HMGN1 Human

High-Mobility Group Nucleosome Binding Domain 1 Human Recombinant
Shipped with Ice Packs
In Stock

Description

Molecular Structure and Domains

HMGN1 consists of 100–108 amino acids (depending on isoform) with a molecular mass of ~11.7 kDa . Its structure includes two functional domains:

  • N-terminal Nucleosome-Binding Domain (NBD): Mediates interaction with nucleosomes, preferentially binding to transcriptionally active chromatin .

  • C-terminal Chromatin-Unfolding Domain (CHUD): Facilitates destabilization of higher-order chromatin structure by reducing linker histone H1 binding .

Therapeutic Efficacy in Murine Models

TreatmentTumor ModelOutcome
HMGN1 + anti-CD4Colon26, B16F1070% tumor regression
minP1 + anti-PD-L1Colon26, LLCDurable rejection in 30–70% of mice

Recombinant HMGN1 Production

Recombinant human HMGN1 is produced in E. coli for research applications :

  • Purity: >95% (SDS-PAGE verified) .

  • Activity: Effective concentration range: 0.6–3 μg/mL in chromatin remodeling assays .

Interaction Networks

HMGN1 interacts with proteins involved in transcription and DNA repair:

InteractorFunctionInteraction Score
HMGN2Chromatin decompaction0.994
EP300Histone acetyltransferase activity0.947
ERCC6Transcription-coupled repair0.961
TLR4Innate immune signalingReported

Clinical Implications and Future Directions

  • Chemotherapy Sensitivity: HMGN1 loss enhances lung adenocarcinoma (LUAD) sensitivity to cisplatin and olaparib by impairing homologous recombination repair .

  • Biomarker Potential: Correlates with immune infiltration in HER2+ breast cancer and head/neck carcinoma .

Ongoing Challenges

  • Species-specific functional disparities (e.g., TCR role in mice vs. humans) .

  • Optimizing HMGN1-derived peptides for clinical use without off-target effects .

Product Specs

Introduction
HMGN1, a non-histone chromosomal protein, binds to nucleosomal DNA, influencing the interaction between DNA and the histone octamer. This interaction plays a crucial role in development by maintaining specific genes in a transcriptionally active chromatin state. HMGN1 has also been shown to inhibit the phosphorylation of nucleosomal histones H3 and H2A by the kinases RPS6KA5/MSK1 and RPS6KA3/RSK2.
Description
Recombinant human HMGN1 protein, expressed in E. coli, is a single, non-glycosylated polypeptide chain comprising 108 amino acids. This includes the full HMGN1 sequence (1-100 amino acids) and an 8 amino acid His-tag fused at the C-terminus. The protein has a molecular weight of 11.7 kDa and is purified using standard chromatographic techniques.
Physical Appearance
Clear, colorless solution, sterilized by filtration.
Formulation
HMGN1 protein is supplied in a solution at a concentration of 0.5 mg/ml. The solution contains 20mM Tris-HCl buffer at pH 8.0, 1mM DTT (reducing agent), 0.1M NaCl (salt), 0.1mM PMSF (protease inhibitor) and 20% glycerol (cryoprotectant).
Stability
For short-term storage (2-4 weeks), the product can be stored at 4°C. For longer storage, it is recommended to freeze the product at -20°C. To ensure maximum stability during long-term storage, adding a carrier protein such as HSA or BSA to a final concentration of 0.1% is advisable. Repeated freezing and thawing of the product should be avoided.
Purity
The purity of the HMGN1 protein is greater than 95% as determined by SDS-PAGE analysis.
Synonyms
HMG14, GC104230, High-Mobility Group Nucleosome Binding Domain 1.
Source
Escherichia Coli.
Amino Acid Sequence
MPKRKVSSAE GAAKEEPKRR SARLSAKPPA KVEAKPKKAA AKDKSSDKKV QTKGKRGAKG KQAEVANQET KEDLPAENGE TKTEESPASD EAGEKEAKSD LEHHHHHH.

Q&A

What is HMGN1 and what are its primary functions in human cells?

HMGN1, also known as HMG14, is a nucleosomal-binding protein that affects the structure and function of chromatin. It binds to the inner side of nucleosomal DNA, thereby altering interactions between DNA and histone octamers. This binding activity is associated with maintaining transcriptionally active chromatin in a unique conformation . HMGN1 is ubiquitously expressed but shows particularly high expression in embryonic tissues, suggesting its importance in developmental processes .

The primary functions of HMGN1 include chromatin binding, nucleosomal DNA binding, and regulation of gene expression. It preferentially localizes to chromatin regulatory sites and to promoters of transcriptionally active genes, allowing it to influence the expression of numerous genes throughout the genome . Additionally, HMGN1 has been shown to inhibit the phosphorylation of nucleosomal histones H3 and H2A by specific kinases, further demonstrating its role in chromatin modification .

HMGN1 expression patterns change significantly during development, with down-regulation occurring throughout the embryo except in committed but continuously renewing cell types undergoing active differentiation. These include the basal layer of the epithelium and kidney cells undergoing mesenchyme to epithelium transition . This dynamic expression pattern highlights HMGN1's role in cellular differentiation processes.

What experimental methods are commonly used to detect and quantify HMGN1 in human samples?

Several established methods are employed to detect and quantify HMGN1 in human samples. Western blotting represents a fundamental approach, typically using a 1:1000 dilution of HMGN1-specific antibodies to detect the protein's characteristic 18 kDa band . For immunohistochemistry applications, HMGN1 antibodies are used at dilutions ranging from 1:50 to 1:500, with positive detection demonstrated in human kidney tissue among other samples .

Immunofluorescence and immunocytochemistry techniques provide spatial information about HMGN1 localization within cells, with recommended antibody dilutions of 1:800 to 1:1600 or 1:200 to 1:500 depending on the specific antibody used . When performing these assays, antigen retrieval is often recommended using TE buffer at pH 9.0 or alternatively with citrate buffer at pH 6.0 to optimize detection .

Quantitative PCR represents another important method for analyzing HMGN1 expression at the transcript level. This approach has been effectively employed in studies examining the regulatory relationship between HMGN1 and MeCP2, demonstrating that HMGN1 functions as a negative regulator of MeCP2 expression . For protein structure analysis, researchers have utilized NMR spectroscopy and circular dichroism to characterize HMGN1's intrinsically disordered regions and conformational changes induced by post-translational modifications .

What are the known interaction partners of HMGN1 in the nucleus?

HMGN1 engages with several key nuclear components, most notably nucleosomal DNA and the histone octamer. The interaction with the nucleosome is particularly important, as HMGN1 binds to the inner side of nucleosomal DNA, altering the DNA-histone octamer interaction . Recent structural biology studies have demonstrated that HMGN1 specifically interacts with the acidic patch of the nucleosome, and this interaction can be modulated by post-translational modifications such as phosphorylation .

The protein also functionally interacts with chromatin modifying enzymes. Research indicates that HMGN1 inhibits the phosphorylation of nucleosomal histones H3 and H2A by kinases RPS6KA5/MSK1 and RPS6KA3/RSK2, suggesting regulatory interactions with these enzymes . Additionally, HMGN1 has been implicated in transcription-coupled repair (TCR) following UV treatment and in the proper activation of ATM, indicating potential interactions with DNA repair machinery .

Genomic profiling studies have revealed that HMGN1 preferentially localizes to chromatin regulatory sites and promoters of actively transcribed genes, suggesting interactions with transcriptional machinery . In the context of CRISPR-based genome editing, HMGN1 has been shown to enhance the efficiency of base editors when fused to Cas9, indicating a functional interaction with the CRISPR-Cas9 system that improves genomic targeting and editing outcomes .

How does phosphorylation impact HMGN1's structure and function?

Phosphorylation of HMGN1 induces significant structural changes that directly impact its functional capabilities. Recent studies employing NMR spectroscopy, circular dichroism, and computational modeling have revealed that serine phosphorylation within the nucleosome binding domain (NBD) of HMGN1 causes local conformational alterations in the peptide backbone . Most notably, this post-translational modification decreases the helical propensity of the NBD, which is critical for proper interaction with nucleosomes .

At the molecular level, advanced modeling studies using AlphaFold3 suggest that phosphorylation specifically disrupts the interface between HMGN1 and the nucleosome acidic patch . This disruption likely reduces HMGN1's binding affinity for nucleosomes, potentially serving as a regulatory mechanism to control HMGN1's chromatin-modifying activities. It's worth noting that computational models tend to over-predict helicity compared to experimental data, highlighting the importance of combining modeling with experimental approaches when studying intrinsically disordered proteins like HMGN1 .

The functional consequences of HMGN1 phosphorylation extend beyond structural changes. As an intrinsically disordered protein, HMGN1 exists in an ensemble of conformational states rather than a single defined structure. Phosphorylation appears to shift this ensemble, potentially altering HMGN1's accessibility to histones and its ability to regulate chromatin structure . This mechanism provides insight into how post-translational modifications might dynamically regulate chromatin-associated proteins to control gene expression in response to cellular signals.

What is the role of HMGN1 in neurodevelopmental disorders, particularly Down Syndrome?

HMGN1's role in neurodevelopmental disorders stems from its genomic location and regulatory functions. The HMGN1 gene is located on chromosome 21 in a segment known as the Down Syndrome critical region (DSCR), which is considered crucial in the etiology of Down Syndrome . Elevated HMGN1 expression has been observed in human Down Syndrome cells and in cells from Ts1Cje mice, which serve as a model for this condition . This overexpression likely contributes to the molecular pathology underlying Down Syndrome.

Mechanistically, HMGN1 has been identified as a negative regulator of methyl CpG-binding protein 2 (MeCP2) expression . MeCP2 is a DNA-binding protein known to affect neurological functions, with mutations or altered levels linked to various neurodevelopmental disorders including Rett syndrome, mental retardation, learning disabilities, and autism spectrum disorders . Quantitative PCR and Western analyses of cell lines and brain tissues from mice that either overexpress or lack HMGN1 have confirmed this regulatory relationship .

The molecular basis for HMGN1's regulation of MeCP2 involves chromatin structure modification. Alterations in HMGN1 levels lead to changes in chromatin structure and histone modifications in the MeCP2 promoter region . Behavioral analyses using multiple assays including open field tests, elevated plus maze, reciprocal social interaction, and automated sociability tests have linked changes in HMGN1 levels to abnormalities in activity and anxiety, as well as social deficits in mice . These findings collectively support the hypothesis that aberrant expression of HMGN1 can contribute to neurodevelopmental disorders through its impact on chromatin structure and gene expression regulation.

How does HMGN1 enhance CRISPR-based genome editing, and what are the mechanisms involved?

HMGN1 significantly enhances CRISPR-directed base editing through its chromatin-associated properties. In a systematic evaluation of chromatin-associated factors, HMGN1 demonstrated a remarkable ability to improve the efficiency of both C-to-G and A-to-G base editing when incorporated into editing constructs . This enhancement effect likely stems from HMGN1's ability to interact with and potentially remodel chromatin structure, making target DNA sequences more accessible to CRISPR machinery.

By fusing HMGN1 to Cas9 along with specific base editors (GBE and ABE), researchers have developed a dual-function A-to-G and C-to-G base editor (GGBE) capable of simultaneous A and C to G conversion with substantially improved editing efficiency . This innovative approach broadens the genome manipulation capacity of CRISPR systems, potentially allowing for more complex editing operations to be performed in a single step.

The mechanisms underlying HMGN1's enhancement effect likely involve its nucleosome-binding capabilities. HMGN1 binds to the inner side of nucleosomal DNA, altering interactions between DNA and histone octamers . This binding may facilitate chromatin opening at target sites, improving accessibility for CRISPR components. Additionally, HMGN1's preferential localization to chromatin regulatory sites and transcriptionally active regions may help guide editing machinery to euchromatic regions, where editing efficiency is typically higher . These properties make HMGN1 a valuable component for improving genome editing technologies, particularly in challenging chromatin contexts.

What are the current approaches for studying HMGN1's intrinsically disordered regions?

Studying the intrinsically disordered regions of HMGN1 requires specialized methodologies that can capture the protein's dynamic conformational ensemble. Nuclear Magnetic Resonance (NMR) spectroscopy represents a gold standard approach, as it can provide atomic-level resolution of protein structure in solution while accounting for conformational heterogeneity . This technique has been successfully employed to study the effects of serine phosphorylation in HMGN1's nucleosome-binding domain, revealing local conformational changes in the peptide backbone upon modification .

Circular dichroism (CD) spectroscopy offers complementary insights by quantifying secondary structure content across the protein population. In HMGN1 research, CD has been instrumental in demonstrating that phosphorylation decreases the helical propensity of the nucleosome binding domain . This finding is particularly important given that helical structure is often induced upon binding to target molecules in intrinsically disordered proteins.

Biochemical approaches such as limited proteolysis, cross-linking mass spectrometry, and hydrogen-deuterium exchange can provide additional insights into HMGN1's structural dynamics and interaction surfaces. Together, these diverse methodologies create a comprehensive toolkit that allows researchers to characterize the conformational landscape of HMGN1 and how it changes in response to post-translational modifications or binding partners.

What are the best antibody-based detection methods for studying HMGN1 expression and localization?

For Western blotting applications, polyclonal antibodies raised against synthetic peptides corresponding to residues surrounding Val65 of human HMGN1 protein have demonstrated high specificity and sensitivity . These antibodies typically perform optimally at a 1:1000 dilution for detecting endogenous HMGN1 levels, which appears as an 18 kDa band . When selecting antibodies, researchers should prioritize those that do not cross-react with other HMGN family members (HMGN2, HMGN3, HMGN4), ensuring signal specificity .

For immunohistochemistry applications, HMGN1 antibodies can be used at dilutions ranging from 1:50 to 1:500, with antigen retrieval recommended using TE buffer at pH 9.0 or alternatively with citrate buffer at pH 6.0 . Positive staining has been consistently observed in human and mouse kidney tissues, making these useful positive controls for protocol optimization . For immunofluorescence and immunocytochemistry, dilutions between 1:200-1:1600 are recommended depending on the specific antibody and cell type being examined .

When studying subcellular localization, it's important to note that HMGN1 is predominantly nuclear with enrichment at chromatin regulatory sites and transcriptionally active regions . Therefore, co-staining with nuclear markers and potentially with markers of active transcription can provide valuable contextual information. For multi-parameter analyses, antibodies compatible with specific epitope tags may be used with recombinant HMGN1 constructs to distinguish between endogenous and exogenous protein or to facilitate co-immunoprecipitation studies examining HMGN1's interaction partners.

How can researchers effectively modulate HMGN1 expression to study its function?

Several complementary approaches can be employed to modulate HMGN1 expression levels for functional studies. For gene knockdown, RNA interference using siRNA or shRNA targeting HMGN1 transcripts has proven effective. This approach is particularly useful for transient reduction of HMGN1 levels to assess immediate functional consequences without allowing for potential compensatory mechanisms that might develop in complete knockout systems .

For overexpression studies, transfection or viral transduction of HMGN1 expression constructs can be utilized. It's worth noting that overexpression of HMGN1 has been shown to inhibit myotube formation in C2C12 myoblast cells and chondrocyte differentiation in primary limb bud mesenchymal cells, suggesting a role in blocking cellular differentiation . When designing overexpression constructs, researchers should consider using inducible promoters to control the timing and level of expression, as HMGN1's effects may be dose-dependent.

For studies specifically examining the effects of HMGN1 in Down Syndrome, the Ts1Cje mouse model, which exhibits elevated HMGN1 expression similar to human Down Syndrome cells, provides a valuable experimental system . Additionally, transgenic mice that either overexpress or lack HMGN1 have been used effectively to study its role in regulating MeCP2 expression and associated behavioral phenotypes .

What experimental approaches can be used to study HMGN1's interactions with chromatin?

Chromatin immunoprecipitation (ChIP) represents a fundamental technique for studying HMGN1's interactions with chromatin. This approach has revealed that HMGN1 preferentially localizes to chromatin regulatory sites and to promoters of transcriptionally active genes . ChIP-seq extends this methodology to provide genome-wide mapping of HMGN1 binding sites, offering comprehensive insights into its distribution across the chromatin landscape.

Biochemical nucleosome binding assays using purified components provide a reductionist approach to studying HMGN1-nucleosome interactions. These assays can be combined with site-directed mutagenesis or phosphomimetic mutations to assess how specific residues or post-translational modifications affect binding affinity and specificity . Electrophoretic mobility shift assays (EMSA) and microscale thermophoresis represent complementary techniques for quantifying binding parameters.

For structural studies of HMGN1-nucleosome complexes, cryo-electron microscopy and X-ray crystallography have been employed, though these approaches face challenges due to HMGN1's intrinsically disordered nature. NMR spectroscopy offers advantages for studying dynamic interactions, as demonstrated in research examining how phosphorylation of HMGN1's nucleosome binding domain affects its interaction with the nucleosome acidic patch .

In cellular contexts, proximity ligation assays can detect HMGN1-chromatin interactions with spatial resolution. Fluorescence recovery after photobleaching (FRAP) provides insights into the dynamics of these interactions by measuring HMGN1's residence time on chromatin. Additionally, Assay for Transposase-Accessible Chromatin with sequencing (ATAC-seq) can be used to assess how modulation of HMGN1 levels affects chromatin accessibility genome-wide, providing functional insights into its chromatin-modifying activities.

How does altered HMGN1 expression contribute to pathology in Down Syndrome?

Elevated HMGN1 expression is a consistent feature observed in human Down Syndrome cells and in the Ts1Cje mouse model of Down Syndrome . This overexpression stems from the genomic location of the HMGN1 gene on chromosome 21, specifically within the Down Syndrome critical region (DSCR) . The increased dosage of HMGN1 contributes to the complex pathology of Down Syndrome through several molecular mechanisms that affect neurodevelopment and cellular function.

A primary mechanism involves HMGN1's regulation of MeCP2 expression. As a negative regulator of MeCP2, elevated levels of HMGN1 in Down Syndrome lead to altered MeCP2 expression . This dysregulation is significant because MeCP2 plays crucial roles in neurological functions, with mutations or altered levels linked to various neurodevelopmental disorders . Quantitative analyses of mouse models have confirmed that HMGN1 levels influence chromatin structure and histone modifications at the MeCP2 promoter, directly affecting its expression .

Behavioral analyses further support HMGN1's role in Down Syndrome pathology. Studies using various behavioral assays, including open field tests, elevated plus maze, reciprocal social interaction, and automated sociability tests, have linked changes in HMGN1 levels to abnormalities in activity, anxiety, and social behavior in mice . These phenotypes parallel certain neurobehavioral aspects of Down Syndrome, suggesting that HMGN1 overexpression contributes to the cognitive and behavioral manifestations of the condition.

Additionally, HMGN1's broad effects on chromatin structure and transcriptional regulation likely contribute to the global gene expression changes observed in Down Syndrome. By altering nucleosome dynamics and affecting the accessibility of transcription machinery to chromatin, elevated HMGN1 may disrupt the precise temporal and spatial gene expression patterns required for normal neurodevelopment, contributing to the characteristic features of Down Syndrome.

What is the potential of HMGN1 as a therapeutic target in genetic and neurological disorders?

HMGN1's involvement in chromatin regulation and its links to neurodevelopmental disorders position it as a potential therapeutic target. For Down Syndrome, normalizing HMGN1 levels could potentially ameliorate some neurological features by restoring proper MeCP2 expression and downstream gene regulation . RNA interference approaches or small molecule inhibitors that modulate HMGN1 activity could be explored as potential therapeutic strategies in this context.

In the field of genome editing, HMGN1's ability to enhance CRISPR-directed base editing efficiency presents translational opportunities . The development of the dual-function A-to-G and C-to-G base editor (GGBE) incorporating HMGN1 demonstrates improved editing capacity that could be valuable for correcting disease-causing mutations . This application highlights HMGN1's potential utility in therapeutic genome editing approaches for genetic disorders.

For targeting HMGN1 itself, understanding its post-translational modifications provides additional therapeutic avenues. Research showing that phosphorylation of HMGN1's nucleosome binding domain decreases its helicity and disrupts interaction with the nucleosome acidic patch suggests that modulating these modifications could alter HMGN1's chromatin-binding properties . Small molecules that either promote or inhibit specific phosphorylation events could potentially be developed as tools to fine-tune HMGN1 activity in pathological contexts.

The development of therapeutic approaches targeting HMGN1 would benefit from further research into its tissue-specific functions and redundancy with other HMGN family members. While HMGN1-/- mice appear largely normal under standard conditions due to potential compensation by proteins like HMGN2, they show hypersensitivity to various stress conditions . This observation suggests that therapeutic modulation of HMGN1 might be most effective under specific stress conditions or in combination with approaches targeting redundant pathways.

How can HMGN1 be utilized to improve modern genome editing technologies?

HMGN1 has demonstrated significant potential for enhancing CRISPR-based genome editing technologies through its chromatin-modifying properties. When fused to Cas9 along with specific base editors, HMGN1 substantially improves editing efficiency for both C-to-G and A-to-G conversions . This enhancement capability has led to the development of a dual-function base editor (GGBE) that can perform simultaneous A and C to G conversions with high efficiency .

The mechanisms underlying HMGN1's enhancement effect likely involve its ability to bind nucleosomal DNA and alter chromatin structure, making target sequences more accessible to editing machinery . This property is particularly valuable for genome editing applications targeting regions with complex chromatin structures or heterochromatic domains, which typically present challenges for standard editing approaches. By incorporating HMGN1 into editing constructs, researchers may overcome some of these accessibility barriers.

Future developments in this area could include optimizing HMGN1 fusion constructs for specific chromatin contexts or cell types. Since HMGN1 expression is tightly linked to cellular differentiation, with varying levels across different tissues and developmental stages , tailored variants might be designed for particular applications. Additionally, combining HMGN1 with other chromatin-modifying factors could potentially create synergistic effects that further enhance editing efficiency or specificity.

Another promising direction involves utilizing HMGN1's preferential localization to chromatin regulatory sites and transcriptionally active regions to guide editing machinery to specific genomic domains. This approach could potentially improve the targeting of therapeutic editing to disease-relevant loci while minimizing off-target effects in other regions of the genome. Such applications would particularly benefit from further research into the sequence and structural determinants of HMGN1's chromatin binding preferences.

What is the role of HMGN1 in cellular stress responses and DNA repair pathways?

HMGN1 plays significant roles in cellular stress responses, particularly following exposure to DNA-damaging agents. Studies with HMGN1-/- mice have revealed hypersensitivity to various stress conditions, including UV light and ionizing radiation (IR) . This hypersensitivity phenotype points to HMGN1's importance in orchestrating appropriate cellular responses to genotoxic stress.

At the molecular level, HMGN1 is required for efficient transcription-coupled repair (TCR) following UV treatment . This specialized DNA repair pathway preferentially removes lesions from actively transcribed regions of the genome, suggesting that HMGN1 helps maintain genomic integrity in transcriptionally active chromatin. Additionally, HMGN1 contributes to proper activation of ATM, a key kinase in the DNA damage response signaling cascade . These functions position HMGN1 as an important factor in the interface between chromatin structure, transcription, and DNA repair.

HMGN1's Gene Ontology annotations specifically include involvement in DNA repair pathways, with particular connections to Transcription-Coupled Nucleotide Excision Repair (TC-NER) . This specialized repair pathway removes bulky DNA lesions from the transcribed strand of active genes, ensuring that transcription can proceed without errors caused by DNA damage. HMGN1's ability to alter nucleosome structure may facilitate access of repair machinery to damaged DNA sites within the context of chromatin.

Future research in this area could explore how HMGN1's post-translational modifications change in response to different types of cellular stress. The finding that phosphorylation affects HMGN1's interaction with the nucleosome acidic patch suggests a potential regulatory mechanism that might be modulated during stress responses. Additionally, investigating how HMGN1 coordinates with other chromatin-associated factors and repair proteins could provide insights into the complex choreography of DNA repair within the nuclear environment.

Product Science Overview

Introduction

High-Mobility Group Nucleosome Binding Domain 1 (HMGN1) is a protein encoded by the HMGN1 gene in humans. This protein is part of the high-mobility group (HMG) family, which is known for its role in chromatin structure and function. HMGN1 is particularly notable for its ability to bind nucleosomal DNA and influence chromatin dynamics.

Structure and Function

HMGN1 binds to the inner side of nucleosomal DNA, altering the interaction between DNA and the histone octamer . This binding is crucial for maintaining an open chromatin configuration around transcribable genes, thereby facilitating gene expression . HMGN1 also inhibits the phosphorylation of nucleosomal histones H3 and H2A by kinases such as RPS6KA5/MSK1 and RPS6KA3/RSK2 .

Biological Role

HMGN1 plays a significant role in various cellular processes, including:

  • Chromatin Remodeling: HMGN1 is involved in chromatin decompaction, which is essential for active transcription .
  • Immune Response: HMGN1 acts as an alarmin, a type of endogenous molecule that signals tissue and cell damage. It is critical for lipopolysaccharide-induced immune responses and promotes the recruitment and activation of antigen-presenting cells (APCs) such as dendritic cells .
  • Gene Expression: By maintaining an open chromatin state, HMGN1 ensures that genes remain in a transcribable conformation, thus facilitating efficient gene expression .
Clinical Significance

Mutations or dysregulation of HMGN1 have been associated with various diseases, including:

  • Van Der Woude Syndrome: A genetic disorder characterized by cleft lip and/or cleft palate .
  • Ossification of the Posterior Longitudinal Ligament of the Spine: A condition where the ligament in the spine becomes ossified, leading to spinal stenosis .
Research and Applications

Recombinant HMGN1 is used in various research applications to study its role in chromatin dynamics and immune responses. Understanding the function of HMGN1 can provide insights into the mechanisms of gene regulation and the development of therapeutic strategies for related diseases.

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.