HMGB1 Human, Sf9

High-Mobility Group Box 1 Human Recombinant, Sf9
Shipped with Ice Packs
In Stock

Description

Production Process and Source

HMGB1 Human, Sf9 is produced via recombinant expression in Sf9 insect cells using baculovirus vectors. Key features include:

  • Host: Sf9 insect cells (derived from Spodoptera frugiperda) .

  • Fusion Tag: C-terminal His-Tag (8 amino acids) for purification .

  • Sequence: Amino acids 1–215, with a molecular mass of ~25 kDa .

  • Purification: Proprietary chromatographic techniques, yielding >90% purity by SDS-PAGE .

ParameterValueSource
Host SystemBaculovirus/Sf9 insect cells
Fusion TagHis-Tag (C-terminal)
Molecular Weight25 kDa
Purity>90% (SDS-PAGE)

Formulation and Stability

  • Buffer: 20 mM Tris-HCl (pH 8), 1 mM EDTA, 0.5 mM DTT, 10% glycerol .

  • Concentration: 1 mg/ml .

  • Storage:

    • Short-term (2–4 weeks): 4°C.

    • Long-term: -20°C; avoid freeze-thaw cycles.

    • Stability Enhancement: Add 0.1% HSA or BSA for prolonged storage .

Stability ConditionRecommendation
Short-term storage4°C, use within 2–4 weeks
Long-term storage-20°C, avoid repeated freeze-thaw
Carrier Protein0.1% HSA/BSA for enhanced stability

Post-Translational Modifications

HMGB1 Human, Sf9 retains glycosylation patterns absent in bacterial systems (e.g., E. coli). Glycosylation may influence receptor binding or immune signaling pathways .

Functional Properties

HMGB1 Human, Sf9 exhibits dual intracellular and extracellular roles:

Intracellular Functions

  • DNA Binding: Bends DNA via A-box and B-box domains, facilitating transcriptional regulation .

  • Chromatin Dynamics: Participates in nucleosome assembly and telomere maintenance .

Extracellular Functions

  • Inflammatory Signaling: Acts as a cytokine via TLR4, RAGE, and CXCR4 receptors, depending on redox states of Cys23, Cys45, and Cys106 .

    • Disulfide HMGB1 (Cys23–Cys45 oxidized, Cys106 reduced): Activates TLR4/MD-2 to induce TNF-α secretion .

    • Fully Reduced HMGB1: Binds CXCR4 and promotes chemotaxis .

    • Sulfonyl HMGB1: Inactive, promoting immune tolerance .

Redox StateReceptorBiological EffectSource
Disulfide (C23-C45)TLR4/MD-2Cytokine release (TNF-α, IL-6)
Fully ReducedCXCR4Chemotaxis (neutrophils)
Sulfonyl (C106 oxidized)NoneImmune tolerance

Inflammation and Immune Response

  • Neuroinflammation: HMGB1 Human, Sf9 induces COX-2, MMP-9, and chemokines (CCL2, CCL5) in astrocytes, promoting leukocyte infiltration .

  • Atherosclerosis: Macrophage-derived HMGB1 amplifies lesion progression via TLR4/NF-κB pathways .

Cancer and Angiogenesis

  • Proangiogenic Activity: HMGB1 binds VEGF receptors, promoting tumor vascularization .

  • Therapeutic Targeting: Neutralizing HMGB1 reduces metastasis and inflammation in preclinical models .

Comparative Analysis: Sf9 vs. Other Systems

ParameterSf9 (Insect Cells)E. coliMammalian (HEK293)
GlycosylationPresent (native-like)AbsentPresent (complex)
Redox SensitivityNative Cys residues preservedMay require refoldingNative
Purity>90%>95% (often GST-tagged)>95%
ApplicationsReceptor signaling studiesStructural studiesNative conformation

Key Research Findings

  • Redox-Dependent Signaling: Sf9-produced HMGB1 retains redox-sensitive cysteines critical for TLR4 activation .

  • ROS-Mediated Aggregation: ROS induces HMGB1 dimerization, enhancing cytokine activity .

Product Specs

Introduction
High mobility group protein B1 (HMGB1) is a highly conserved protein present in the nuclei and cytoplasm of eukaryotic cells. It plays a multifaceted role in cellular processes, including DNA replication, repair, transcription, and inflammation. HMGB1 is involved in various disease processes, including cancer, autoimmune diseases, and infectious diseases.
Description
Recombinant human HMGB1 protein, expressed in baculovirus-infected insect cells (Sf9), is a single, glycosylated polypeptide chain with a His-tag at the C-terminus. It encompasses amino acids 1-215, resulting in a protein of 223 amino acids with a molecular weight of 25 kDa. Purification is achieved using proprietary chromatographic methods.
Physical Appearance
Clear, colorless, and sterile-filtered solution.
Formulation
The HMGB1 protein is supplied in a solution containing 20mM Tris-HCl (pH 8.0), 1mM EDTA, 0.5mM DTT, and 10% glycerol at a concentration of 1mg/ml.
Stability
For short-term storage (up to 4 weeks), the product can be stored at 4°C. For long-term storage, it is recommended to store the protein at -20°C. The addition of a carrier protein, such as HSA or BSA (0.1%), is advised for extended storage. Repeated freezing and thawing should be avoided.
Purity
The purity of the HMGB1 protein is greater than 90% as determined by SDS-PAGE analysis.
Synonyms
HMG1, HMG3, SBP-1, Amphoterin, HMGB1, High-Mobility Group Box 1.
Source

Sf9 Insect Cells.

Amino Acid Sequence
MGKGDPKKPR GKMSSYAFFV QTCREEHKKK HPDASVNFSE FSKKCSERWK TMSAKEKGKF EDMAKADKAR YEREMKTYIP PKGETKKKFK DPNAPKRPPS AFFLFCSEYR PKIKGEHPGL SIGDVAKKLG EMWNNTAADD KQPYEKKAAK LKEKYEKDIA AYRAKGKPDA AKKGVVKAEK SKKKKEEEED EEDEEDEEEE EDEEDEDEEE DDDDELEHHH HHH.

Q&A

What is HMGB1 and what are its primary biological functions?

HMGB1 (High-Mobility Group Box 1, also known as HMG1, HMG3, SBP-1, or Amphoterin) is an abundant chromatin-binding protein found in eukaryotic cell nuclei that serves dual functions depending on its location. Inside cells, HMGB1 binds to DNA and participates in transcriptional regulation, DNA replication, repair, and chromatin remodeling . It enhances the binding affinity of transcription factors including p53, Rb, and NF-κB to DNA by bending the DNA molecule . In the cytoplasm, HMGB1 regulates apoptosis and autophagy by binding to proteins like Beclin1 and modulating caspase-3 activation . Extracellularly, HMGB1 functions as a pro-inflammatory cytokine with activities resembling tumor necrosis factor (TNF), contributing to various inflammatory and autoimmune conditions .

How does the redox state of HMGB1 affect its biological activity?

The immunological activity of HMGB1 is critically determined by the oxidation states of its three cysteine residues (Cys23, Cys45, and Cys106), creating three distinct forms with different functions :

Researchers must carefully consider HMGB1's redox state when designing experiments, as the biological outcomes can vary dramatically depending on which form predominates.

What post-translational modifications occur in HMGB1 and how do they influence function?

HMGB1 undergoes several important post-translational modifications that regulate its localization and function:

  • N-glycosylation: Occurs at residues N37, N134, and N135 . N-glycosylation affects HMGB1's nuclear mobility and reduces its DNA binding affinity through steric hindrance . This has been confirmed through FRAP (Fluorescence Recovery After Photobleaching) analysis showing that non-glycosylated HMGB1 mutants (N37Q/N134Q and N37Q/N135Q) have slower nuclear mobility than wild-type HMGB1 .

  • Acetylation: While not directly discussed in the provided materials, acetylation of lysine residues in HMGB1's nuclear localization sequences is known to prevent nuclear re-entry, facilitating cytoplasmic accumulation and subsequent secretion.

  • Oxidation: As detailed above, oxidation of cysteine residues creates functionally distinct forms of HMGB1 with different receptor binding capabilities and downstream effects .

When designing studies involving HMGB1, researchers should consider how these modifications might influence experimental outcomes and interpret results accordingly.

What are the technical advantages of expressing human HMGB1 in Sf9 cells?

Expressing human HMGB1 in Sf9 insect cells offers several significant advantages for research applications:

  • Post-translational modifications: Sf9 cells can perform important eukaryotic post-translational modifications including N-glycosylation, which has been confirmed for HMGB1 at residues N37, N134, and N135 . This allows researchers to study how these modifications affect HMGB1 function.

  • High protein yield: The baculovirus expression system in Sf9 cells typically produces higher protein yields than mammalian expression systems, facilitating structural and functional studies that require substantial amounts of purified protein.

  • Proper protein folding: Sf9 cells provide a eukaryotic environment that supports proper folding of complex proteins like HMGB1, preserving their functional domains and activities.

  • Simplified purification: The ability to add purification tags such as the 6×His tag allows for efficient purification using affinity chromatography followed by size exclusion chromatography to achieve high protein homogeneity .

  • Reduced endotoxin contamination: Unlike bacterial expression systems, insect cell systems produce proteins with minimal endotoxin contamination, which is crucial for immunological studies involving HMGB1.

What is the optimal expression and purification protocol for HMGB1 in Sf9 cells?

Based on the research literature, the following optimized protocol is recommended for HMGB1 expression and purification from Sf9 cells:

  • Cloning and baculovirus generation:

    • Clone the HMGB1 gene into a baculovirus transfer vector such as pFastBac HT-B

    • Include an N-terminal 6×His tag for purification

    • Generate recombinant baculovirus according to manufacturer's protocols (e.g., Invitrogen's system)

  • Protein expression:

    • Infect Sf9 cells with the recombinant baculovirus

    • Express for approximately 36 hours at 27°C

  • Cell lysis and initial purification:

    • Harvest cells and lyse in buffer containing 25 mM Tris-HCl pH 7.5, 300 mM NaCl, 10% glycerol, 30 mM imidazole, and 5 mM β-mercaptoethanol

    • Purify using Ni-NTA affinity chromatography

    • Elute with buffer containing increased imidazole concentration (300 mM)

  • Tag removal and final purification:

    • Treat with TEV protease for 16 hours at 4°C to remove the His tag

    • Perform size exclusion chromatography using a Superdex-200 column with buffer containing 25 mM Tris-HCl pH 7.5 and 150 mM NaCl

  • Quality control and storage:

    • Verify purity by SDS-PAGE

    • Store in buffer containing 20 mM Tris-HCl pH 8, 1 mM EDTA, 0.5 mM DTT, and 10% glycerol

    • For long-term storage, aliquot and keep at 4°C (if using within 2-3 weeks) or -80°C for extended periods

How can researchers verify the N-glycosylation status of recombinant HMGB1?

Verifying the N-glycosylation status of recombinant HMGB1 is crucial for functional studies. The following complementary approaches can be employed:

  • Enzymatic deglycosylation:

    • Treat purified HMGB1 with PNGase F, which specifically cleaves N-linked glycans

    • Resuspend the protein in 20 mM ammonium bicarbonate buffer (pH 8.0)

    • Heat at 98°C for 10 minutes to denature the protein

    • Incubate with PNGase F overnight at 37°C

    • Analyze by SDS-PAGE; a downward migration shift indicates the presence of N-glycosylation

  • Glycosylation inhibitor treatment:

    • Treat cells expressing HMGB1 with tunicamycin (an inhibitor of N-linked glycosylation)

    • Compare migration patterns of HMGB1 from treated and untreated cells by western blotting

    • A band shift indicates the presence of N-glycosylation in the native protein

  • LC-MS/MS analysis:

    • Perform liquid chromatography-tandem mass spectrometry on purified HMGB1

    • After PNGase F treatment, look for a mass increase of 0.98 Da at glycosylation sites, resulting from the conversion of asparagine to aspartic acid

    • This technique can precisely identify the glycosylation sites (N37, N134, and N135 in HMGB1)

  • Mutational analysis:

    • Generate HMGB1 mutants where potential glycosylation sites are replaced (e.g., N37Q, N134Q)

    • Compare the molecular weight and migration patterns of wild-type and mutant proteins

    • Differences confirm the presence and location of glycosylation sites

How can researchers assess HMGB1's DNA binding properties and the impact of glycosylation?

HMGB1's DNA binding properties and the effects of glycosylation can be evaluated using several complementary techniques:

  • Electrophoretic Mobility Shift Assay (EMSA):

    • Incubate purified HMGB1 (wild-type or glycosylation mutants) with plasmid DNA

    • Analyze complex formation by gel electrophoresis

    • Research has shown that non-glycosylated HMGB1 (N37Q/N134Q) forms stronger complexes with DNA than wild-type glycosylated HMGB1, as evidenced by retarded migration and some complexes remaining in the wells

  • Fluorescence Recovery After Photobleaching (FRAP):

    • Express EGFP-tagged HMGB1 variants in cells

    • Photobleach a nuclear area and monitor fluorescence recovery

    • Non-glycosylated HMGB1 mutants (N37Q/N134Q and N37Q/N135Q) show lower recovery percentages than wild-type HMGB1, indicating reduced mobility due to stronger DNA binding

  • Molecular modeling:

    • Model the interactions between N-acetyl-D-glucosamine (GlcNAc)-tagged HMGB1 A or B box and double-stranded DNA

    • This approach has revealed that GlcNAc molecules at N37 and N134 create steric hindrance that interferes with the phosphate backbone or nucleic acids of dsDNA

These techniques provide complementary insights into how glycosylation modulates HMGB1's interaction with DNA, which is essential for understanding its nuclear functions.

What experimental approaches can distinguish between different redox forms of HMGB1?

Distinguishing between HMGB1's redox forms is crucial for understanding its various biological activities. Researchers can employ these approaches:

  • Mass spectrometry-based redox proteomics:

    • Treat samples with alkylating agents to trap the existing redox state

    • Digest the protein and analyze peptide fragments by LC-MS/MS

    • Identify the presence of free thiols, disulfide bonds, or oxidized forms at Cys23, Cys45, and Cys106

  • Functional bioassays:

    • Test the ability of different HMGB1 preparations to activate TLR4 signaling (characteristic of disulfide HMGB1)

    • Assess chemotactic activity through CXCR4 (characteristic of fully reduced HMGB1)

    • Measure cytokine production in macrophages or dendritic cells

  • Redox-specific antibodies:

    • Use antibodies that specifically recognize different redox forms of HMGB1

    • Apply these in western blotting, ELISA, or immunohistochemistry

  • Site-directed mutagenesis:

    • Create cysteine-to-serine mutations to mimic the reduced state

    • Create cysteine-to-aspartate mutations to mimic the oxidized state

    • Compare the functional properties of these mutants with wild-type protein

These approaches allow researchers to correlate specific redox forms with biological activities, providing insight into how HMGB1 functions in different physiological and pathological contexts.

How does HMGB1 contribute to cellular processes like autophagy and what methods can assess this function?

HMGB1 plays critical roles in regulating autophagy through several mechanisms :

  • Binding to Beclin1: HMGB1 interacts with Beclin1, a key regulator of autophagy, promoting autophagosome formation while limiting apoptosis.

  • Protection of autophagy proteins: HMGB1 binds to Beclin1 and ATG5, suppressing calpain-induced cleavage and modulating the transition from autophagy to apoptosis during inflammation.

  • STAT3 signaling inhibition: HMGB1 may inhibit STAT3-mediated signaling, promoting autophagy and providing protection against infection in intestinal epithelial cells.

Methods to assess HMGB1's role in autophagy include:

  • Autophagy flux assays:

    • Monitor LC3-II levels with and without lysosomal inhibitors in cells expressing wild-type or mutant HMGB1

    • Use GFP-LC3 puncta formation as an indicator of autophagosome formation

  • Protein interaction studies:

    • Perform co-immunoprecipitation experiments to detect HMGB1-Beclin1 interactions

    • Use proximity ligation assays to visualize these interactions in situ

  • HMGB1 knockdown/knockout approaches:

    • Use siRNA, shRNA, or CRISPR-Cas9 to reduce or eliminate HMGB1 expression

    • Assess the impact on autophagy markers and autophagic flux

  • Subcellular localization tracking:

    • Monitor HMGB1 translocation from the nucleus to the cytoplasm during autophagy induction

    • Use fluorescently tagged HMGB1 or immunofluorescence with subcellular markers

These methods can reveal how HMGB1 contributes to autophagy regulation in different physiological and pathological contexts.

How can researchers investigate HMGB1's non-classical secretion mechanisms?

HMGB1 lacks an ER-targeting signal peptide yet undergoes N-glycosylation and is secreted through non-classical pathways. To investigate these mechanisms:

  • Secretion pathway inhibitor studies:

    • Treatment with brefeldin A and nocodazole (inhibitors of ER-Golgi transport and microtubule polymerization) does not decrease LPS-stimulated HMGB1 secretion, indicating that the conventional secretion pathway is not involved

    • Use other pathway-specific inhibitors to identify the mechanisms involved

  • Live-cell imaging:

    • Create fluorescently tagged HMGB1 constructs

    • Track their movement from the nucleus to the extracellular space in real-time

    • Co-label with markers for different vesicular compartments

  • Proteomics approach:

    • Identify HMGB1-interacting proteins during the secretion process using proximity labeling techniques

    • Compare the interactomes of wild-type and glycosylation-mutant HMGB1

  • Glycosylation status analysis:

    • Compare the glycosylation patterns of intracellular versus secreted HMGB1

    • Determine if glycosylation status changes during the secretion process

  • Secretion kinetics:

    • Use pulse-chase experiments with metabolic labeling to track the time course of HMGB1 synthesis, modification, and secretion

These approaches can help elucidate the unconventional secretion mechanisms of HMGB1 and the role of glycosylation in this process.

What is the relationship between HMGB1 forms and disease pathogenesis?

HMGB1 is implicated in various disease processes, with different forms and modifications playing distinct roles:

  • Liver diseases:

    • HMGB1 contributes to alcoholic liver disease (ALD) progression by promoting inflammation and lipid accumulation

    • HMGB1 knockdown reduces SREBP-1 synthesis and lipid accumulation, providing protection against disease progression

    • In liver fibrosis, HMGB1 activates pMEK1/2/pERK1/2/pcJun and PI3K/Akt signaling, enhancing Collagen type I synthesis via RAGE

  • Inflammatory conditions:

    • In Sjögren's syndrome, increased extracellular HMGB1 in salivary glands indicates involvement in the inflammatory process

    • In sepsis, HMGB1 catalyzes movement of LPS monomers to CD14, initiating TLR4-mediated proinflammatory responses

  • Cancer:

    • HMGB1 plays a role in the relationship between necrosis and malignancy in glioma tumors

    • Overexpression is common in gastrointestinal stromal tumors and related to KIT mutation

Research methods to investigate these relationships include:

  • Redox-specific detection in patient samples

  • Animal models with redox-locked HMGB1 variants

  • Cell-type specific knockout studies

  • Therapeutic targeting of specific HMGB1 forms

Understanding which HMGB1 forms predominate in different disease states could guide the development of more targeted therapeutic approaches.

How can researchers develop HMGB1-targeted therapeutic strategies?

Based on HMGB1's roles in disease pathogenesis, several therapeutic strategies can be explored:

  • Redox-specific targeting:

    • Develop compounds that selectively bind to and neutralize pro-inflammatory forms of HMGB1

    • Create redox-modulating agents that promote conversion to non-inflammatory forms

  • Post-translational modification inhibitors:

    • Design inhibitors of specific enzymes that modify HMGB1 (e.g., glycosylation inhibitors)

    • Target acetylation pathways to prevent nuclear-cytoplasmic translocation and secretion

  • Receptor antagonists:

    • Develop antagonists for HMGB1 receptors (TLR4, RAGE) to block downstream signaling

    • Create decoy receptors to capture extracellular HMGB1

  • Domain-specific antibodies:

    • Generate antibodies that recognize specific domains or modified forms of HMGB1

    • Use these for both diagnostic purposes and therapeutic neutralization

  • Gene therapy approaches:

    • Explore CRISPR-Cas9 or antisense oligonucleotides to modulate HMGB1 expression

    • Develop cell-type specific delivery systems for maximal therapeutic effect

For all these approaches, researchers should:

  • Validate targets in relevant disease models

  • Assess specificity to minimize off-target effects

  • Evaluate effects on both pathological and physiological HMGB1 functions

  • Consider combinatorial approaches targeting multiple aspects of HMGB1 biology

What are common challenges in expressing and purifying functional HMGB1 from Sf9 cells?

Researchers frequently encounter these challenges when working with HMGB1 in Sf9 cells:

  • Protein aggregation:

    • HMGB1 can form aggregates during expression or purification

    • Solution: Optimize buffer conditions, include stabilizing agents like glycerol (10%), and maintain reducing conditions with DTT (0.5 mM)

  • Proteolytic degradation:

    • HMGB1 may be susceptible to proteases

    • Solution: Include protease inhibitors during lysis and purification, work at 4°C, and minimize processing time

  • Inconsistent glycosylation:

    • Insect cells may produce heterogeneous glycosylation patterns

    • Solution: Verify glycosylation status using PNGase F treatment and LC-MS/MS analysis

  • Maintaining redox state:

    • Different redox forms have distinct functions

    • Solution: Work under controlled redox conditions and verify the redox state using mass spectrometry

  • Endotoxin contamination:

    • Even low levels can affect functional assays

    • Solution: Use endotoxin-free reagents and include endotoxin removal steps during purification

  • Tag interference:

    • Purification tags may affect protein function

    • Solution: Include TEV protease treatment to remove tags, followed by additional purification steps

Careful attention to these factors will help ensure the production of homogeneous, functional HMGB1 suitable for structural and functional studies.

How can researchers accurately interpret contradictory findings about HMGB1 in the literature?

The HMGB1 literature contains apparent contradictions that can be resolved by considering several key factors:

  • Redox state variations:

    • Different laboratories may work with HMGB1 in different redox states

    • Solution: Always characterize and report the redox state of HMGB1 preparations using mass spectrometry or functional assays

  • Post-translational modification heterogeneity:

    • Different expression systems and purification methods may yield HMGB1 with varying modifications

    • Solution: Fully characterize modifications and consider their impact on experimental outcomes

  • Species differences:

    • HMGB1 from different species may have subtle functional differences

    • Solution: Clearly specify the species origin and consider evolutionary conservation when comparing results

  • Experimental context variations:

    • HMGB1 functions differently depending on cell type, disease state, and microenvironment

    • Solution: Carefully document experimental conditions and avoid overgeneralizing findings

  • Technical approaches:

    • Different detection methods may have varying sensitivities and specificities

    • Solution: Use multiple complementary techniques to confirm findings

When reviewing literature, researchers should pay particular attention to these factors and design experiments that specifically address potential sources of contradiction.

What techniques can distinguish between intracellular and extracellular functions of HMGB1?

Distinguishing between HMGB1's intracellular and extracellular functions requires specialized approaches:

  • Compartment-specific manipulations:

    • Use cell-impermeable neutralizing antibodies to target only extracellular HMGB1

    • Create fusion proteins with compartment-targeting sequences to restrict HMGB1 to specific locations

  • Conditional knockout/knockin models:

    • Generate models with tissue-specific or inducible HMGB1 manipulation

    • Create mutants with altered secretion but preserved intracellular functions

  • Secretion pathway analysis:

    • Use inhibitors of conventional (brefeldin A) and non-conventional secretion pathways

    • Monitor the impact on HMGB1-dependent processes

  • Receptor antagonism:

    • Block extracellular receptors (TLR4, RAGE) without affecting intracellular HMGB1

    • Compare effects with direct HMGB1 inhibition

  • Fractionation approaches:

    • Carefully separate nuclear, cytoplasmic, and extracellular fractions

    • Analyze HMGB1 forms and binding partners in each compartment

  • Real-time imaging:

    • Track fluorescently tagged HMGB1 movement between compartments

    • Correlate localization changes with functional outcomes

These approaches can help researchers determine whether observed effects are due to intracellular functions of HMGB1 (DNA binding, autophagy regulation) or its extracellular activities (cytokine-like functions, receptor activation).

Product Science Overview

Introduction

High-Mobility Group Box 1 (HMGB1) is a highly conserved, non-histone chromatin-binding protein found in the nucleus of eukaryotic cells. It plays a crucial role in the regulation of DNA-dependent processes such as transcription, replication, and repair. HMGB1 is also known for its role as a cytokine, influencing various cellular processes outside the nucleus.

Structure and Function

HMGB1 consists of 215 amino acids and has a molecular mass of approximately 25 kDa . It contains two DNA-binding domains known as HMG-boxes and an acidic C-terminal tail. The protein can bind to DNA in a non-sequence-specific manner, facilitating the bending and looping of DNA, which is essential for the formation of nucleoprotein complexes .

Inside the cell, HMGB1 binds to DNA and is involved in transcriptional regulation. It acts as an architectural factor, stabilizing nucleoprotein complexes and facilitating the interaction of transcription factors with their target DNA sequences . Outside the cell, HMGB1 functions as a cytokine with activities resembling those of tumor necrosis factor (TNF). It can activate monocytes and neutrophils, playing a significant role in inflammation and immune responses .

Recombinant HMGB1 (Sf9)

Recombinant HMGB1 is produced using the baculovirus expression system in Sf9 insect cells. This method allows for the production of a glycosylated form of the protein, which closely resembles the native protein found in human cells . The recombinant HMGB1 is fused to an 8 amino acid His-Tag at the C-terminus, facilitating its purification through chromatographic techniques .

The recombinant HMGB1 (Human, Sf9) is available in various quantities and is shipped with ice packs to maintain its stability. It is formulated as a sterile, filtered, colorless solution containing 20mM Tris-HCl (pH 8), 1mM EDTA, 0.5mM DTT, and 10% glycerol . The protein is stable at 4°C for short-term storage and at -20°C for long-term storage, with the addition of a carrier protein to prevent multiple freeze-thaw cycles .

Applications

HMGB1 has a wide range of applications in research and clinical settings. It is used to study chromatin dynamics, transcriptional regulation, and DNA repair mechanisms. Additionally, HMGB1 is involved in various pathological conditions, including cancer, sepsis, and autoimmune diseases . Its role as a cytokine makes it a target for therapeutic interventions aimed at modulating immune responses and inflammation.

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.