STIP1 Human

Stress-Induced-Phosphoprotein 1 Human Recombinant
Shipped with Ice Packs
In Stock

Description

Molecular Structure and Functional Domains

STIP1 contains three tetratricopeptide repeat (TPR) domains (TPR1, TPR2A, TPR2B) and two aspartate/proline-rich (DP) domains . These structural motifs enable interactions with:

  • HSP70 (via TPR1 and DP2 domains)

  • HSP90 (via TPR2A and TPR2B domains)

  • LSD1 (lysine-specific demethylase 1) and GSK3β (glycogen synthase kinase-3 beta)

Table 1: Key Domains and Binding Partners of STIP1

DomainBinding PartnerFunctional Role
TPR1HSP70, LSD1Facilitates HSP70 substrate transfer
TPR2AHSP90, GSK3βRegulates HSP90 ATPase activity
TPR2BLSD1, GSK3βScaffolds LSD1-GSK3β phosphorylation
DP2HSP70Enhances HSP70 ATPase activity

Scaffold for LSD1-GSK3β Phosphorylation

STIP1 stabilizes the interaction between LSD1 and GSK3β, enabling GSK3β-mediated phosphorylation of LSD1 at residues S707/S711. This modification:

  • Increases LSD1 stability by preventing nuclear export

  • Enhances cancer cell proliferation and migration

  • Correlates with poor prognosis in oral squamous cell carcinoma (OSCC) and ovarian cancer

Regulation of Protein Stability

STIP1-HSP90 complexes maintain LSD1 stability by inhibiting proteasomal degradation. Silencing STIP1 reduces LSD1 levels, which is reversible with proteasome inhibitors like MG132 .

Prognostic Biomarker

Table 2: Clinical Correlations of STIP1 Overexpression

Cancer TypeHazard Ratio (HR)Outcome MeasureSource
OSCC3.07 (CSS)Reduced 5-year survival
Ovarian Cancer2.56 (OS)Advanced tumor grade
Colorectal Cancer2.10 (DFS)Metastasis risk

Inhibitor Synergy

Combined inhibition of LSD1 (SP2509) and GSK3β (LY2090314) synergistically induces cancer cell death in vitro .

Intracellular Targeting Strategies

  • Anti-STIP1 antibodies: Disrupt STIP1-HSP90 interactions, reducing LSD1 stability .

  • Peptide 520: A cell-penetrating peptide that blocks STIP1 scaffolding, impairing cancer cell migration and invasion .

Table 3: Preclinical Therapeutic Efficacy

InterventionCancer ModelEffect ObservedReference
SP2509 + LY2090314OSCC, OvarianSynergistic apoptosis
Peptide 520Gastric, HCCReduced metastasis

Research Tools and Recombinant Proteins

Recombinant human STIP1 (e.g., ab222344, Cusabio EP022831HU) is widely used to study its autocrine effects. Key applications include:

  • Cell proliferation assays: STIP1 stimulates migration in ovarian cancer cells .

  • Protein interaction studies: Mapping TPR domain binding via pull-down assays .

Future Directions

  • Biomarker validation: Multicenter studies to confirm STIP1’s prognostic utility in OSCC .

  • Combination therapies: Testing LSD1/GSK3β inhibitors in xenograft models .

  • Chemoresistance: Exploring STIP1’s role in cisplatin resistance .

Product Specs

Introduction
STIP1 is an adaptor protein that facilitates the functions of HSP70 and HSP90 in the protein folding process. It acts as a mediator by binding to both HSP90 and substrate-bound HSP70, thereby promoting the transfer of proteins from HSP70 to HSP90. STIP1 exhibits regulatory effects on these chaperones by stimulating the ATPase activity of HSP70 and inhibiting the ATPase activity of HSP90, suggesting its role in modulating their conformations and ATPase cycles. Notably, genetic variations in STIP1 have been implicated in the regulation of corticosteroid response among asthmatic individuals experiencing reduced lung function.
Description
STIP1 Human Recombinant, produced in E. coli, is a single, non-glycosylated polypeptide chain consisting of 543 amino acids (1-543 a.a.). It has a molecular mass of 62.6 kDa. The protein undergoes purification using proprietary chromatographic techniques.
Physical Appearance
Sterile Filtered colorless solution.
Formulation
The STIP1 protein solution is provided at a concentration of 0.5 mg/ml. It is formulated in a buffer containing 20mM Tris-HCl (pH 8.0), 10% glycerol, 1mM DTT, and 0.1M NaCl.
Stability
For short-term storage (2-4 weeks), the product can be stored at 4°C. For extended storage, it is recommended to freeze the product at -20°C. To ensure optimal stability during long-term storage, consider adding a carrier protein (0.1% HSA or BSA). It is important to avoid subjecting the product to multiple freeze-thaw cycles.
Purity
The purity of the STIP1 protein is greater than 95.0%, as determined by SDS-PAGE analysis.
Synonyms
Stress Induced Phosphoprotein 1, Transformation-Sensitive Protein IEF SSP 3521, Renal Carcinoma Antigen NY-REN-11, Stress-Induced-Phosphoprotein 1, Hsp70/Hsp90-Organizing Protein, Hsc70/Hsp90-Organizing Protein, STI1, HOP, Epididymis Secretory Sperm Binding Protein Li 94n, NY-REN-11 Antigen, IEF-SSP-3521, HEL-S-94n, STI1L, P60.
Source
Escherichia Coli.
Amino Acid Sequence
MEQVNELKEK GNKALSVGNI DDALQCYSEA IKLDPHNHVL YSNRSAAYAK KGDYQKAYED GCKTVDLKPD WGKGYSRKAA ALEFLNRFEE AKRTYEEGLK HEANNPQLKE GLQNMEARLA ERKFMNPFNM PNLYQKLESD PRTRTLLSDP TYRELIEQLR NKPSDLGTKL QDPRIMTTLS VLLGVDLGSM DEEEEIATPP PPPPPKKETK PEPMEEDLPE NKKQALKEKE LGNDAYKKKD FDTALKHYDK AKELDPTNMT YITNQAAVYF EKGDYNKCRE LCEKAIEVGR ENREDYRQIA KAYARIGNSY FKEEKYKDAI HFYNKSLAEH RTPDVLKKCQ QAEKILKEQE RLAYINPDLA LEEKNKGNEC FQKGDYPQAM KHYTEAIKRN PKDAKLYSNR AACYTKLLEF QLALKDCEEC IQLEPTFIKG YTRKAAALEA MKDYTKAMDV YQKALDLDSS CKEAADGYQR CMMAQYNRHD SPEDVKRRAM ADPEVQQIMS DPAMRLILEQ MQKDPQALSE HLKNPVIAQK IQKLMDVGLI AIR.

Q&A

What is STIP1 and what are its primary functions in human cells?

STIP1, also known as HSP70/90 organizing protein (HOP), is a 62.6-kDa co-chaperone protein that mediates the interaction between heat shock proteins HSP70 and HSP90. It contains three tetratricopeptide repeat (TPR) domains that enable it to serve as a scaffold for protein-protein interactions . STIP1 plays critical roles in:

  • Protein folding and stabilization

  • Modulation of chaperone activities beyond HSP70 and HSP90

  • Regulation of cell proliferation pathways

  • Supporting cellular responses to stress conditions

  • Mediating protein trafficking between cytoplasm and nucleus

In research contexts, STIP1 is typically studied using co-immunoprecipitation assays to identify binding partners, immunoblotting to quantify expression levels, and immunofluorescence to determine subcellular localization.

How is STIP1 expression regulated in normal human tissues versus disease states?

STIP1 expression varies significantly between normal and pathological conditions. In normal tissues, STIP1 maintains baseline expression with upregulation occurring during cellular stress. In cancer and neurodegenerative conditions, STIP1 regulation becomes dysregulated through multiple mechanisms:

  • Cancer tissues consistently show elevated STIP1 expression compared to adjacent normal tissues

  • Following ischemic events, STIP1 expression rapidly increases and then gradually decreases in affected tissues

  • In neurodegenerative disorders, STIP1 has been found to interact with prion protein, preventing amyloid-β-induced synaptic loss and neuronal death

Methodologically, researchers investigating STIP1 regulation should employ qRT-PCR for transcript analysis, western blotting for protein quantification, and chromatin immunoprecipitation (ChIP) assays to identify transcription factors regulating STIP1 expression.

What cellular compartments contain STIP1 and how does its localization impact function?

STIP1 demonstrates dynamic localization patterns that directly influence its functional capabilities:

  • Primarily cytoplasmic under normal conditions

  • Nuclear translocation observed under specific stress conditions

  • Cell surface localization in certain cell types, particularly cancer cells

  • Secreted forms detected in extracellular environments

Notably, research has demonstrated that mutations affecting LSD1 phosphorylation sites (S707A/S711A) result in translocation of LSD1 from the nucleus to the cytoplasm, a process mediated by STIP1 scaffolding functions . This highlights how STIP1 localization directly impacts its ability to regulate client protein distribution and function.

To study STIP1 localization, researchers should utilize subcellular fractionation followed by western blotting, confocal microscopy with immunofluorescence, and live cell imaging with fluorescently tagged STIP1 constructs.

How does STIP1 function as a molecular scaffold in signaling pathways, and what methodologies best capture these interactions?

STIP1 serves as a sophisticated molecular scaffold that facilitates the assembly of multi-protein complexes critical for signal transduction. Research has revealed several key insights:

STIP1's scaffolding properties are domain-specific, with different domains mediating distinct protein interactions:

  • TPR1 and TPR2B domains bind to the AOL domain of LSD1

  • TPR2A and TPR2B domains interact with the kinase domain of GSK3β

This precise molecular architecture enables STIP1 to position enzymes and substrates in optimal configurations for post-translational modifications, as demonstrated by its facilitation of GSK3β-mediated phosphorylation of LSD1 .

To effectively study these scaffolding functions, researchers should employ:

  • Proximity ligation assays to detect protein-protein interactions in situ

  • FRET (Förster resonance energy transfer) analysis to measure real-time interactions

  • Hydrogen-deuterium exchange mass spectrometry to map interaction interfaces

  • Domain deletion/mutation studies to identify specific interaction regions

These techniques collectively enable researchers to characterize the dynamic nature of STIP1-mediated complex formation and its functional consequences.

What are the mechanisms through which STIP1 modulates inflammatory responses in neurological conditions?

STIP1 demonstrates significant immunomodulatory functions in neurological disorders, particularly in ischemia-reperfusion injury contexts. Multiple mechanisms have been identified:

  • STIP1 is co-localized with Iba-1 (a microglial marker) in rat spinal cord following ischemia-reperfusion injury, indicating direct interaction with microglial cells

  • STIP1 overexpression significantly reduces pro-inflammatory cytokine production (TNF-α and IL-6) in ischemic spinal cord tissue

  • STIP1 deactivates the NF-κB signaling pathway by:

    • Increasing IκBβ expression

    • Reducing nuclear translocation of NF-κB p65

    • Competing with IκBβ for binding to HSPA8 (heat shock protein family A member 8)

These findings suggest that STIP1's neuroprotective effects are partially mediated through its anti-inflammatory actions. The research methodologies required to investigate these pathways include:

  • Immunoprecipitation to identify binding partners in inflammatory pathways

  • Cytokine ELISAs to measure inflammatory mediator production

  • Luciferase reporter assays to quantify NF-κB activity

  • Flow cytometry to assess microglial activation states

  • In vivo models of ischemia-reperfusion with STIP1 overexpression or knockdown

What post-translational modifications of STIP1 have been identified, and how do they regulate its function?

STIP1 undergoes various post-translational modifications (PTMs) that fine-tune its activity, stability, and interaction profile. Current research has identified:

Modification TypeSpecific SitesFunctional ImpactResearch Methods
PhosphorylationMultiple Ser/Thr residuesAlters binding affinity to client proteins; Regulates cellular localizationMass spectrometry; Phospho-specific antibodies; Site-directed mutagenesis
UbiquitinationNot fully characterizedControls protein stability and turnoverImmunoprecipitation with ubiquitin antibodies; Proteasome inhibition studies
SUMOylationUnder investigationMay regulate nuclear-cytoplasmic shuttlingSUMO-IP; In vitro SUMOylation assays

Researchers investigating STIP1 PTMs should implement:

  • Phosphatase treatments to assess global phosphorylation effects

  • 2D gel electrophoresis to separate modified isoforms

  • Targeted proteomics approaches to quantify specific modifications

  • Functional assays comparing wild-type versus PTM-deficient mutants

These studies reveal that STIP1's function is not static but dynamically regulated through a complex pattern of modifications that respond to cellular context and stress conditions.

How does STIP1 contribute to oncogenesis, and what experimental approaches best characterize its oncogenic mechanisms?

STIP1 promotes oncogenesis through multiple distinct mechanisms that collectively enhance cancer cell survival, proliferation, and metastasis:

  • Formation of an HSP90-JAK2-STAT3 complex that activates pro-survival signaling

  • Facilitation of GSK3β-mediated phosphorylation of LSD1, enhancing cell proliferation

  • Promotion of epithelial-mesenchymal transition via increased nuclear shuttling of Snail1 in hepatocellular carcinoma

  • Activation of ERK or ALK2-SMAD signaling pathways when secreted extracellularly

The tissue-specific patterns of STIP1-mediated oncogenesis include its overexpression in liver, pancreatic, ovarian, colon, and breast tumors . Importantly, both intracellular and extracellular forms of STIP1 contribute to cancer progression through distinct molecular mechanisms.

To effectively study STIP1's oncogenic functions, researchers should employ:

  • Cancer cell line models with STIP1 knockdown or overexpression

  • Patient-derived xenografts to assess clinical relevance

  • Phospho-proteomics to identify signaling pathway alterations

  • Chromatin immunoprecipitation sequencing (ChIP-seq) to identify transcriptional changes

  • Co-culture systems to assess tumor-stroma interactions mediated by STIP1

What therapeutic approaches targeting STIP1 show promise for cancer treatment?

Emerging research suggests multiple strategies for targeting STIP1 therapeutically:

  • Combinatorial approaches using LSD1 inhibitors (SP2509) and GSK3β inhibitors (LY2090314) have demonstrated synergistic antitumor effects in preclinical models

  • Intracellular targeting of STIP1 has been shown to inhibit cancer cell line growth and promote caspase 3-dependent apoptotic cell death

  • Antibody-based approaches targeting extracellular STIP1 to prevent receptor activation

  • Peptide-based inhibitors designed to disrupt specific STIP1 protein-protein interactions

When designing STIP1-targeted therapeutic studies, researchers should consider:

  • Specificity of STIP1 inhibition to minimize off-target effects

  • Differential targeting of intracellular versus extracellular STIP1

  • Tissue-specific effects given STIP1's varied roles across cell types

  • Potential for resistance mechanisms through compensatory pathways

  • Combined inhibition strategies targeting multiple STIP1-dependent processes

How does the STIP1 interactome differ between normal and cancer cells?

The STIP1 protein interaction network undergoes significant remodeling during oncogenic transformation:

  • Cancer cells show enhanced interaction between STIP1 and oncogenic client proteins

  • The stoichiometry of STIP1-HSP90-HSP70 complexes is altered in malignant cells

  • Novel cancer-specific interactions emerge that are absent in normal tissues

  • Subcellular redistribution of STIP1 complexes occurs in tumor cells

Research methodologies to characterize these differential interactomes include:

  • BioID or APEX proximity labeling to identify context-specific interactions

  • Quantitative interaction proteomics comparing normal versus cancer cells

  • Super-resolution microscopy to visualize interaction complexes in situ

  • Correlation of interactome changes with cancer progression stages

Understanding these cancer-specific interactions provides opportunities for developing highly selective therapeutic interventions that disrupt oncogenic functions while preserving normal STIP1 activities.

What neuroprotective mechanisms are mediated by STIP1, and how can they be therapeutically exploited?

STIP1 demonstrates significant neuroprotective properties through several complementary mechanisms:

  • Binding to prion protein to prevent interaction with soluble amyloid-β oligomers

  • Prevention of amyloid-β-induced synaptic loss and neuronal death in cultured neurons

  • Inhibition of long-term potentiation disruption in hippocampal specimens

  • Amelioration of ischemia/reperfusion-induced neuronal injury and inflammation in rat spinal cord

  • Promotion of recruitment of bone marrow-derived cells to ischemic brain regions

Studies in heterozygous STIP1 knockout mice have confirmed its neuroprotective role, as these animals display aggravated ischemic damage in the brain, while extracellular STIP1 treatment prevents ischemia-mediated neuronal cell death .

Researchers investigating STIP1's neuroprotective functions should employ:

  • Primary neuronal cultures with STIP1 modulation

  • Electrophysiological recordings to assess synaptic function

  • Cerebral ischemia models with STIP1 intervention

  • Behavioral testing to assess cognitive and motor outcomes

  • Cell-specific conditional knockout models to dissect cell-autonomous effects

How does STIP1 influence microglial activation and neuroinflammation?

STIP1 exerts significant effects on microglial function and neuroinflammatory processes:

  • Co-localization with Iba-1 confirms STIP1 expression in microglial cells in rat spinal cord

  • STIP1 overexpression inhibits microglial activation following ischemic injury

  • STIP1 suppresses production of pro-inflammatory cytokines (TNF-α and IL-6) in activated microglia

  • Mechanistically, STIP1 deactivates the NF-κB signaling pathway by:

    • Increasing IκBβ expression

    • Decreasing HSPA8 expression

    • Reducing binding of IκBβ to HSPA8

These findings suggest that STIP1 competes with IκBβ for binding to HSPA8, thereby stabilizing IκBβ and preventing NF-κB activation. This represents a novel mechanism through which STIP1 restrains excessive inflammatory responses after neurological injury.

Research approaches to study STIP1 in neuroinflammation include:

  • Mixed glial culture systems with STIP1 manipulation

  • In vivo models of neuroinflammation with cell-specific STIP1 modulation

  • NF-κB reporter systems to quantify pathway activity

  • Cytokine/chemokine profiling in response to STIP1 intervention

  • Single-cell RNA sequencing to identify microglial phenotype changes

What are the optimal methods for detecting and quantifying both intracellular and extracellular STIP1?

Accurate detection and quantification of STIP1 across different cellular compartments requires specialized methodologies:

For intracellular STIP1:

  • Western blotting remains the gold standard for quantification in cell lysates

  • Immunofluorescence provides spatial information about subcellular localization

  • Flow cytometry allows for single-cell analysis of intracellular STIP1 levels

  • Subcellular fractionation enables compartment-specific quantification

For extracellular STIP1:

  • ELISA assays specifically optimized for STIP1 detection in culture media or biological fluids

  • Proximity extension assays for highly sensitive detection in complex samples

  • Surface plasmon resonance to measure binding kinetics with cell surface receptors

  • Immunoprecipitation from conditioned media followed by western blotting

When designing STIP1 detection protocols, researchers should carefully validate antibody specificity, establish appropriate controls, and consider both total and post-translationally modified forms of the protein.

What genetic models and tools are available for manipulating STIP1 expression in research?

Researchers have multiple options for modulating STIP1 expression and function:

Genetic Knockout/Knockdown Models:

  • Complete STIP1 knockout causes embryonic lethality, necessitating conditional approaches

  • Heterozygous STIP1 knockout mice are viable but show enhanced vulnerability to stress conditions

  • Inducible and tissue-specific STIP1 knockout models using Cre-loxP systems

  • CRISPR/Cas9-mediated knockout in cell lines and primary cultures

Overexpression Systems:

  • Lentiviral vectors containing STIP1 coding sequences for stable overexpression

  • Transient transfection approaches for acute overexpression studies

  • Inducible expression systems for temporal control of STIP1 induction

Domain-Specific Manipulation:

  • Expression of isolated TPR domains to dissect domain-specific functions

  • Point mutations in specific binding interfaces to disrupt selected interactions

  • Domain deletion constructs to eliminate particular functional regions

Researchers should carefully select the appropriate model based on their specific research question, considering aspects such as temporary versus permanent manipulation, cell-type specificity, and potential compensatory mechanisms.

What emerging technologies are advancing our understanding of STIP1 biology?

Several cutting-edge technologies are transforming STIP1 research:

Structural Biology Approaches:

  • Cryo-electron microscopy to visualize STIP1 complexes at near-atomic resolution

  • Hydrogen-deuterium exchange mass spectrometry to map dynamic conformational changes

  • NMR spectroscopy to analyze solution-state protein dynamics and interactions

Single-Cell Technologies:

  • Single-cell RNA sequencing to identify cell populations with differential STIP1 expression

  • Mass cytometry to simultaneously measure STIP1 and multiple signaling nodes

  • Live-cell imaging with fluorescent biosensors to track STIP1 activity in real-time

Multi-omics Integration:

  • Proteogenomic approaches linking STIP1 genomic variations to protein expression patterns

  • Integrative network analysis to position STIP1 within broader cellular systems

  • Spatial transcriptomics to map STIP1 expression patterns within complex tissues

Therapeutic Development Platforms:

  • High-throughput screening systems to identify STIP1 modulators

  • Peptide display technologies to develop interaction-specific inhibitors

  • PROTAC (proteolysis targeting chimera) approaches for selective STIP1 degradation

These innovative technologies are enabling researchers to address previously intractable questions about STIP1 biology and accelerating the translation of basic discoveries into potential therapeutic applications.

Product Science Overview

Gene and Protein Structure

The STIP1 gene is located on chromosome 11q13.1 and consists of 14 exons . The protein itself is characterized by the presence of nine tetratricopeptide repeat (TPR) motifs, which are clustered into domains of three TPRs each . These motifs are essential for protein-protein interactions, allowing STIP1 to bind both Hsp70 and Hsp90, facilitating the transfer of substrate proteins between these chaperones .

Function and Mechanism

STIP1 functions as a molecular scaffold, bringing together Hsp70 and Hsp90 to form a chaperone complex that assists in the proper folding of newly synthesized proteins and the refolding of misfolded proteins . This process is vital for maintaining cellular protein homeostasis, especially under stress conditions where the demand for protein folding increases .

In addition to its role in protein folding, STIP1 is involved in various cellular processes, including signal transduction, cell cycle regulation, and apoptosis . It has been shown to interact with several signaling molecules, such as the protein tyrosine kinase JAK2, and modulate their activity through phosphorylation .

Clinical Significance

STIP1 is commonly overexpressed in various types of cancer, where it contributes to tumor progression by promoting cell proliferation and survival . Its interaction with Hsp90 is particularly important in cancer cells, as Hsp90 stabilizes many oncogenic proteins. Therefore, targeting the STIP1-Hsp90 interaction is being explored as a potential therapeutic strategy for cancer treatment .

Recombinant STIP1

Human recombinant STIP1 is produced using recombinant DNA technology, which involves inserting the human STIP1 gene into a suitable expression system, such as bacteria or yeast, to produce the protein in large quantities. This recombinant protein is used in research to study its structure, function, and interactions with other proteins, as well as in drug development to screen for potential inhibitors of its 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.