LSM3 Human

LSM3 Homolog, U6 Small Nuclear RNA Associated Human Recombinant
Shipped with Ice Packs
In Stock

Description

LSM3 Human Recombinant produced in E. coli is a single polypeptide chain containing 125 amino acids (1-102) and having a molecular mass of 14.3kDa.
LSM3 is fused to a 23 amino acid His-tag at N-terminus & purified by proprietary chromatographic techniques.

Product Specs

Introduction
LSM3, a member of the snRNP Sm protein family, is found in numerous organisms and identified by sequence homology. These proteins possess an Sm sequence motif with two regions separated by a variable-length linker that folds into a loop. LSM3, like other Sm-like proteins, forms a stable heteromer within tri-snRNP particles essential for pre-mRNA splicing.
Description
Recombinant human LSM3, produced in E. coli, is a single polypeptide chain of 125 amino acids (residues 1-102) with a molecular weight of 14.3 kDa. The protein includes a 23 amino acid His-tag at the N-terminus and is purified using proprietary chromatographic techniques.
Physical Appearance
Clear, colorless solution, sterile-filtered.
Formulation
LSM3 is supplied in a solution at a concentration of 0.5 mg/mL in 20 mM Tris-HCl buffer (pH 8.0), 100 mM NaCl, 2 mM DTT, and 20% glycerol.
Stability
For short-term storage (2-4 weeks), store at 4°C. For extended storage, freeze at -20°C. Adding a carrier protein like 0.1% HSA or BSA is recommended for long-term storage. Avoid repeated freeze-thaw cycles.
Purity
Purity exceeds 90% as assessed by SDS-PAGE.
Synonyms
LSM3 homolog U6 small nuclear RNA associated (S. cerevisiae), U6 snRNA-associated Sm-like protein LSm3, SMX4, USS2, YLR438C.
Source
E.coli.
Amino Acid Sequence
MGSSHHHHHH SSGLVPRGSH MGSMADDVDQ QQTTNTVEEP LDLIRLSLDE RIYVKMRNDR ELRGRLHAYD QHLNMILGDV EETVTTIEID EETYEEIYKS TKRNIPMLFV RGDGVVLVAP PLRVG

Q&A

What is LSM3 and what complexes does it participate in?

LSM3 (Sm-like protein 3) in humans is a small protein belonging to the Sm-like protein family that participates in two distinct heptameric ring complexes: the cytoplasmic Lsm1-7 complex involved in mRNA decay and the nuclear Lsm2-8 complex that participates in pre-mRNA splicing . These complexes share several subunits (Lsm2-7) but differ in their unique components (Lsm1 or Lsm8) and cellular localization. The participation of LSM3 in these two complexes demonstrates its dual functionality in RNA processing pathways. Biochemical isolation of these complexes typically requires different approaches depending on which complex is being targeted - including differential cellular fractionation followed by immunoprecipitation with complex-specific antibodies.

How is LSM3 function conserved between humans and model organisms?

LSM3 shows remarkable evolutionary conservation from yeast to humans, particularly in its core RNA-binding domains. In yeast, Lsm3 has been demonstrated to interact with chromatin-associated Mediator at intron-containing ribosomal protein genes . Similar interactions likely exist in human cells, though with important species-specific differences in target genes and regulatory networks. When designing experiments using model organisms, researchers should consider that while the core functions of LSM3 are conserved, the regulatory contexts may differ significantly. Cross-species complementation assays can be particularly informative for assessing functional conservation of specific domains or mutations.

How can RNA-seq data be analyzed to identify LSM3-dependent splicing events?

RNA-seq analysis for LSM3-dependent splicing requires specialized analytical pipelines. First, establish LSM3 knockdown or knockout cell lines alongside controls. After RNA extraction and sequencing, differential splicing analysis should be performed using tools such as rMATS, MAJIQ, or VAST-TOOLS that specifically quantify various splicing events (exon skipping, intron retention, alternative 5' or 3' splice sites). Calculate splicing ratios across conditions and identify events that change significantly (typically using a threshold of p<0.05 and PSI change >10%). To distinguish direct from indirect effects, integrate these data with LSM3 binding profiles from CLIP-seq or similar techniques. Advanced analysis should include motif enrichment around affected splice sites and distinguishing between effects on co-transcriptional versus post-transcriptional splicing.

What are the optimal ChIP-seq protocols for studying LSM3 chromatin interactions?

For effective LSM3 ChIP-seq in human cells, begin with dual crosslinking using DSG (disuccinimidyl glutarate) followed by formaldehyde to capture potentially indirect chromatin interactions . Cell lysis should be performed under conditions that preserve nuclear integrity, followed by chromatin fragmentation to approximately 200-300bp fragments. The choice of antibody is critical - monoclonal antibodies against human LSM3 typically show higher specificity than polyclonal alternatives, though validation through western blotting and immunoprecipitation efficiency tests is essential. Include appropriate negative controls such as IgG and untagged wild-type strains. For peak calling, utilize a false discovery rate threshold of <0.05 and filter for peaks detected across at least three biological replicates with >2-fold enrichment . When analyzing the data, pay particular attention to intronic and exonic regions of genes involved in RNA processing and ribosome biogenesis, as these have been identified as enrichment hotspots in yeast studies.

How should researchers design co-immunoprecipitation experiments to study LSM3 complexes?

Design co-immunoprecipitation (co-IP) experiments for LSM3 by first determining which complex you're targeting (Lsm1-7 or Lsm2-8). For studying interactions with larger complexes like Mediator, optimize extraction conditions to preserve chromatin-associated interactions. The protocol should include:

  • Cell fractionation to separate cytoplasmic and nuclear components

  • Chromatin extraction using a buffer containing 300-400mM NaCl and 0.1% NP-40

  • Immunoprecipitation using LSM3-specific antibodies or epitope tags (FLAG, TAP, or Myc tags have all proven effective)

  • Stringent washing (at least 5 washes) to remove non-specific interactions

  • Western blotting for specific components of target complexes

When comparing results across different experimental conditions, ensure that loading controls and IP efficiency controls are included. Quantitative assessment of interaction strengths can be achieved through densitometric analysis of western blot signals normalized to input and IP efficiency.

ComplexRecommended TagBuffer ConditionsPositive Controls
Lsm1-7FLAG-Lsm1/Lsm3150mM NaCl, 0.1% NP-40Pat1, Xrn1
Lsm2-8FLAG-Lsm8/Lsm3250mM NaCl, 0.1% NP-40SmB, SmD1
Chromatin-boundTAP-Lsm3400mM NaCl, 0.1% NP-40Med1, Med15

What approaches effectively distinguish between LSM3's roles in the Lsm1-7 versus Lsm2-8 complexes?

To distinguish LSM3's functions in the two different complexes, employ these methodological approaches:

Complex-specific protein tags can clearly separate the two complexes - use Lsm1-FLAG to pull down the cytoplasmic Lsm1-7 complex and Lsm8-FLAG for the nuclear Lsm2-8 complex . When analyzing the immunoprecipitated material, probe for complex-specific factors: Lsm1-7 associates with decapping factors (Dcp1/2) and decay machinery, while Lsm2-8 associates with spliceosomal components.

Subcellular fractionation followed by western blotting can separate the cytoplasmic Lsm1-7 from the nuclear Lsm2-8 complex. Functional assays including RNA stability measurements (for Lsm1-7) and splicing efficiency analysis (for Lsm2-8) provide complementary evidence for complex-specific activities. Knockdown of complex-specific components (Lsm1 or Lsm8) followed by phenotypic and RNA processing analysis can further distinguish the roles of each complex.

How does LSM3 occupancy correlate with gene expression across different cell states?

LSM3 occupancy shows significant correlation with gene expression levels, particularly for intron-containing ribosomal protein genes. Analysis of RNA-seq data from different growth conditions in yeast revealed that genes co-occupied by Mediator and Lsm3 display higher and more homogeneous splice ratios as well as higher expression levels compared to genes binding neither factor .

When analyzing human cells, researchers should consider growth-dependent regulation - as cells transition from early to late logarithmic growth phase, LSM3-occupied genes show significant reductions in both splice ratios and expression levels . This correlation suggests a regulatory role for LSM3 in coordinating splicing efficiency with growth conditions.

To effectively study this correlation in human cells:

  • Perform parallel ChIP-seq for LSM3 and RNA-seq across different cellular states

  • Calculate splice ratios and expression levels for all genes

  • Compare these metrics between LSM3-occupied and non-occupied genes

  • Analyze changes in these parameters across cellular transitions (e.g., cell cycle progression, differentiation, or stress response)

What computational approaches best analyze LSM3 binding patterns genome-wide?

For comprehensive analysis of LSM3 binding patterns, implement this computational workflow:

First, perform peak calling using MACS2 with parameters optimized for transcription factor binding (narrow peaks) at an average fragment size of 300bp and FDR<0.05 . Filter peaks to retain only those detected across all biological replicates with >2-fold enrichment in at least one replicate.

For meta-gene analysis, normalize all genes to a standard length and generate heatmaps of LSM3 occupancy across gene bodies, with special attention to promoter regions, exon-intron boundaries, and 3'-exons where LSM3 shows preferential binding . Implement k-means clustering to identify groups of genes with similar binding patterns, followed by GO analysis of each cluster to identify functional correlations.

Integration with other datasets is crucial - correlate LSM3 binding with RNA-seq data (expression levels and splicing patterns), other protein occupancy data (especially Mediator components), and chromatin accessibility data. For binding motif analysis, extract sequences under peak summits (±50bp) and use tools like MEME or HOMER to identify enriched sequence motifs that might direct LSM3 recruitment.

How can CRISPR-Cas9 approaches be optimized for studying LSM3 function?

CRISPR-Cas9 approaches for LSM3 functional studies must balance complete gene disruption against potentially lethal phenotypes. Design multiple sgRNAs targeting different exons, with preference for early constitutive exons. For validation, design primers spanning the targeted region for genomic PCR and Sanger sequencing, as well as qPCR primers for measuring transcript levels.

Given LSM3's essential role in RNA processing, complete knockout may be lethal or severely growth-inhibiting. Therefore, consider these alternative approaches:

  • Inducible knockdown systems using CRISPRi with dCas9-KRAB to enable temporal control of repression

  • Domain-specific mutations that disrupt specific functions while preserving others

  • Degron tagging for rapid, inducible protein degradation

  • Homology-directed repair to introduce epitope tags for immunoprecipitation studies

After generating modified cell lines, comprehensive phenotypic analysis should include:

  • Growth rate measurements

  • RNA processing assessments (splicing efficiency, mRNA stability)

  • RNA-seq to identify global changes in gene expression and splicing

  • Co-IP studies to assess complex formation

What methods best assess LSM3 as a potential biomarker in human diseases?

To evaluate LSM3 as a potential biomarker, researchers should employ a multi-faceted approach:

Start with bioinformatic analysis of existing cancer genomics databases (TCGA, ICGC) to assess expression, mutation status, and correlation with patient outcomes across cancer types. Follow with validation using tissue microarrays with immunohistochemistry to quantify LSM3 protein levels across large cohorts of patient samples. Quantitative assessment requires digital image analysis for precise scoring of staining intensity and subcellular localization.

For liquid biopsy applications, develop sensitive assays to detect LSM3 mRNA or protein in blood, urine, or other accessible fluids. These might include digital PCR for rare transcript detection or ELISA/Luminex approaches for protein quantification. Single-cell RNA-seq of patient samples can reveal heterogeneity in LSM3 expression across different cell populations within tumors, potentially identifying specific cellular states associated with disease progression.

Functional validation of biomarker status should include correlation of LSM3 levels with therapeutic response in patient-derived xenograft models or organoid cultures, establishing whether LSM3 is merely correlative or functionally relevant to disease mechanisms.

How do RNA processing defects linked to LSM3 dysfunction contribute to pathogenesis?

RNA processing defects stemming from LSM3 dysfunction can contribute to pathogenesis through multiple mechanisms. To investigate these connections:

Perform global analysis of splicing patterns in patient samples or disease models using RNA-seq with specialized splice junction analysis. Key metrics to assess include: intron retention rates, exon skipping events, and usage of alternative 5' or 3' splice sites. Particular attention should be paid to cancer-relevant genes that may generate oncogenic splice variants.

Transcript stability analysis can reveal whether LSM3 dysfunction leads to inappropriate stabilization of transcripts that should be degraded, or premature decay of essential transcripts. This can be approached using actinomycin D chase experiments followed by RNA-seq or targeted qPCR.

To connect processing defects with cellular phenotypes, perform rescue experiments where wild-type LSM3 or specific mutant variants are reintroduced into knockout or patient-derived cells. Assess whether restoration of proper RNA processing correlates with normalization of disease-related cellular behaviors (proliferation, migration, or differentiation).

The most challenging aspect involves distinguishing primary from secondary effects, since global disruption of RNA processing can have cascading consequences throughout the transcriptome. Integration of direct binding data (from CLIP-seq) with functional outcomes can help establish causal relationships between specific LSM3-RNA interactions and pathogenic mechanisms.

How do post-translational modifications regulate human LSM3 function?

Post-translational modifications (PTMs) of LSM3 likely play critical roles in regulating its complex formation, localization, and activity. To characterize these modifications:

Begin with mass spectrometry analysis of immunoprecipitated LSM3 to identify sites of phosphorylation, ubiquitination, methylation, and other PTMs. Pay particular attention to modifications that change in response to cellular stresses or cell cycle progression. Site-directed mutagenesis of identified modification sites (e.g., changing phosphorylatable serines to alanines or phosphomimetic aspartates) can reveal the functional importance of specific modifications.

Analyzing the subcellular distribution of modified versus unmodified LSM3 provides insights into how PTMs affect complex formation and localization. Antibodies specific to modified forms can be particularly valuable, though these may need to be custom developed. Temporal dynamics of modifications can be studied using synchronized cell populations or rapid induction of stress responses.

Integration with signaling pathway analysis is essential - identify kinases, phosphatases, or other modifying enzymes that target LSM3 and determine how these activities change across cellular conditions. This may reveal how RNA processing is coordinated with broader cellular regulatory networks.

What are the latest techniques for assessing LSM3-RNA interactions at single-molecule resolution?

Single-molecule approaches have revolutionized our understanding of LSM3-RNA interactions. To implement these techniques:

Single-molecule FRET (smFRET) can reveal the dynamics of LSM3 complex assembly on RNA substrates by labeling both the protein complex and RNA with appropriate fluorophores. This approach requires careful protein purification and labeling strategies but provides unparalleled insights into binding kinetics and conformational changes.

For in vivo studies, consider APEX2 proximity labeling coupled with RNA-seq (APEX-RIP) to identify RNAs in the immediate vicinity of LSM3 in living cells. Alternative approaches include single-molecule fluorescence in situ hybridization (smFISH) combined with immunofluorescence to visualize co-localization of LSM3 with specific transcripts at the single-cell level.

Nanopore direct RNA sequencing offers exciting possibilities for detecting LSM3-dependent RNA modifications or structural changes, while cryoEM of LSM3 complexes bound to RNA substrates can provide structural insights into binding interfaces and complex assembly.

These advanced techniques require specialized equipment and expertise but provide unprecedented resolution of molecular interactions that cannot be achieved through bulk biochemical methods.

What are the most significant unanswered questions about LSM3 function in human cells?

Despite significant progress, several critical questions about LSM3 remain unanswered. The precise mechanism by which LSM3 influences splice site selection remains unclear - does it directly participate in splice site recognition or act through recruitment of other factors? The coordination between LSM3's dual roles in splicing and mRNA decay is poorly understood, particularly how these functions are balanced in different cellular contexts.

The broader question of how LSM3-containing complexes contribute to integrated RNA regulation across transcription, processing, export, and decay requires systems-level investigation. Furthermore, the potential moonlighting functions of LSM3 beyond its established roles in Lsm1-7 and Lsm2-8 complexes remain an intriguing possibility that merits exploration.

Product Science Overview

Function and Mechanism

LSM3 is involved in pre-mRNA splicing as a component of the U4/U6-U5 tri-snRNP complex, which is essential for spliceosome assembly. The spliceosome is a complex molecular machine responsible for removing introns from pre-mRNA. LSM3 is also part of the precatalytic spliceosome (spliceosome B complex) .

The heptameric LSM2-8 complex, which includes LSM3, binds specifically to the 3’-terminal U-tract of U6 snRNA. This binding is crucial for the stability and function of U6 snRNA, which is a key component of the spliceosome .

Biological Pathways

LSM3 is associated with several important biological pathways, including:

  • Processing of Capped Intron-Containing Pre-mRNA: This pathway involves the modification and splicing of pre-mRNA to produce mature mRNA.
  • Deadenylation-dependent mRNA Decay: This pathway is responsible for the degradation of mRNA molecules, which is a critical process for regulating gene expression .
Clinical Significance

Mutations or dysregulation of the LSM3 gene can be associated with various diseases. For example, LSM3 has been linked to conditions such as External Pathological Resorption and Tooth Resorption .

Research and Applications

The recombinant form of LSM3, known as Human Recombinant LSM3, is used in various research applications to study its function and role in RNA processing. This recombinant protein is produced using recombinant DNA technology, which allows for the expression of the LSM3 protein in a controlled laboratory environment .

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.