpression

  • Stable integration for long-term studies

  • Inducible systems (Tet-On/Tet-Off) for temporal control

CRISPR/Cas9 Gene Editing:

  • Complete knockout of SPI1

  • Knockin modifications for structure-function studies

  • CRISPRa/CRISPRi for endogenous expression modulation

Experimental Design Considerations:

  • Cell type-specific effects must be considered

  • Dosage sensitivity is critical, as both under and overexpression affect phenotype

  • Timing of modulation affects outcomes, particularly in developmental contexts

  • Compensatory mechanisms may emerge with long-term modulation

What model systems are most appropriate for studying SPI1 function in different contexts?

Various model systems offer distinct advantages for SPI1 research:

Cell Line Models:

  • Hematopoietic cell lines (K562, HL-60, U937)

  • Glioma cell lines for cancer studies (U87, U251)

  • Advantages: Ease of genetic manipulation, scalability

  • Limitations: May not fully recapitulate primary cell behavior

Primary Cell Models:

  • Isolated human hematopoietic stem/progenitor cells

  • Primary microglia for neuroinflammation studies

  • More physiologically relevant but limited availability

In Vitro Differentiation Systems:

  • Human pluripotent stem cell differentiation:

    • Monolayer-based, chemically-defined systems

    • Recapitulate endothelial-to-hematopoietic transition

    • Enable temporal analysis of SPI1 during development

  • iPSC-derived microglia for neurodegeneration studies

Animal Models:

  • Transgenic mouse models with SPI1 modulation

  • Mouse models of Alzheimer's disease show that increasing SPI1 expression improves disease symptoms, while reducing SPI1 exacerbates pathology

  • Zebrafish models for visualization of hematopoietic development

Selection Considerations:

  • Research question should guide model choice

  • Multi-model approaches often provide complementary insights

  • Consider translational relevance when designing studies

How does SPI1 regulate endothelial-to-hematopoietic transition and lineage commitment?

SPI1 plays a crucial role in the endothelial-to-hematopoietic transition (EHT), a fundamental process in blood system development:

Transcriptional Network Regulation:

  • Functions within a complex network involving multiple transcription factors

  • Coordinates with factors like RUNX1, GATA2, and TAL1

Lineage-Specific Control:

  • The SPI1-KLF1 axis directs erythroid/myeloid development

  • The SPI1-LYL1 axis guides lymphoid lineage development

Expression Dynamics:

  • SPI1 expression increases during transition from hemogenic endothelial cells to hematopoietic progenitors

  • This pattern is conserved between in vivo (human AGM region) and in vitro models

Functional Impact of Disruption:

  • SPI1 knockdown during in vitro EHT results in:

    • Decreased generation of hematopoietic progenitor cells

    • Reduced differentiation potential

    • Cell cycle dysregulation and increased apoptosis

Multi-omic analyses reveal that SPI1 contributes to hematopoietic stem cell heterogeneity during embryonic development, suggesting its role in establishing diverse blood cell fates.

What mechanisms underlie SPI1's role in neurodegenerative diseases like Alzheimer's?

Recent research has identified SPI1 as an important factor in Alzheimer's disease (AD) pathology:

Genetic Association and Expression Effects:

  • SPI1 has been genetically linked to Alzheimer's disease risk

  • The relationship between SPI1 levels and AD pathology shows a bidirectional effect:

    • Reducing SPI1 expression worsens AD symptoms in mouse models

    • Increasing SPI1 expression improves AD-related characteristics

Mechanism Insights:

  • SPI1 modulation affects multiple aspects of AD pathology

  • Effects likely involve microglial function, as SPI1 regulates microglial development

  • Precise SPI1 levels appear crucial for proper function in the CNS context

Therapeutic Implications:

  • Finding that increased SPI1 expression improves AD symptoms suggests potential therapeutic avenues

  • Researchers at Indiana University School of Medicine are working on drug discovery targeting SPI1-related pathways for AD treatment

  • Challenge lies in achieving precisely controlled modulation, as both insufficient and excessive SPI1 function may be detrimental

This research represents a significant expansion of SPI1's biological relevance beyond hematopoiesis to neurodegenerative conditions.

How should contradictory findings about SPI1 function in different contexts be reconciled?

Resolving seemingly contradictory findings about SPI1 requires consideration of several factors:

Context-Dependent Functions:

  • Cell type specificity: SPI1 interacts with different cofactors across cell types

  • Developmental stage effects: SPI1's role evolves during developmental progression

  • Disease context: Different pathological states may alter SPI1's function

Dosage-Dependent Effects:

  • SPI1 exhibits concentration-dependent effects in many systems

  • Both insufficient and excessive SPI1 can be detrimental

  • Different SPI1-regulated processes may have unique threshold requirements

Methodological Considerations:

  • Acute versus chronic modulation yields different results

  • Complete knockout versus partial knockdown reveals different aspects of function

  • Model system variations contribute to apparent contradictions

For example, the contradiction between SPI1's apparent oncogenic role in glioma and its protective role in Alzheimer's disease can be reconciled by recognizing:

  • Different cellular contexts (glial tumor cells versus microglia/neurons)

  • Distinct molecular partners and downstream targets in each system

  • Different cellular processes being regulated

What are the most effective approaches for validating putative SPI1 target genes?

Validating SPI1 targets requires a multi-layered approach:

Bioinformatic Prediction:

  • Identify putative SPI1 binding motifs in gene regulatory regions

  • Analyze existing ChIP-seq datasets across relevant cell types

Experimental Validation:

  • Chromatin immunoprecipitation (ChIP):

    • Confirms direct binding of SPI1 to target gene regulatory regions

    • Has been used to demonstrate SPI1 binding to the PAICS promoter

  • Reporter assays:

    • Luciferase assays with wild-type and mutated binding sites

    • Confirms functional impact of SPI1 binding on transcription

    • Double luciferase activity assays have confirmed SPI1's regulation of PAICS

Functional Validation:

  • Expression analysis after SPI1 modulation:

    • Measure target gene changes following SPI1 knockdown/overexpression

    • Determine dose-dependency and kinetics of the effect

  • Rescue experiments:

    • Restore expression of specific targets in SPI1-deficient background

    • Studies show KLF1 or LYL1 overexpression partially rescues defects caused by SPI1 knockdown

Mechanistic Confirmation:

  • Site-directed mutagenesis to confirm specific binding sites

  • DNA-protein interaction assays (EMSA, microscale thermophoresis)

  • Chromatin conformation studies to assess enhancer-promoter interactions

A comprehensive validation pipeline combines multiple approaches to provide strong evidence for direct regulation by SPI1.

What specialized techniques are necessary for studying SPI1 in specific cellular contexts?

Investigating SPI1's context-specific roles requires tailored methodological approaches:

Cell Type-Specific Manipulation:

  • Conditional expression systems (Cre-loxP technology)

  • Cell type-specific promoters driving SPI1 expression or knockdown

  • FACS-based isolation of specific cell populations for targeted analysis

Temporal Control Strategies:

  • Inducible expression systems for precise timing of SPI1 modulation

  • Developmental time course analyses capturing dynamic changes

  • Particularly important when studying SPI1 during endothelial-to-hematopoietic transition

Single-Cell Analysis Approaches:

  • Single-cell RNA sequencing:

    • Resolves heterogeneity within populations

    • Identifies cell subsets differentially affected by SPI1

    • Has been used to map SPI1 expression during human AGM hematopoiesis

  • Single-cell ATAC-seq for chromatin accessibility changes

  • CyTOF or spectral flow cytometry for multi-parameter phenotyping

Multi-omics Integration:

  • Combined analysis of transcriptome, chromatin accessibility, and SPI1 binding

  • Provides comprehensive view of SPI1's regulatory network

  • Helps distinguish direct from indirect effects

Functional Assays for Phenotypic Assessment:

  • Hematopoietic differentiation: Colony formation assays

  • Cell migration: Transwell and wound healing assays used in glioma studies

  • Cell cycle analysis: Flow cytometry showing G1 phase stagnation in SPI1-knockdown glioma cells

  • Apoptosis: Annexin V staining demonstrating increased apoptosis with SPI1 downregulation

The selection of techniques should be tailored to the specific cellular context and SPI1 functions under investigation.

How might SPI1-targeted therapies be developed for Alzheimer's disease?

Recent findings suggest promising therapeutic potential in targeting SPI1 for Alzheimer's disease:

Current Evidence Base:

  • Research at Indiana University School of Medicine demonstrated that increased SPI1 expression improves Alzheimer's disease characteristics in mouse models

  • The study revealed that reducing SPI1 worsened disease symptoms, while increasing it had beneficial effects

Therapeutic Approaches Under Investigation:

  • Researchers are working with the Target Enablement to Accelerate Therapy Development for Alzheimer's Disease (TREAT-AD) program on potential drug discovery efforts

  • The goal is developing compounds that can precisely modulate SPI1 activity

Methodological Considerations:

  • Dose precision is critical: Both insufficient and excessive SPI1 function may be harmful

  • Cell-type specificity: Targeting SPI1 in specific cell populations (like microglia) while sparing others

  • Delivery systems: Developing approaches to modulate SPI1 within the central nervous system

  • Temporal control: Determining optimal timing for intervention in disease progression

Potential Screening Platforms:

  • High-throughput compound screening against SPI1 or its regulatory pathways

  • iPSC-derived microglia from AD patients for personalized drug testing

  • Mouse models with humanized SPI1 for in vivo validation

Challenges and Considerations:

  • The precise level of SPI1 modulation appears crucial for therapeutic benefit

  • Understanding the complete mechanism of SPI1's protective effects in AD

  • Potential off-target effects on hematopoiesis must be monitored

  • Biomarkers for patient selection and treatment response need development

What approaches can be used to investigate SPI1's role in cancer development and progression?

SPI1 has been implicated in various cancers, particularly gliomas, requiring specialized investigative approaches:

Molecular Mechanism Studies:

  • SPI1 promotes glioma cell proliferation and migration through:

    • Regulation of cell cycle progression (G1 phase control)

    • Modulation of apoptotic pathways

    • Transcriptional regulation of oncogenic targets like PAICS

Experimental Models:

  • Patient-derived glioma cell lines and xenografts

  • Cancer stem cell models to assess SPI1's role in tumor initiation

  • Genetic manipulation in existing glioma models:

    • SPI1 knockdown studies show reduced migration in transwell assays

    • Decreased wound healing capacity with SPI1 downregulation

Clinical Correlation Approaches:

  • Analysis of SPI1 expression in patient tumor samples

  • Correlation with clinical parameters and outcomes

  • Single-cell analysis of tumor heterogeneity and SPI1 expression patterns

Therapeutic Targeting Strategies:

  • Development of SPI1 inhibitors or modulators

  • Identification of druggable downstream targets

  • Combination approaches targeting SPI1-regulated pathways

Molecular Interaction Analyses:

  • Identification of cancer-specific SPI1 cofactors

  • Mapping of SPI1 regulatory networks in malignant versus normal cells

  • Epigenetic regulation of SPI1 in cancer contexts

These approaches provide a comprehensive framework for understanding SPI1's oncogenic mechanisms and potential for therapeutic targeting.

How do changes in SPI1 expression impact hematopoietic differentiation potential?

SPI1 plays a critical role in directing hematopoietic differentiation, with expression changes having profound effects:

Expression Level Effects:

  • Precise SPI1 levels are crucial for normal hematopoiesis

  • SPI1 knockdown during in vitro endothelial-to-hematopoietic transition leads to:

    • Decreased generation of hematopoietic progenitor cells

    • Reduced differentiation potential of these cells

Lineage-Specific Regulation:

  • The SPI1-KLF1 axis directs erythroid and myeloid lineage development

  • The SPI1-LYL1 axis guides lymphoid lineage development

  • This dual regulatory activity contributes to balanced blood cell production

Rescue Experiments Reveal Mechanistic Insights:

  • Overexpression of KLF1 partially rescues myeloid lineage potential in SPI1-knockdown cells

  • LYL1 overexpression re-establishes lymphoid lineage potential

  • These findings demonstrate how SPI1 directs lineage commitment through specific downstream factors

Methodological Approaches for Assessment:

  • Colony-forming unit assays to quantify progenitor populations

  • Flow cytometry analysis of lineage markers

  • In vitro differentiation systems recapitulating developmental processes

  • Transplantation assays for in vivo reconstitution potential

Therapeutic Implications:

  • Understanding SPI1-regulated hematopoiesis informs development of treatments for blood disorders

  • Potential applications in improving hematopoietic stem cell transplantation

  • Opportunities for directed differentiation in regenerative medicine approaches

Product Science Overview

Introduction

The Spi-1 proto-oncogene, also known as SPI1 or PU.1, is a critical transcription factor involved in the regulation of gene expression during the development of myeloid and B-lymphoid cells. This gene plays a pivotal role in the immune system, influencing the differentiation and function of various immune cells.

Gene and Protein Structure

SPI1 encodes a protein that belongs to the ETS-domain transcription factor family. The ETS domain is a conserved DNA-binding domain that allows the protein to bind to specific DNA sequences and regulate the transcription of target genes. The SPI1 protein binds to a purine-rich sequence known as the PU-box, which is located near the promoters of its target genes .

Function and Mechanism

The primary function of SPI1 is to activate gene expression during the development of myeloid and B-lymphoid cells. It achieves this by binding to the PU-box and regulating the transcription of genes involved in cell differentiation and proliferation. Additionally, SPI1 can bind RNA and modulate pre-mRNA splicing, further influencing gene expression .

Role in Immune System

SPI1 is essential for the development and function of the immune system. It regulates the differentiation of myeloid cells, such as macrophages and neutrophils, as well as B-lymphocytes. By controlling the expression of genes involved in immune cell development, SPI1 ensures the proper functioning of the immune response .

Clinical Significance

Mutations or dysregulation of SPI1 can lead to various hematological disorders and cancers. For instance, overexpression of SPI1 has been associated with certain types of leukemia. As a result, SPI1 is considered a potential target for therapeutic interventions in hematological malignancies .

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 2025 TheBiotek. All Rights Reserved.