Recombinant Human SWI/SNF-related matrix-associated actin-dependent regulator of chromatin subfamily D member 3 (SMARCD3)

Shipped with Ice Packs
In Stock

Description

General Information

Recombinant Human SWI/SNF-related matrix-associated actin-dependent regulator of chromatin subfamily D member 3 (SMARCD3), also known as BAF60C, CRACD3, or Rsc6p, is a protein-coding gene located on chromosome 7 . SMARCD3 is a subunit of the SWI/SNF complex, which is involved in chromatin remodeling and the regulation of gene expression .

Function

SMARCD3 plays a crucial role in regulating gene expression by remodeling chromatin, the complex of DNA and proteins that make up chromosomes . It alters the structure of nucleosomes, the basic units of chromatin, in an ATP-dependent manner, making genes more or less accessible to cellular machinery . SMARCD3 is a component of several SWI/SNF chromatin remodeling complexes, including the neural progenitors-specific complex (npBAF) and the neuron-specific complex (nBAF) . During neural development, SMARCD3 is involved in the transition from npBAF to nBAF complexes, which is essential for the differentiation of neural stem cells into neurons. SMARCD3 also plays a role in regulating the activity of genes involved in dendrite growth .

Role in Cancer

SMARCD3 has been implicated in various cancers, with its role varying depending on the cancer type.

  • Pancreatic Cancer: SMARCD3 is amplified in pancreatic cancer and enriched in pancreatic cancer stem cells, and it is upregulated in human disease . It is required for the growth of aggressive cancer stem cells and regulates the epigenetic landscape and BAF complex binding at FOXA1 binding sites in mouse pancreatic cancer cells . SMARCD3 controls the landscape of lipid metabolism in pancreatic cancer cells .

  • Breast Cancer: SMARCD3 expression is associated with hormone-positive (ER+) breast cancer . Lower proliferation rates are observed in SMARCD3-depleted cells, reflecting a failure to fully progress through G2/M and an increase in endoreplication . In the absence of SMARCD3, p21 accumulates in cells, but does not halt the cell cycle, and DNA damage accumulates and remains unrepaired .

  • Medulloblastoma (MB): Increased SMARCD3 expression activates Reelin–DAB1-mediated Src kinase signaling, which results in an MB response to Src inhibition . Higher levels of SMARCD3 expression are significantly correlated with poorer prognosis of patients with MB across all subgroups, independent of age and sex .

  • Lung Adenocarcinoma (LUAD): SMARCD3 expression is low in LUAD cancer tissues compared to normal tissues .

  • Other Cancers: SMARCD3 is a risk factor for COAD, HNSC, mesothelioma (MESO), STAD, UCEC, and UVM and a protective factor against PAAD .

Involvement in Other Diseases

SMARCD3 is associated with several diseases, including endometrial cancer, urinary bladder cancer, Mobius syndrome, achalasia-alacrima syndrome, and left ventricular noncompaction .

SMARCD3 and SWI/SNF Complex

SMARCD3 is one of the mutually exclusive, tissue-specific SMARCD1/2/3 subunits (also known as Baf60a/b/c) within the SWI/SNF complex . These subunits are only partially redundant, as they cannot complement each other . SMARCD3 is a recruiter of the SWI/SNF complex to target sites on the chromatin through interaction with protein partners .

SMARCD3 in Gene Regulation

SMARCD3 regulates the epigenetic landscape and BAF complex binding . It is predominantly incorporated into the more abundant BAF complex and, to some extent, the PBAF complex . SMARCD3 coordinates FOXA1/BAF activity at a subset of sites, controlling downstream transcriptional programs with diverse functions, including extracellular matrix organization, glycosylation, and immune signaling .

Prognostic and Biomarker Potential

SMARCD3 might act as a tumor suppressor through the regulation of cell cycle checkpoints and could be a reliable and specific breast cancer prognostic biomarker . In ER+ breast cancer patients, low SMARCD3-expressing tumors exhibit reduced survival rates compared to patients expressing normal or higher levels of SMARCD3 .

Tables Summarizing Research Findings

DiseaseSMARCD3 RolePrognostic/Biomarker Potential
Pancreatic CancerAmplified, enriched in cancer stem cells, upregulated in human disease, regulates epigenetic landscapePotential therapeutic target
Breast Cancer (ER+)Associated with hormone-positive breast cancer, affects cell cycle progression, DNA damage repairReliable and specific prognostic biomarker; low expression correlated with reduced survival rates
MedulloblastomaIncreased expression activates Reelin–DAB1-mediated Src kinase signalingHigh expression correlated with poorer prognosis
Lung AdenocarcinomaLow expression in cancer tissuesPotential diagnostic marker
COAD, HNSC, MESO, STAD, UCEC, UVMRisk factorPrognostic indicator
PAADProtective factorPrognostic indicator

Product Specs

Form
Lyophilized powder
Note: We will prioritize shipping the format currently in stock. However, if you have specific format requirements, please indicate them during order placement. We will accommodate your request to the best of our ability.
Lead Time
Delivery times may vary depending on the purchase method and location. Please contact your local distributor for precise delivery estimates.
Note: All proteins are shipped with standard blue ice packs unless otherwise requested. Dry ice shipping requires advance notice and incurs additional charges.
Notes
Avoid repeated freeze-thaw cycles. Store working aliquots at 4°C for up to one week.
Reconstitution
Centrifuge the vial briefly before opening to settle the contents. Reconstitute the protein in sterile, deionized water to a concentration of 0.1-1.0 mg/mL. We recommend adding 5-50% glycerol (final concentration) and aliquoting for long-term storage at -20°C/-80°C. Our default glycerol concentration is 50%, which can serve as a guideline for your reconstitution.
Shelf Life
Shelf life depends on several factors: storage conditions, buffer composition, temperature, and inherent protein stability. Generally, liquid formulations have a 6-month shelf life at -20°C/-80°C, while lyophilized forms have a 12-month shelf life at -20°C/-80°C.
Storage Condition
Upon receipt, store at -20°C/-80°C. Aliquoting is recommended for multiple uses. Avoid repeated freeze-thaw cycles.
Tag Info
Tag type is determined during the manufacturing process.
The tag type will be determined during production. If you require a specific tag, please inform us, and we will prioritize its development.
Synonyms
60 kDa BRG 1/Brm associated factor subunit C; 60 kDa BRG-1/Brm-associated factor subunit C; BAF60C; BRG1 associated factor 60C; BRG1-associated factor 60C; Chromatin remodeling complex BAF60C subunit; CRACD3; Mammalian chromatin remodeling complex BRG1 associated factor 60C; MGC111010; Rsc6p; Smarcd3; SMRD3_HUMAN; SWI/SNF complex 60 kDa subunit C; SWI/SNF related matrix associated actin dependent regulator of chromatin subfamily d member 3; SWI/SNF related, matrix associated, actin dependent regulator of chromatin, subfamily d, member 3, isoform 1; SWI/SNF-related matrix-associated actin-dependent regulator of chromatin subfamily D member 3; Swp73 like protein
Buffer Before Lyophilization
Tris/PBS-based buffer, 6% Trehalose.
Datasheet
Please contact us to get it.
Expression Region
2-483
Protein Length
Full Length of Mature Protein
Purity
>85% (SDS-PAGE)
Species
Homo sapiens (Human)
Target Names
SMARCD3
Target Protein Sequence
AADEVAGGA RKATKSKLFE FLVHGVRPGM PSGARMPHQG APMGPPGSPY MGSPAVRPGL APAGMEPARK RAAPPPGQSQ AQSQGQPVPT APARSRSAKR RKMADKILPQ RIRELVPESQ AYMDLLAFER KLDQTIMRKR VDIQEALKRP MKQKRKLRLY ISNTFNPAKP DAEDSDGSIA SWELRVEGKL LDDPSKQKRK FSSFFKSLVI ELDKDLYGPD NHLVEWHRTP TTQETDGFQV KRPGDLSVRC TLLLMLDYQP PQFKLDPRLA RLLGLHTQSR SAIVQALWQY VKTNRLQDSH DKEYINGDKY FQQIFDCPRL KFSEIPQRLT ALLLPPDPIV INHVISVDPS DQKKTACYDI DVEVEEPLKG QMSSFLLSTA NQQEISALDS KIHETIESIN QLKIQRDFML SFSRDPKGYV QDLLRSQSRD LKVMTDVAGN PEEERRAEFY HQPWSQEAVS RYFYCKIQQR RQELEQSLVV RNT
Uniprot No.

Target Background

Function
Recombinant Human SWI/SNF-related matrix-associated actin-dependent regulator of chromatin subfamily D member 3 (SMARCD3) is involved in transcriptional activation and repression of specific genes through chromatin remodeling (altering DNA-nucleosome topology). It is a component of SWI/SNF chromatin remodeling complexes, which perform crucial enzymatic activities, modifying chromatin structure by altering DNA-histone interactions within nucleosomes in an ATP-dependent manner. SMARCD3 stimulates nuclear receptor-mediated transcription and is part of both the neural progenitor-specific chromatin remodeling complex (npBAF) and the neuron-specific chromatin remodeling complex (nBAF). During neural development, a shift from a stem/progenitor to a postmitotic chromatin remodeling mechanism occurs as neurons exit the cell cycle and differentiate. This transition involves a change in the subunit composition of npBAF and nBAF complexes. As neural progenitors differentiate into neurons, npBAF complexes containing ACTL6A/BAF53A and PHF10/BAF45A are replaced by homologous subunits ACTL6B/BAF53B and DPF1/BAF45B or DPF3/BAF45C in neuron-specific (nBAF) complexes. The npBAF complex is essential for the self-renewal and proliferation of multipotent neural stem cells. The nBAF complex, along with CREST, regulates the expression of genes essential for dendrite growth.
Gene References Into Functions
  1. GATA binding protein 4 (Gata4), T-box transcription factor 5 (Tbx5), and BRG1-associated factor 60C protein (Baf60c) together induce the differentiation of adipose tissue-derived mesenchymal stem cells (ADMSCs) into cardiomyocytes. PMID: 26071180
  2. Knockdown or antibody blockade of Wnt5a reverses Smarcd3-induced epithelial-mesenchymal transition, indicating that Smarcd3 epigenetically regulates this transition by activating Wnt signaling pathways. PMID: 23716599
  3. Deptor is induced by the Baf60c-Six4 transcriptional complex and mediates Baf60c's activation of Akt and glycolytic metabolism in a cell-autonomous manner. PMID: 23563706
  4. BAF60c promotes lipogenesis *in vivo* and elevates triglyceride levels, highlighting its role in metabolic adaptation and activation of the lipogenic program in response to feeding and insulin. PMID: 23219531
  5. Five genes (BCL6, T (BRACHYURY), c-MYC, MITF, and BAF60C (SMARCD3)) efficiently convert postnatal chorion and decidual cells into chondrocytes. PMID: 22833560
  6. Phosphorylation of BAF60c on a conserved threonine promotes its incorporation into a Brg1-based SWI/SNF complex, leading to chromatin remodeling and activation of MyoD target genes. PMID: 22068056
  7. GTNB directs cardiogenesis from human embryonic stem cells and promotes proper cardiac development. PMID: 21694703
  8. BAF60c acts as a coregulator, serving as a crucial anchor point for recruiting the SWI/SNF complex to nuclear receptors and other transcription factors. PMID: 14701856
  9. Tat-mediated activation of the HIV promoter requires the SWI/SNF complex in synergy with the coactivator p300. PMID: 16687403
  10. Baf60c is re-expressed in Muller glial cells that re-enter the cell cycle after neurotoxic injury. PMID: 18816825
Database Links

HGNC: 11108

OMIM: 601737

KEGG: hsa:6604

STRING: 9606.ENSP00000262188

UniGene: Hs.647067

Protein Families
SMARCD family
Subcellular Location
Nucleus.
Tissue Specificity
Isoform 2 and isoform 1 are expressed in brain, heart, kidney, placenta, prostate, salivary gland, spleen, testis, thyroid, trachea and uterus. Isoform 1 is also expressed in skeletal muscle and adipose tissue.

Q&A

What is SMARCD3 and what is its role in the SWI/SNF complex?

SMARCD3, also known as BAF60C, is a subunit of the SWI/SNF (SWItch/Sucrose Non-Fermentable) chromatin remodeling complex. This complex plays an essential role in regulating transcriptional networks and the timing of gene expression through ATP-dependent nucleosome displacement along or from DNA molecules . SMARCD3 specifically functions as an accessory subunit that confers functional specificity to the SWI/SNF complex . Unlike core catalytic subunits that provide ATPase activity, SMARCD3 ensures tissue specificity and mediates transcriptional or co-transcriptional activities . This allows the complex to target specific genomic regions and regulate distinct gene expression programs.

How does SMARCD3 differ from other SMARCD family members?

The SMARCD family consists of three mutually exclusive accessory subunits (SMARCD1/BAF60A, SMARCD2/BAF60B, and SMARCD3/BAF60C) that can be incorporated into the SWI/SNF complex. Research has demonstrated that:

  • Expression patterns differ significantly: SMARCD3 exhibits tissue-specific expression patterns, particularly in cardiac and muscle tissues, while SMARCD1 has broader expression .

  • Their expression levels are not correlated either positively or negatively, indicating they do not compensate for each other when one is absent or downregulated .

  • SMARCD3 often shows more pronounced alterations in cancer compared to other family members: In prostate cancer, SMARCD3 exhibits clearly higher expression levels in malignant cells compared to non-malignant RWPE-1 cells, while SMARCD1 and SMARCD2 levels remain relatively consistent .

  • Only SMARCD3 has demonstrated significant prognostic value in certain cancers (such as breast cancer), with low SMARCD3 correlating with worse patient outcomes .

Research in prostate cancer models has shown that these proteins "exhibit independent, shared and redundant functions," yet maintain distinct roles in cellular processes .

How is SMARCD3 expression detected in research settings?

Multiple methodologies are employed to quantify SMARCD3 expression:

  • mRNA expression analysis: RNA sequencing (RNA-seq) and quantitative PCR are commonly used to measure SMARCD3 transcript levels. Single-cell RNA sequencing (scRNA-seq) has been used to demonstrate subgroup-specific expression in medulloblastoma, showing that 40.98% of Group 3 medulloblastoma cells express SMARCD3 compared with lower percentages in other subgroups (Group 4: 15.67%; SHH: 5.43%; WNT: 13.14%) .

  • Protein detection: Immunohistochemistry (IHC) using human tissue microarrays has been effectively employed to assess SMARCD3 protein levels in patient samples. This method has revealed correlations between SMARCD3 expression and clinicopathological variables, such as estrogen receptor (ER) and progesterone receptor (PR) status in breast cancer .

  • Proteomics approaches: Mass spectrometry-based proteomics has successfully identified differential SMARCD3 protein expression across cancer subtypes, as demonstrated in medulloblastoma research .

How do SMARCD3 expression patterns correlate with disease prognosis?

SMARCD3 expression exhibits complex relationships with disease outcomes that vary by cancer type:

Medulloblastoma: Higher levels of SMARCD3 expression correlate with poorer prognosis across all subgroups, independent of age and sex. Immunohistochemistry analysis confirmed that high SMARCD3 levels are associated with worse patient outcomes .

Breast Cancer: In contrast to medulloblastoma, low SMARCD3 expression correlates with worse outcomes in breast cancer. Patients with unaltered SMARCD3 have been shown to survive an average of 673 additional days (1.9 years) compared to patients with low SMARCD3 expression . Nuclear SMARCD3 expression has demonstrated predictive value specifically for survival in ER-positive breast cancer patients.

Gastric Cancer: SMARCD3 overexpression has been identified as a negative prognostic marker for gastric cancer through Kaplan-Meier analysis .

This contradictory prognostic significance across different cancer types highlights the context-dependent nature of SMARCD3 function, necessitating cancer-specific research approaches.

What experimental models are most appropriate for studying SMARCD3 function?

Researchers have successfully employed several model systems to investigate SMARCD3 function:

Cell Line Models:

  • Medulloblastoma: MED8A, D341, D458, and D425 cell lines have been used effectively for SMARCD3 functional studies .

  • Breast Cancer: MDA-MB-231 and MDA-MB-468 have demonstrated utility for proliferation and migration assays .

  • Prostate Cancer: RWPE-1 (non-malignant), LnCAP, C4-2, PC3, and DU145 have been used to study SMARCD3 in the context of prostate cancer .

Animal Models:

  • Mouse orthotopic xenograft models have been employed to study SMARCD3's role in tumor metastasis, particularly for medulloblastoma research .

  • Genetically engineered mouse models (GEMMs) with conditional knockout capabilities, such as the FSF-Kras G12D/+; p53 frt/frt; Pdx-Flp dual-recombinase model, allow for genetic deletion post-tumor establishment .

  • Virus-induced spontaneous tumor formation in postnatal C57BL/6J mice has been used to assess SMARCD3's contribution to tumor development .

Patient-Derived Models:

  • Three-dimensional (3D) culture systems using patient-derived organoids provide physiologically relevant environments for studying SMARCD3 function .

What genetic manipulation techniques are effective for SMARCD3 functional studies?

Several approaches have demonstrated efficacy:

CRISPR-Cas9 Gene Editing:

  • CRISPR-Cas9-mediated SMARCD3 knockout (KO) has been successfully employed in multiple cancer cell lines to study effects on migration, metastasis, and signaling pathways .

RNAi-Mediated Knockdown:

  • shRNA-mediated knockdown of SMARCD3 has been effective, with multiple studies utilizing at least two independent shRNAs to ensure specificity of observed phenotypes .

Overexpression Systems:

  • Viral vectors for SMARCD3 overexpression have been used to investigate gain-of-function effects .

Inducible Systems:

  • Tamoxifen-inducible R26-CreER^T2 CRE systems permit temporally controlled deletion of SMARCD3 in mouse models .

These methodologies allow researchers to manipulate SMARCD3 levels in both in vitro and in vivo settings, facilitating comprehensive functional characterization.

How does SMARCD3 contribute to cancer metastasis?

SMARCD3 promotes metastatic capacity through multiple mechanisms:

Medulloblastoma Metastasis:

  • SMARCD3 regulates Disabled 1 (DAB1)-mediated Reelin signaling to drive medulloblastoma metastatic dissemination .

  • SMARCD3 deletion decreases cell migration in scratch-wound healing and Transwell assays, and reduces spinal metastasis in orthotopic xenograft models .

  • SMARCD3 knockout reduces the number of circulating tumor cells (CTCs) in peripheral blood, while overexpression increases CTCs and spinal metastasis .

  • Notably, SMARCD3 is highly expressed in the tumor margin compared to the tumor center, suggesting that cells with high SMARCD3 levels tend to spread from the primary site .

Epithelial-Mesenchymal Transition (EMT):

  • In gastric cancer, SMARCD3 overexpression promotes EMT, thereby enhancing invasion and migration capabilities .

  • Functional assays with SMARCD3 knockdown and overexpression have demonstrated that its depletion reduces cell migration and invasion, while overexpression increases cellular irregularity .

These findings support SMARCD3's role as a key driver of metastatic progression across multiple cancer types, with evidence suggesting it contributes to poor prognosis primarily through enhanced metastatic potential rather than increased proliferation .

What signaling pathways does SMARCD3 regulate in cancer?

SMARCD3 influences multiple oncogenic signaling cascades:

PI3K/AKT Pathway:

  • SMARCD3 overexpression increases p-AKT-S473 levels in gastric cancer cells (MKN-74) .

  • It also enhances PI3Kp85 activity in KATO III cells, while knockout decreases this activity in SNU 601 cells .

MAPK/ERK Pathway:

  • Elevated SMARCD3 expression boosts p-ERK levels in gastric cancer models .

Wnt/β-catenin Signaling:

  • SMARCD3 overexpression increases β-catenin activity in KATO III cells .

  • In cardiac development, SMARCD3 has been shown to regulate Wnt signaling .

Androgen Receptor (AR) Signaling:

  • All SMARCD family proteins (including SMARCD3) are involved in the regulation of hormone-dependent AR-driven genes in prostate cancer .

  • SMARCD proteins can regulate AR-downstream targets even in androgen-depleted cells, suggesting involvement in progression to castration resistance .

DNA Damage Repair Pathways:

  • Low SMARCD3 expression correlates with increased homologous recombination deficiency (HRD) scores and markers in breast cancer, suggesting its involvement in DNA repair mechanisms .

This multilayered involvement in critical oncogenic pathways makes SMARCD3 a significant factor in cancer biology and a potential therapeutic target.

How does SMARCD3 influence cell cycle progression?

SMARCD3 plays a complex role in cell cycle regulation:

G1/S Transition:

  • SMARCD3 depletion leads to modest accumulation in G1 phase and slight decrease in S phase entry .

  • Knockdown of SMARCD3 results in strong accumulation of p21 (cyclin-dependent kinase inhibitor), though interestingly, this elevated p21 doesn't always correlate with reduced proliferation rates, suggesting cells might not respond properly to activated checkpoints .

S Phase Duration:

  • Single-cell tracking using the FUCCI system has revealed that individual cell cycle phases, particularly S-phase, are longer in SMARCD3-depleted cells .

S/G2 Transition and Endoreplication:

  • SMARCD3 depletion compromises the S/G2 transition, with some cells initiating a new cycle of DNA synthesis instead of entering mitosis (endoreplication) .

  • This results in whole-genome doubling, a phenomenon estimated to occur in 45% of breast cancers .

  • Consistent with this finding, SMARCD3 expression is significantly lower in breast tumors that have undergone whole-genome doublings .

G2/M Progression:

  • Lower proliferation rates observed in SMARCD3-depleted cells reflect failure to fully progress through G2/M phase .

These findings suggest SMARCD3 functions as a key regulator of cell cycle checkpoints, with its loss potentially contributing to genomic instability.

What is SMARCD3's relationship with DNA damage repair?

SMARCD3 appears to play a crucial role in DNA damage response:

DNA Damage Accumulation:

  • In SMARCD3-depleted cells, DNA damage accumulates and remains unrepaired .

Homologous Recombination Deficiency:

  • Analysis of breast cancer samples revealed that the number of homologous recombination deficiency (HRD) markers and the total HRD score are significantly higher in SMARCD3-low than in SMARCD3-high ER+ samples .

  • These markers include telomeric allelic imbalances (NtAI), large-scale state transitions (LST), and genomic segments with loss of heterozygosity (HRD-LOH) .

Correlation with HDR Signature:

  • SMARCD3 expression levels directly correlate with the Homology Directed Repair (HDR) signature, which encompasses genes whose expression decreases in response to DNA damage .

Genotoxic Sensitivity:

  • SMARCD3-depleted cells exhibit higher sensitivity to genotoxic agents such as radiation, despite their slower proliferation rates .

These findings suggest that SMARCD3 may function as a tumor suppressor in certain contexts by maintaining genomic stability through efficient DNA damage repair.

How can SMARCD3's chromatin remodeling activity be specifically assessed?

Methodologies for investigating SMARCD3's chromatin remodeling function include:

Chromatin Immunoprecipitation (ChIP):

  • ChIP-seq analysis can identify genomic regions where SMARCD3-containing SWI/SNF complexes bind, revealing its target genes and regulatory elements .

  • Integrated RNA-seq and ChIP-seq via network analysis has shown that SMARCD3 knockdown drives losses in BAF complex binding and H3K27-acetylation at active enhancers cobound by transcription factors like FOXA1 .

ATAC-seq (Assay for Transposase-Accessible Chromatin):

  • This method can identify changes in chromatin accessibility resulting from SMARCD3 activity or depletion, providing insights into its chromatin remodeling function .

Nucleosome Positioning Assays:

  • These assays can determine how SMARCD3 affects nucleosome organization and positioning around specific genomic regions.

Co-Immunoprecipitation (Co-IP):

  • Co-IP experiments can identify SMARCD3's protein interaction partners within the SWI/SNF complex and with other transcription factors .

  • Flag co-immunoprecipitation has been used to demonstrate interaction between PHF7 and SMARCD3 .

Proximity Labeling Approaches:

  • Methods like BioID or TurboID (as in PHF7 mTurbo experiments) can identify proteins in close proximity to SMARCD3 in living cells, revealing its interactome in different cellular contexts .

These complementary approaches allow researchers to comprehensively characterize SMARCD3's role in chromatin remodeling and transcriptional regulation.

What are the challenges in reconciling SMARCD3's apparently contradictory roles in different cancer types?

Resolving SMARCD3's context-dependent functions requires addressing several research challenges:

Tissue-Specific Effects:

  • SMARCD3's opposing roles in different cancers (oncogenic in medulloblastoma vs. tumor-suppressive in breast cancer) likely reflect tissue-specific functions .

  • Research should focus on identifying tissue-specific interaction partners and transcriptional programs regulated by SMARCD3.

Methodological Limitations:

  • The Cancer Dependency Map (DepMap) database, which explores gene function in 2D cancer cell lines, shows SMARCD3 as an enrichment in cancer cell lines, contradicting findings from more physiologically relevant 3D and in vivo models .

  • This discrepancy highlights the importance of using appropriate model systems: "2D functional screening may miss or underestimate the impact of genes playing a role in invasiveness, 3D/in vivo growth, or stemness" .

Subunit Composition Differences:

  • The specific composition of SWI/SNF complexes containing SMARCD3 likely differs between cancer types, affecting function.

  • Comprehensive proteomics analysis of SMARCD3-containing complexes across different tissues and cancer types could help resolve these contradictions.

Genomic Context Variations:

  • The genomic landscape of different cancers influences how SMARCD3 affects gene expression.

  • Integrative multi-omics approaches combining expression data with mutation profiles, epigenomic landscapes, and chromatin accessibility patterns are needed.

Addressing these challenges requires multidisciplinary approaches and careful consideration of cellular context when interpreting SMARCD3 functional data.

What is SMARCD3's role in normal development and non-cancer pathologies?

SMARCD3 functions in several developmental and disease contexts:

Cardiac Development:

  • SMARCD3 (BAF60c) orchestrates cardiac development by regulating transcriptional networks .

  • It interacts with PHF7 and can recruit cardiac transcription factors like Gata4 .

Neurodevelopment:

  • SMARCD3 regulates Disabled 1 (DAB1)-mediated Reelin signaling in Purkinje cell migration during cerebellar development .

  • This neurodevelopmental program is later hijacked in medulloblastoma to promote metastasis .

Muscle Development and Function:

  • The long non-coding RNA SMARCD3-OT1 promotes muscle hypertrophy and fast fiber-type differentiation by enhancing SMARCD3X4 expression at both mRNA and protein levels .

  • This mechanism improves myoblast proliferation and myotube formation .

Neurodegenerative Diseases:

  • SMARCD3 has been identified in transcriptomic signatures associated with cortical thickness in behavioral variant frontotemporal dementia (bvFTD) .

  • It shows differential expression patterns in patients with pathogenic variants in C9orf72, GRN, or MAPT genes .

These diverse roles highlight SMARCD3's importance beyond cancer, suggesting its broader significance in tissue development and homeostasis.

How do epigenetic modifications influence SMARCD3 expression and function?

Epigenetic regulation of SMARCD3 occurs through several mechanisms:

Promoter Methylation:

  • In triple-negative breast cancer (TNBC), SMARCD3 silencing through promoter methylation may contribute to cancer progression .

  • This epigenetic inactivation, rather than mutation or copy number loss, appears to be a significant mechanism for SMARCD3 downregulation in certain cancers .

Histone Modifications:

  • As part of the SWI/SNF complex, SMARCD3 both regulates and is regulated by histone modifications.

  • SMARCD3 knockdown leads to reduced H3K27-acetylation at active enhancers, suggesting a positive feedback mechanism .

Long Non-coding RNA Regulation:

  • SMARCD3-OT1, a long non-coding RNA, promotes SMARCD3X4 expression in muscle development .

  • This represents an additional layer of epigenetic control over SMARCD3 function in specific tissues.

Reader Protein Interactions:

  • SMARCD3 utilizes histone reader proteins such as PHF7 to identify target sites and exert its chromatin-modifying effects .

  • This interaction allows for context-specific targeting of SMARCD3-containing SWI/SNF complexes.

Understanding these epigenetic regulatory mechanisms provides opportunities for therapeutic interventions targeting SMARCD3 expression or function in various disease contexts.

What are the most promising approaches for targeting SMARCD3 therapeutically?

Several strategies show potential for SMARCD3-directed therapies:

Synthetic Lethality Approaches:

  • Exploiting vulnerabilities created by SMARCD3 deficiency, particularly in DNA damage repair pathways, could lead to targeted therapies for SMARCD3-low cancers .

  • The correlation between low SMARCD3 and increased homologous recombination deficiency suggests PARP inhibitors might be effective in SMARCD3-deficient tumors .

Protein-Protein Interaction Disruptors:

  • Small molecules targeting specific interactions between SMARCD3 and other proteins (such as transcription factors or other SWI/SNF components) could modulate its function in a context-dependent manner.

  • The identified interaction between SMARCD3 and PHF7 represents one potential target .

Epigenetic Modifiers:

  • For cancers where SMARCD3 is silenced by promoter methylation, demethylating agents could restore expression and tumor suppressor function .

RNA-Based Therapeutics:

  • siRNA or antisense oligonucleotides targeting SMARCD3 could be effective in cancers where it functions as an oncogene, such as medulloblastoma .

Pathway-Specific Inhibitors:

  • Since SMARCD3 regulates multiple signaling pathways (PI3K/AKT, MAPK/ERK, Wnt/β-catenin), combinatorial approaches targeting these downstream effectors might be effective .

The context-dependent nature of SMARCD3 function necessitates careful consideration of cancer type and molecular context when developing therapeutic strategies.

What key research gaps remain in our understanding of SMARCD3 biology?

Despite significant advances, several critical knowledge gaps persist:

Structural Determinants of Specificity:

  • The structural basis for SMARCD3's unique functions compared to other SMARCD family members remains poorly understood.

  • Structural studies of SMARCD3-containing SWI/SNF complexes could reveal targeting opportunities.

Tissue-Specific Interaction Networks:

  • Comprehensive mapping of SMARCD3's protein interactions across different tissues and disease states would help explain its context-dependent functions.

Post-Translational Modifications:

  • The regulation of SMARCD3 through post-translational modifications and how these affect its function within the SWI/SNF complex require further investigation.

Non-Canonical Functions:

  • Potential roles of SMARCD3 beyond chromatin remodeling, such as in RNA processing or metabolism, remain unexplored.

Predictive Biomarkers:

  • Development of reliable biomarkers to identify patients who would benefit from SMARCD3-targeted therapies represents a significant clinical need.

Addressing these gaps requires interdisciplinary approaches combining structural biology, systems biology, biochemistry, and clinical research to fully elucidate SMARCD3's complex biology and therapeutic potential.

Table 1: SMARCD3 Expression and Prognostic Significance Across Cancer Types

Cancer TypeSMARCD3 Expression PatternPrognostic SignificanceReference
MedulloblastomaHighly expressed in Group 3 (40.98% of cells) compared to other subgroups (Group 4: 15.67%; SHH: 5.43%; WNT: 13.14%)Higher expression correlates with poorer prognosis
Breast CancerOften downregulated compared to normal tissue; correlated with ER and PR statusLow expression associated with worse outcomes; patients with normal SMARCD3 survive 673 days longer
Prostate CancerElevated in malignant cell lines compared to non-malignant RWPE-1 cellsAltered in 6% of non-metastatic and 5-9% of metastatic cases
Gastric CancerFrequently overexpressedOverexpression serves as negative prognostic marker
Pancreatic CancerElevated in pancreatic ductal adenocarcinomaAssociated with stem cell characteristics and therapy resistance

Table 2: Functional Effects of SMARCD3 Manipulation in Experimental Models

Experimental ApproachCancer ModelObserved EffectsReference
CRISPR-Cas9 KnockoutMedulloblastoma (MED8A, D341)Decreased cell migration and spinal metastasis
shRNA KnockdownPancreatic Cancer (KPC, KP^f/f C)Reduced 3D growth by >50%, inhibited proliferation, increased cell death
shRNA KnockdownBreast Cancer (MDA-MB-231, MDA-MB-468)Lower proliferation rates, G1 accumulation, increased p21
OverexpressionMedulloblastoma (D425)Increased cell migration, spinal metastasis, and circulating tumor cells
OverexpressionPancreatic Cancer (KP^f/f C)Increased 3D growth by 2-fold, sustained CD133+ stem cell fraction
Conditional KnockoutPancreatic Cancer (Smarcd3^f/f; FSF-Kras^G12D/+; p53^frt/frt; Pdx-Flp)Suppressed tumor development when deleted post-establishment

Quick Inquiry

Personal Email Detected
Please use an institutional or corporate email address for inquiries. Personal email accounts ( such as Gmail, Yahoo, and Outlook) are not accepted. *
© Copyright 2025 TheBiotek. All Rights Reserved.