HMGB2 belongs to the high-mobility group (HMG) protein family, serving as a non-histone nuclear protein containing HMG-box motifs. It functions primarily as an architectural facilitator in nucleoprotein complex assembly by bending DNA and modulating the association of transcription factors to their DNA-binding sites . In human cells, HMGB2:
Acts upstream of OCT4/SOX2 signaling to regulate stem cell pluripotency
Facilitates the recruitment of transcription factors to specific genomic sites
Interacts with several proteins including casein kinase 1α and tumor suppressor p53
Influences mesenchymal stem cell differentiation and erythroid differentiation
HMGB2 expression is developmentally regulated, with high expression in undifferentiated stem cells and significant decrease during cellular differentiation processes .
HMGB2 expression is regulated through multiple mechanisms:
Transcriptional regulation: The nuclear receptor small heterodimer partner (SHP) and transcription factor E2F1 control HMGB2 mRNA expression .
Post-transcriptional regulation: MicroRNA-127 (miR-127) acts as a translational repressor of HMGB2 protein expression by targeting its 3′ untranslated region . This creates a negative feedback loop as miR-127 is upregulated during differentiation processes.
Protein-protein interactions: HMGB2 interacts with various proteins that can affect its stability and function, including its interaction with OCT4 which enhances OCT4 and SOX2 protein expression and transcriptional activities .
Post-translational modifications: Similar to other HMG proteins, HMGB2 can undergo modifications that affect its function and cellular localization.
For effective analysis of HMGB2 in tissue samples, researchers should consider the following methodological approaches:
Immunohistochemistry (IHC): Effective for detecting HMGB2 protein levels in patient tissues, as demonstrated in studies of glioblastoma patients where IHC was used to measure HMGB2 protein levels in 51 GBM patients .
Quantitative RT-PCR: For measuring HMGB2 mRNA levels, as shown in studies of IgA nephropathy where qRT-PCR was used to measure HMGB2 expression .
Western blotting: For protein quantification and validation of HMGB2 expression patterns across different tissues or under different experimental conditions .
Single-cell RNA sequencing (scRNA-seq): For evaluating HMGB2 expression patterns at single-cell resolution, particularly valuable in heterogeneous tissues such as tumors .
Spatial transcriptomics (ST): For analyzing the spatial distribution of HMGB2 expression within tissue architecture, providing insights into its role in the tissue microenvironment .
HMGB2 contributes to cancer progression through multiple molecular mechanisms:
Regulation of cell viability and invasion: In glioblastoma multiforme (GBM), HMGB2 knockdown decreases cell viability and invasion in vitro and reduces tumor volume in vivo . This effect involves changes in p53 expression and alterations in the balance of matrix metalloproteinase 2 (MMP2) and tissue inhibitors of metalloproteinase 2 (TIMP2) .
Modulation of chemotherapeutic sensitivity: Silencing HMGB2 significantly increases the sensitivity of GBM cells to temozolomide chemotherapy, suggesting its role in chemoresistance .
Promotion of stem-like properties: In liver cancer, HMGB2 is markedly induced in tumor-initiating cells, and diminishing HMGB2 expression impairs spheroid formation, indicating its role in maintaining cancer stem cell properties .
Immunomodulatory effects: HMGB2 shapes the tumor microenvironment in hepatocellular carcinoma (HCC), correlating with an immunosuppressive microenvironment, particularly evident in exhausted T cells .
Association with metastasis: HMGB2 is significantly upregulated in HCC transitional subgroups associated with aggressive metastasis .
Elevated HMGB2 expression has been observed in multiple human cancers, including colon adenocarcinoma, head and neck squamous cell carcinoma, uterine corpus endometrial carcinoma, and breast cancers, and is associated with lower patient survival rates .
Several emerging therapeutic strategies targeting HMGB2 are being investigated:
Small molecule inhibitors: Compounds like inflachromene (ICM) have been identified to inhibit HMGB proteins by perturbing their post-translational modifications. Treatment with ICM decreases OCT4 and SOX2 expression and facilitates stem cell differentiation .
RNA interference approaches:
microRNA-based therapies: Ectopic expression of miR-127, which targets HMGB2, decreases HMGB2 expression and facilitates differentiation of embryonic stem cells .
Modulation of SHP expression: The nuclear receptor SHP suppresses HMGB2 messenger RNA expression, providing another potential therapeutic avenue .
Immunotherapy combinations: Given HMGB2's role in shaping the immunosuppressive tumor microenvironment, combining HMGB2 inhibition with immune checkpoint inhibitors represents a promising strategy, particularly in HCC .
HMGB2 plays a crucial role in stem cell pluripotency through the following mechanisms:
Interaction with core pluripotency factors: HMGB2 interacts with OCT4, increases protein expression of OCT4 and SOX2, and enhances their transcriptional activities, placing HMGB2 upstream of OCT4/SOX2 signaling in controlling embryonic stem cell pluripotency .
Developmental regulation: HMGB2 protein is highly expressed in undifferentiated cells (similar to OCT4 and SOX2) but undergoes rapid decline during embryonic body formation .
Response to differentiation cues: HMGB2 expression decreases during differentiation processes, with concomitant increases in its negative regulators miR-127 and SHP .
Experimental manipulation approaches:
Genetic modulation:
Knockdown using shRNA or siRNA
Overexpression using expression vectors
CRISPR/Cas9-mediated genome editing to modify HMGB2 or its regulatory elements
Pharmacological inhibition: Using small molecule inhibitors like inflachromene to disrupt HMGB2 function .
Indirect regulation:
Differentiation protocols: Monitoring and manipulating HMGB2 levels during directed differentiation of stem cells to understand its role in lineage specification.
HMGB2 contributes to IgA nephropathy (IgAN) through the following mechanisms:
Increased expression in disease state: HMGB2 expression is significantly greater in IgAN patients compared to healthy controls .
Regulation of APRIL expression: HMGB2 promotes proliferation-inducing ligand (APRIL) expression, which is an important cause of glycosylation deficiency of IgA1 (Gd-IgA1), a key trigger of IgAN .
Interaction with HMGA1:
Effect on disease progression: The HMGB2-HMGA1-APRIL axis ultimately leads to Gd-IgA1 overexpression, which is central to IgAN pathogenesis .
Experimental evidence supports this pathway:
Knockdown of HMGB2 downregulates HMGA1, APRIL, and Gd-IgA1 expression
Knockdown of HMGA1 reduces Gd-IgA1 concentration in cell supernatant
Co-immunoprecipitation confirms the physical interaction between HMGB2 and HMGA1
HMGB2 plays a significant role in shaping the tumor microenvironment (TME) in hepatocellular carcinoma (HCC):
Association with aggressive phenotype: HMGB2 is significantly upregulated in HCC transitional subgroups associated with aggressive metastasis .
Correlation with immunosuppression: HMGB2 expression positively correlates with an immunosuppressive microenvironment in HCC . This is particularly evident in:
Exhausted T cells
Expression of immunosuppressive markers
Spatial expression patterns: Spatial transcriptomics and multiplex immunohistochemistry have validated the specific spatial expression patterns of HMGB2 within the TME, providing evidence of its role in HCC progression and immune evasion .
Impact on prognosis: HMGB2 expression correlates with poor prognosis in HCC patients across multiple cohorts, suggesting its role as a prognostic biomarker .
Potential impact on immunotherapy response: The association between HMGB2 and immunosuppression suggests it may influence response to immune checkpoint inhibitors, which is particularly relevant given the importance of immunotherapy in HCC management .
The choice of experimental model depends on the specific aspect of HMGB2 function being investigated:
Cell line models:
Embryonic stem cells: CGR8 cells have been used effectively to study HMGB2's role in pluripotency .
Cancer cell lines: Multiple GBM cell lines have been employed for invasion and viability studies related to HMGB2 .
B cells: Important for studying HMGB2's role in immune-related conditions like IgA nephropathy .
Animal models:
Patient-derived samples:
Primary cells: Peripheral blood mononuclear cells from IgAN patients have been used to study HMGB2's role in disease progression .
Tissue microarrays: Used for large-scale analysis of HMGB2 expression across patient cohorts.
Patient-derived xenografts: To maintain tumor heterogeneity while studying HMGB2 function.
Advanced techniques:
Organoids: Provide three-dimensional culture systems that better recapitulate in vivo conditions.
Single-cell analysis platforms: For studying heterogeneity in HMGB2 expression and function across cell populations .
Spatial omics approaches: To understand HMGB2's role in the tissue microenvironment .
Distinguishing between HMGB2 expression and its functional activity requires multiple complementary approaches:
Expression measurement:
mRNA quantification: qRT-PCR for HMGB2 transcript levels
Protein quantification: Western blotting, ELISA, or immunohistochemistry for HMGB2 protein levels
Single-cell approaches: scRNA-seq for cell-specific expression patterns
Activity assessment:
DNA-binding assays: Electrophoretic mobility shift assays (EMSA) to assess HMGB2 binding to DNA
Chromatin immunoprecipitation (ChIP): To identify genomic regions bound by HMGB2
Transcriptional reporter assays: Using constructs containing HMGB2-responsive elements
Interaction studies:
Functional readouts:
Pluripotency markers: OCT4/SOX2 expression levels as downstream readouts of HMGB2 activity in stem cells
Invasion assays: Transwell and wound-healing assays to assess the impact of HMGB2 on cell invasion
Differentiation assays: EB formation to assess HMGB2's impact on differentiation potential
Drug sensitivity tests: To evaluate HMGB2's influence on chemotherapeutic resistance
Dynamics and localization:
Live-cell imaging: With fluorescently tagged HMGB2 to track localization and dynamics
Subcellular fractionation: To assess distribution between nuclear and cytoplasmic compartments
Immunofluorescence microscopy: For spatial localization of HMGB2 within cells and tissues
When designing experiments to study HMGB2, researchers must consider several context-specific factors:
Cancer research considerations:
Heterogeneity issues:
Microenvironment factors:
Therapeutic relevance:
Clinical correlation:
Developmental biology considerations:
Temporal dynamics:
Lineage specificity:
Assess HMGB2's role across different lineages (ectoderm, mesoderm, endoderm)
Use lineage tracing approaches to track cell fate decisions
Compare HMGB2 function in different stem cell types (embryonic, adult, induced)
Interaction with core pluripotency network:
Physiological relevance:
Maintain physiologically relevant culture conditions
Consider the impact of oxygen tension on HMGB2 function
Validate in vitro findings in appropriate in vivo models
Common methodological considerations:
Controls and validation:
Use multiple HMGB2 modulation approaches (siRNA, CRISPR, inhibitors)
Include rescue experiments to confirm specificity
Employ positive and negative controls for all assays
Dose and timing:
Establish dose-response relationships for inhibitors or activators
Consider temporal aspects of HMGB2 manipulation
Account for potential compensatory mechanisms
Regulatory mechanisms:
Several promising research directions for HMGB2 in human disease warrant further investigation:
HMGB2 as a therapeutic target:
Development of selective HMGB2 inhibitors with improved pharmacokinetic properties
Evaluation of combination therapies targeting HMGB2 alongside standard treatments
Investigation of tissue-specific delivery methods for HMGB2-targeting therapeutics
Biomarker development:
Immunomodulatory roles:
Broader disease associations:
Expansion of studies beyond cancer and IgA nephropathy to other inflammatory and autoimmune conditions
Investigation of HMGB2's role in aging-related diseases
Examination of potential involvement in metabolic disorders
Mechanistic insights:
Detailed structural studies of HMGB2 interactions with binding partners
Comprehensive mapping of HMGB2 genomic binding sites across cell types
Investigation of post-translational modifications that regulate HMGB2 function
Single-cell and spatial transcriptomics offer powerful approaches to advance HMGB2 research:
Cellular heterogeneity insights:
Identification of cell populations with differential HMGB2 expression
Characterization of cell type-specific HMGB2 regulatory networks
Detection of rare cell populations with unique HMGB2-associated properties
Spatial context advantages:
Mapping of HMGB2 expression patterns within tissue architecture
Identification of spatial relationships between HMGB2-expressing cells and other cell types
Correlation of HMGB2 expression with tissue microenvironmental features
Disease progression understanding:
Tracking changes in HMGB2 expression during disease evolution
Identifying cellular transitions associated with HMGB2 regulation
Mapping the spatial reorganization of HMGB2-expressing cells during pathogenesis
Therapeutic response monitoring:
Assessment of cell type-specific responses to HMGB2-targeting therapies
Identification of resistance mechanisms at single-cell resolution
Spatial mapping of therapy-induced changes in the tumor microenvironment
Technical integration approaches:
Combined single-cell RNA-seq with ATAC-seq to link HMGB2 expression to chromatin accessibility
Integration of spatial transcriptomics with multiplex protein imaging for multi-omic insights
Computational methods to infer cell-cell communication networks involving HMGB2
As demonstrated in recent HCC research, these techniques have already provided valuable insights into HMGB2's role in shaping the tumor microenvironment and its association with immunosuppressive features .
Several important contradictions and knowledge gaps exist in HMGB2 research:
Contextual function paradoxes:
Regulatory mechanism uncertainties:
The relative contributions of transcriptional regulation (via SHP) versus post-transcriptional regulation (via miR-127) in different contexts remain unclear
The triggers that initiate HMGB2 downregulation during normal differentiation versus disease intervention are not fully understood
Further investigation of upstream regulators and their tissue specificity is needed
Therapeutic targeting challenges:
Given HMGB2's role in normal stem cell function, the potential for off-target effects of HMGB2 inhibition requires careful assessment
The optimal timing and context for therapeutic intervention targeting HMGB2 remain undefined
Development of strategies to achieve selective inhibition in disease contexts while sparing normal function is needed
Interaction with other HMGB family members:
The potential functional redundancy or compensation between HMGB1, HMGB2, and other family members is not fully characterized
The unique versus overlapping functions of HMGB family proteins require further delineation
Understanding of how these proteins interact or compensate for each other during development and disease is incomplete
Technical limitations:
Current antibodies may have cross-reactivity issues between HMGB family members
Challenges in distinguishing nuclear versus cytoplasmic functions of HMGB2
Limited availability of specific HMGB2 inhibitors hampers functional studies
HMGB2 contains two basic DNA-binding domains, known as HMG-box A and B, and a C-terminal acidic tail . This structure allows HMGB2 to bind to non-canonical DNA structures such as single-stranded DNA, enhancing DNA flexibility by looping. This mechanism promotes activities on various gene promoters by enhancing transcription factor binding and/or bringing distant regulatory sequences into close proximity .
HMGB2 plays multiple roles in different cellular compartments. In the nucleus, it is involved in transcription, chromatin remodeling, and V(D)J recombination . It acts as a cofactor of the RAG complex, stimulating cleavage and RAG protein binding at the 23 bp spacer of conserved recombination signal sequences (RSS) . Additionally, HMGB2 is involved in the final ligation step in DNA end-joining processes of DNA double-strand breaks repair and V(D)J recombination .
Unlike HMGB1, which is ubiquitously expressed, HMGB2 expression is restricted mainly to lymphoid organs and testes, although it is widely expressed during embryogenesis . In human aortic smooth muscle cells, HMGB2 can enhance cell proliferation and migration by inducing reactive oxygen species . In vivo and in vitro analyses indicate that HMGB2 loss in cardiomyocytes causes hypertrophy and increased expression of genes responsible for hypertrophic cell growth .
HMGB2 has been associated with various diseases. For instance, it is linked to ceroid lipofuscinosis, neuronal, 6A . Studies have shown that HMGB2 plays a role in germ cell differentiation, as hmgb2−/− mice exhibit reduced fertility . Additionally, these mice possess reduced superficial zone cellularity and accelerated development of osteoarthritis .
Recombinant HMGB2 is produced using genetic engineering techniques, where the HMGB2 gene is cloned and expressed in suitable host cells. This allows for the production of large quantities of HMGB2 protein for research and therapeutic purposes. Recombinant HMGB2 retains the functional properties of the native protein, making it a valuable tool for studying its biological roles and potential therapeutic applications.