SMNDC1 Human

Survival Motor Neuron Domain Containing 1 Human Recombinant
Shipped with Ice Packs
In Stock

Description

Functional Roles in RNA Splicing and Disease

SMNDC1 regulates splicing through its Tudor domain and phase-separating properties . Key findings include:

Role in Spliceosome Assembly

SMNDC1 recruits U4/U5/U6 tri-snRNPs to pre-spliceosomes via Tudor domain-mediated interactions . Disruption of this function alters splicing of genes involved in chromatin remodeling (e.g., ATRX) and transcription factors (e.g., PDX1) .

Disease Associations

DiseaseMechanismSource
DiabetesSMNDC1 knockdown induces insulin production in pancreatic α-cells .
CancerOverexpression correlates with poor survival in hepatocellular carcinoma .
NeurodegenerationTudor domain interactions mirror SMN protein dynamics in motor neuron diseases .

Autoregulation via Poison Exons

A conserved poison exon in SMNDC1 introduces premature termination codons (PTCs) when included, triggering nonsense-mediated decay (NMD) . Loss of this exon increases SMNDC1 protein levels by ~8–15% in mice, disrupting RNA processing homeostasis .

Therapeutic Targeting and Inhibitors

Recent advances in SMNDC1 inhibition highlight its potential as a therapeutic target:

Small-Molecule Inhibitors

Kubicek et al. identified inhibitors that bind the Tudor domain, disrupting:

  • Protein interactions: Loss of binding to sDMA-marked Sm proteins .

  • Subcellular localization: Dissolution of nuclear speckles and phase-separated condensates .

  • Splicing outcomes: Altered splicing of SMNDC1-dependent genes (e.g., PDX1, ATRX) .

Inhibitor Characteristics

ParameterDetail
Binding SiteTudor domain’s sDMA-binding pocket
Cellular EffectSplicing changes in α-cells, promoting insulin
Therapeutic UseDiabetes, hepatocellular carcinoma

Research Tools and Antibodies

Proteintech’s SMNDC1 antibody (68344-1-Ig) enables detection via:

ApplicationDilutionReactivity
Western Blot1:5,000–1:50,000Human
Flow Cytometry0.8 µg/10⁶ cellsHuman

Validated Cell Lines

Cell LineDetection
HeLaWB, FC (Intra)
HEK-293WB
JurkatWB

Product Specs

Introduction
SMNDC1, a member of the SMN family, is an essential splicing factor crucial for spliceosome assembly. This protein contains a Tudor domain that shares significant similarity with the Survival Motor Neuron (SMN) protein. SMNDC1 is found in various tissues, including the heart, pancreas, and skeletal muscle, and localizes to Cajal bodies and nuclear speckles within cells. Notably, mutations in the SMNDC1 gene can lead to autosomal recessive proximal spinal muscular atrophy.
Description
This product consists of the SMNDC1 protein produced in E. coli. It is a single, non-glycosylated polypeptide chain comprising 258 amino acids (amino acids 1-238) and has a molecular weight of 28.9 kDa. The protein includes a 20 amino acid His-tag fused at the N-terminus to facilitate purification, which is carried out using proprietary chromatographic techniques.
Physical Appearance
A clear solution that has undergone sterile filtration.
Formulation
The SMNDC1 protein is provided at a concentration of 0.5 mg/ml in a 20mM Tris-HCl buffer with a pH of 8.0. The buffer also contains 1mM DTT, 100mM NaCl, and 10% glycerol.
Purity
The purity of the SMNDC1 protein is greater than 90%, as determined by SDS-PAGE analysis.
Stability
For short-term storage (2-4 weeks), the product can be stored at 4°C. For longer-term storage, it is recommended to freeze the product at -20°C. Repeated freezing and thawing of the product should be avoided.
Synonyms
SMNR, SMN-related protein, SPF30, Survival Motor Neuron Domain Containing 1, 30kDa Splicing Factor SMNrp, Splicing Factor 30, Survival of Motor Neuron-related.
Source
Escherichia Coli.
Amino Acid Sequence
MGSSHHHHHH SSGLVPRGSH MSEDLAKQLA SYKAQLQQVE AALSGNGENE DLLKLKKDLQ EVIELTKDLL STQPSETLAS SDSFASTQPT HSWKVGDKCM AVWSEDGQCY EAEIEEIDEE NGTAAITFAG YGNAEVTPLL NLKPVEEGRK AKEDSGNKPM SKKEMIAQQR EYKKKKALKK AQRIKELEQE REDQKVKWQQ FNNRAYSKNK KGQVKRSIFA SPESVTGKVG VGTCGIADKP MTQYQDTSKY NVRHLMPQ

Q&A

What is SMNDC1 and what is its relationship to SMN1?

SMNDC1 (survival motor neuron domain containing 1) is a protein-coding gene located on chromosome 10q25.2 in humans. It functions as a paralog of the SMN1 gene, which encodes the survival motor neuron protein implicated in autosomal recessive proximal spinal muscular atrophy . While sharing similar cellular functions with SMN1, SMNDC1 is distinctly identified as a nuclear protein and constituent of the spliceosome complex . Research approaches to understand this relationship typically involve comparative genetic analysis, protein domain mapping, and functional studies using knockout models to identify shared and divergent pathways between these related genes.

What are the known cellular functions of SMNDC1?

SMNDC1 functions primarily as a splicing factor involved in RNA processing within the nucleus . Methodologically, its functions have been established through:

  • Spliceosome complex isolation and characterization studies

  • RNA interference screens demonstrating its role in regulating gene expression

  • Chromatin immunoprecipitation experiments revealing its interaction with chromatin remodeling complexes

Research has demonstrated that SMNDC1 participates in transforming RNA into final messenger RNA that carries genetic information, effectively influencing the expression of many other proteins throughout the cell . Notably, it plays a critical role in connecting splicing and chromatin remodeling mechanisms to control insulin expression in human and mouse islet cells .

How is SMNDC1 expression regulated throughout the body?

SMNDC1 is differentially expressed throughout the body, with particularly abundant levels detected in skeletal muscle tissue . This expression pattern has been established through:

  • Tissue-specific quantitative PCR analysis

  • RNA sequencing of different human tissues

  • Immunohistochemical profiling of protein expression

Interestingly, SMNDC1 is subject to regulation by alternative splicing via the inclusion of a highly conserved poison exon, a regulatory mechanism preserved across different taxonomic kingdoms from plants to humans . Research investigating this regulation typically employs splicing reporter assays, CRISPR-mediated genetic modification of regulatory elements, and cross-species comparative genomics.

How does SMNDC1 contribute to diabetes pathophysiology?

SMNDC1 has emerged as a critical regulator of pancreatic cell function with significant implications for diabetes research. Methodologically, its role has been established through:

  • RNA interference screens in murine alpha cell lines that identified SMNDC1 as a silencer of insulin expression

  • Mechanistic studies showing that SMNDC1 knockdown triggers global repression of alpha cell gene-expression programs while increasing beta cell markers

  • Functional studies demonstrating that SMNDC1 loss in human pancreatic islets improves glucose sensitivity and enhances insulin secretion

When SMNDC1 is downregulated, research has shown upregulation of PDX1 (a key beta cell transcription factor) through modulation of the BAF and Atrx chromatin remodeling complexes . This suggests potential therapeutic approaches targeting SMNDC1 to repurpose alpha cells for insulin production in diabetes treatment strategies.

What connections exist between SMNDC1 and cancer development?

Research has identified associations between SMNDC1 and cancer pathways, though detailed mechanisms remain an active area of investigation. Current methodological approaches include:

  • Genome-wide association studies linking SMNDC1 variants to specific cancer types

  • Gene expression profiling in tumor versus normal tissues

  • Functional studies examining how SMNDC1-mediated splicing affects oncogenes and tumor suppressors

Scientists have noted that as an essential gene present in nearly every cell, SMNDC1 dysregulation can potentially contribute to cancerous transformation through altered splicing patterns of critical regulatory genes . Research teams are currently exploring the therapeutic potential of SMNDC1 inhibitors, following the successful identification of compounds that can regulate this essential protein .

Is SMNDC1 implicated in other human diseases?

Beyond diabetes and cancer, SMNDC1 has been linked to several other human disorders. Research methodologies exploring these connections include:

  • Genome-wide association studies that have identified SMNDC1 as a susceptibility locus for Crohn's disease in Korean populations

  • Investigation of SMNDC1's role in spinal muscular atrophy pathways, given its paralogous relationship with SMN1

  • Splicing analysis in various disease models to identify aberrant SMNDC1 activity

Researchers typically approach these disease associations through comparative transcriptomics, splicing pattern analysis, and genetic screening in affected populations, followed by functional validation in cellular and animal models.

What techniques are most effective for studying SMNDC1's splicing activity?

To effectively study SMNDC1's splicing function, researchers employ several specialized techniques:

  • Modified Sm-ring assembly assays to detect ring assembly on polyA-enriched RNA using anti-Sm RNA immunoprecipitation and next-generation sequencing (RIP-Seq)

  • RNA immunoprecipitation experiments to confirm association of Sm-site containing mRNAs with Sm proteins in the cytoplasm

  • Bioinformatic pipelines to identify Sm-site containing RNAs in transcriptomes

  • Splicing reporter constructs containing minigenes to assess SMNDC1-dependent exon inclusion/exclusion events

These methodologies have revealed that SMNDC1 participates in Sm-ring assembly on mRNAs containing Sm-sites, particularly those enriched in 3' untranslated regions, providing direct links between SMNDC1 function and RNA processing events .

How can researchers effectively knockdown or modulate SMNDC1 in experimental models?

Successful modulation of SMNDC1 in research settings employs several complementary approaches:

  • RNA interference using siRNA or shRNA libraries, which has been effectively used in alpha cell lines to demonstrate SMNDC1's role in insulin expression

  • CRISPR-Cas9 gene editing to create knockout or knockin models for studying complete loss or specific mutations

  • Pharmacological inhibition using recently identified compounds that regulate SMNDC1 activity

  • Overexpression systems using vectors containing wild-type or mutant SMNDC1 constructs

When implementing these approaches, researchers must carefully consider cell type-specific effects, as SMNDC1 is essential for most cell types and its complete loss impairs cellular viability . Partial knockdown or tissue-specific conditional knockout models often provide more interpretable results for understanding SMNDC1 function.

What are the recommended protocols for investigating SMNDC1 interactions with chromatin remodeling complexes?

To study SMNDC1's interactions with chromatin remodeling complexes, researchers typically employ these methodological approaches:

  • Chromatin immunoprecipitation followed by sequencing (ChIP-seq) to identify genomic binding sites of SMNDC1 and associated chromatin remodelers

  • Co-immunoprecipitation assays to detect direct protein-protein interactions between SMNDC1 and components of the BAF and Atrx complexes

  • Proximity ligation assays to visualize protein interactions in situ

  • ATAC-seq (Assay for Transposase-Accessible Chromatin using sequencing) before and after SMNDC1 modulation to assess changes in chromatin accessibility

These techniques have revealed that SMNDC1 knockdown affects the activities of BAF and Atrx chromatin remodeling complexes, leading to upregulation of PDX1 and subsequent changes in cell identity and function . When implementing these protocols, researchers should include appropriate controls and validation approaches to confirm the specificity of observed interactions.

How does the phase-separating property of SMNDC1 influence its cellular functions?

Recent research has uncovered that SMNDC1 undergoes phase separation, forming membraneless organelles within the nucleus. Methodological approaches to study this phenomenon include:

  • Fluorescence recovery after photobleaching (FRAP) to measure protein dynamics in cellular condensates

  • Optogenetic tools to control phase separation in living cells

  • In vitro reconstitution assays using purified components to assess concentration-dependent phase separation

  • Pharmacological perturbation using compounds that specifically disrupt phase-separated structures

Studies have shown that the pharmacological perturbation of phase-separating SMNDC1 protein can modulate its function, which has potential therapeutic applications . When investigating phase separation properties, researchers should consider how physiological conditions, post-translational modifications, and protein-protein interactions might influence SMNDC1 condensate formation and function.

What is the significance of SMNDC1's conserved poison exon across species?

The presence of a highly conserved poison exon in SMNDC1 represents an important evolutionary feature. Research approaches to understand its significance include:

  • Comparative genomics across diverse species to track evolutionary conservation

  • Targeted mutagenesis of the poison exon to assess functional consequences

  • Splicing analysis under various cellular conditions to determine regulatory mechanisms

  • Phylogenetic studies examining selection pressure on this genomic element

Research has revealed that this poison exon plays a crucial role in regulating SMNDC1 expression and function across different kingdoms from plants to humans . Researchers investigating this feature should employ cross-species models and consider how environmental stressors or developmental cues might affect poison exon inclusion and subsequent protein expression patterns.

How might SMNDC1 be therapeutically targeted for diabetes treatment?

The development of therapeutic approaches targeting SMNDC1 for diabetes represents an emerging research direction. Methodological considerations include:

  • High-throughput screening platforms to identify small molecules that modulate SMNDC1 activity

  • Structure-based drug design targeting specific SMNDC1 domains or interactions

  • Cell-based assays measuring insulin production after SMNDC1 modulation

  • Preclinical models evaluating pancreatic alpha-to-beta cell reprogramming strategies

Current research indicates that SMNDC1 inhibition could stimulate alpha cells to produce insulin and improve glucose sensitivity in pancreatic islets . When pursuing this therapeutic direction, researchers should carefully evaluate potential off-target effects, considering SMNDC1's essential role in many cell types and its involvement in multiple cellular processes beyond pancreatic function.

What are the most reliable antibodies and detection methods for SMNDC1 research?

Selecting appropriate tools for SMNDC1 detection is critical for research reproducibility. Methodological recommendations include:

  • Validation of antibody specificity using knockout controls or multiple antibodies targeting different epitopes

  • Implementation of both Western blotting and immunofluorescence approaches to confirm subcellular localization

  • Mass spectrometry-based detection for unbiased protein identification and quantification

  • Generation of tagged SMNDC1 constructs (with careful consideration of tag position to avoid functional interference)

When reporting SMNDC1 research, detailed documentation of antibody sources, catalog numbers, dilutions, and validation approaches should be provided to ensure reproducibility and facilitate cross-study comparisons.

What bioinformatic approaches best identify Sm-sites in transcriptomic data?

Computational identification of Sm-sites requires sophisticated bioinformatic pipelines. Effective methodological approaches include:

  • Sequence motif analysis using position weight matrices based on validated Sm-sites

  • Secondary structure prediction to assess accessibility of putative Sm-sites

  • Evolutionary conservation analysis to identify functionally important sites

  • Integration of RIP-Seq data to correlate predicted sites with experimental binding evidence

Research has established that Sm-sites are enriched in mRNA 3' untranslated regions , and bioinformatic approaches should be calibrated to detect both canonical and non-canonical sites with appropriate false discovery rate controls.

Product Science Overview

Introduction

Survival Motor Neuron Domain Containing 1 (SMNDC1), also known as Survival of Motor Neuron-Related-Splicing Factor 30 (SPF30), is a protein encoded by the SMNDC1 gene in humans. This protein is a constituent of the spliceosome complex, which is essential for RNA splicing, a critical process in gene expression. SMNDC1 is a paralog of the SMN1 gene, which encodes the survival motor neuron protein, mutations in which are the cause of autosomal recessive proximal spinal muscular atrophy .

Gene and Protein Structure

The SMNDC1 gene is located on chromosome 10 at the band 10q25.2 and spans approximately 14,208 base pairs. The protein encoded by this gene is a nuclear protein that has been identified as a constituent of the spliceosome complex. It is involved in the assembly of the U4/U5/U6 tri-small nuclear ribonucleoprotein into the spliceosome .

Function

SMNDC1 plays a crucial role in RNA splicing, a process that removes introns from pre-mRNA and joins exons together to form mature mRNA. This process is vital for the proper expression of genes and the production of functional proteins. The protein is also involved in mRNA processing and the apoptotic process .

Expression

The SMNDC1 gene is differentially expressed in various tissues, with abundant levels in skeletal muscle. It is also expressed in other tissues such as the amniotic fluid, buccal mucosa, germinal epithelium, cartilage tissue, gingival epithelium, parietal pleura, retinal pigment epithelium, jejunal mucosa, and palpebral conjunctiva .

Clinical Significance

Mutations in the SMN1 gene, a paralog of SMNDC1, are known to cause autosomal recessive proximal spinal muscular atrophy, a severe genetic disorder characterized by the loss of motor neurons in the spinal cord and brainstem, leading to muscle wasting and weakness. While SMNDC1 itself is not directly implicated in this disorder, its role in RNA splicing and its similarity to SMN1 suggest that it may share similar cellular functions .

Research and Applications

Research on SMNDC1 is ongoing to better understand its role in RNA splicing and its potential implications in genetic disorders. The recombinant form of this protein is used in various research applications to study its function and interactions within the spliceosome complex .

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.