LSM4 Human

LSM4 Homolog, U6 Small Nuclear RNA Associated Human Recombinant
Shipped with Ice Packs
In Stock

Description

LSM4 Human Recombinant fused with a 20 amino acid His tag at N-terminus produced in E.Coli is a single, non-glycosylated, polypeptide chain containing 159 amino acids (1-139 a.a.) and having a molecular mass of 17.5kDa. The LSM4 is purified by proprietary chromatographic techniques.

Product Specs

Introduction
LSM4 (U6 snRNA-associated Sm-like protein) forms a seven-part ring-shaped complex involved in RNA processing. It facilitates interactions between RNA and proteins and is crucial for the structural changes needed during the assembly of ribosomal subunits. LSM4 specifically binds to the U-tract at the 3' end of U6 snRNA.
Description
Recombinant human LSM4, produced in E. coli, is a single, non-glycosylated polypeptide chain. It consists of 159 amino acids (with a 20 amino acid His tag at the N-terminus, covering amino acids 1-139), resulting in a molecular weight of 17.5 kDa. Purification is achieved through proprietary chromatographic techniques.
Physical Appearance
A clear, colorless solution, sterilized by filtration.
Formulation
The LSM4 protein is provided at a concentration of 0.5 mg/ml in a buffer consisting of 20mM Tris-HCl (pH 8.0), 20% glycerol, 0.1M NaCl, and 1mM DTT.
Stability
For short-term storage (2-4 weeks), keep at 4°C. For longer periods, freeze at -20°C. Adding a carrier protein (0.1% HSA or BSA) is recommended for long-term storage. Avoid repeated freeze-thaw cycles.
Purity
Purity is determined to be greater than 90% via SDS-PAGE analysis.
Synonyms
U6 snRNA-associated Sm-like protein LSm4, Glycine-rich protein, GRP, LSM4, YER112W.
Source
Escherichia Coli.
Amino Acid Sequence

MGSSHHHHHH SSGLVPRGSH MLPLSLLKTA QNHPMLVELK NGETYNGHLV SCDNWMNINL REVICTSRDG DKFWRMPECY IRGSTIKYLR IPDEIIDMVK EEVVAKGRGR GGLQQQKQQK GRGMGGAGRG VFGGRGRGGI PGTGRGQPEK KPGRQAGKQ.

Q&A

What is LSM4 and what is its primary function in human cells?

LSM4 is a member of the "Like-Smith" (LSM) family of RNA-binding proteins that appears in essentially all cellular organisms. The protein plays a crucial role in pre-mRNA splicing as a component of the U4/U6-U5 tri-snRNP complex involved in spliceosome assembly and functions in the precatalytic spliceosome (spliceosome B complex) . The LSM family was first discovered in a patient with systemic lupus erythematosus, where these Sm proteins are antigens targeted by anti-Sm antibodies . LSM4 consists of 139 amino acids in humans and is involved in RNA metabolism pathways . Research methodology to study LSM4 function typically includes RNA immunoprecipitation, splicing assays, and gene expression analysis following LSM4 modulation.

How does LSM4 expression differ between normal tissues and cancerous tissues?

LSM4 shows significantly higher expression levels in breast tumors and other cancer types compared to normal tissues . Analysis using The Cancer Genome Atlas (TCGA) data integrated with the cBioPortal database has demonstrated that LSM4 mRNA expression is significantly upregulated in breast cancer tissues . This pattern extends beyond breast cancer, with significant overexpression observed in adenoid cystic carcinoma, esophageal carcinoma, colon cancer, and lung adenocarcinomas . Using the UALCAN database, researchers found that LSM4 was among several LSM family members (including LSM1, LSM2, LSM3, LSM5, LSM7, LSM8, LSM10, and LSM12) that showed significantly higher expression in breast cancer tissues compared to healthy controls . To study these expression differences, researchers should employ multiple detection methods, including RNA-seq, RT-qPCR, and immunohistochemistry, with appropriate normalization to reference genes or tissues.

What techniques are most reliable for studying LSM4 protein expression in tissue samples?

For reliable LSM4 protein detection, researchers should consider multiple complementary techniques:

  • Immunohistochemistry (IHC): Enables visualization of LSM4 distribution within tissue architecture using validated antibodies. The Human Protein Atlas database contains IHC images from tissue microarrays that have been used to visualize LSM4 across different tissues .

  • Immunofluorescence (IF): Provides higher resolution and multiplexing capabilities for co-localization studies. The Human Protein Atlas includes high-resolution IF images for LSM4 detection .

  • Western Blotting: Offers semi-quantitative analysis of LSM4 protein levels with appropriate loading controls.

  • Mass Spectrometry: Provides highly specific protein identification and quantification, particularly useful for detecting post-translational modifications.

  • Proximity Ligation Assay: Enables detection of protein-protein interactions involving LSM4 in situ.

When designing experiments, researchers should validate antibody specificity, include appropriate positive and negative controls, and perform technical replicates. For quantitative comparisons, standardized scoring systems and digital image analysis should be employed to minimize subjective interpretation.

How does LSM4 expression correlate with cancer progression and patient survival outcomes?

LSM4 expression shows significant correlation with cancer progression and poorer patient survival. Research using TCGA data revealed that LSM4 expression levels were highly associated with poor prognostic outcomes in breast cancer, with a hazard ratio of 1.35 (95% confidence interval 1.21–1.51, p for trend = 3.4 × 10^-7) . Additionally, significant correlations were found between LSM4 expression and advancing tumor stages in breast cancer patients, demonstrating an upward trend as the disease progresses .

To analyze such correlations, researchers should:

  • Use Kaplan-Meier survival analysis with appropriate cutoff values for LSM4 expression

  • Employ Cox proportional hazards regression to adjust for confounding factors

  • Stratify analyses by molecular subtypes (basal, HER2, luminal A, and luminal B)

  • Validate findings across independent patient cohorts

The relationship between LSM4 and poor outcomes may be mediated through its involvement in critical cellular pathways, including "Cell cycle role of APC in cell cycle regulation" and "Immune response IL-15 signaling via MAPK and PI3K cascade" as revealed by MetaCore and GlueGo analyses .

What is the relationship between LSM4 expression and immune cell infiltration in tumors?

LSM4 expression shows intriguing correlations with immune cell infiltration in the tumor microenvironment, particularly in breast cancer. Research has demonstrated that infiltration levels of various immune cell types, including CD4+ T cells, CD8+ T cells, T-cell follicular helpers, and myeloid-derived suppressor cells, are positively correlated with LSM4 expression in several breast cancer subtypes (basal, HER2, luminal A, and luminal B) .

This relationship suggests that LSM4 may influence the tumor immune microenvironment through several possible mechanisms:

  • Modulation of cytokine or chemokine expression

  • Regulation of pathways affecting immune cell recruitment

  • Influence on tumor immunogenicity through altered RNA processing

To study this relationship, researchers should employ:

  • Multiplexed immunohistochemistry to visualize LSM4 and immune markers simultaneously

  • Flow cytometry for quantitative assessment of tumor-infiltrating lymphocytes

  • Single-cell RNA sequencing to characterize immune cell populations in relation to LSM4 expression

  • Spatial transcriptomics to map the physical relationship between LSM4-expressing cells and immune infiltrates

Understanding this relationship could have important implications for immunotherapy response prediction and combination treatment strategies targeting both LSM4 and immune checkpoints.

How might alterations in LSM4 function affect RNA splicing patterns in cancer cells?

Given LSM4's role in the spliceosome as a component of the U4/U6-U5 tri-snRNP complex , alterations in its expression or function likely impact RNA splicing patterns in cancer cells. While specific splicing changes induced by LSM4 dysregulation are not detailed in the provided search results, research approaches to investigate this question should include:

  • Transcriptome-wide splicing analysis:

    • RNA-seq with junction-focused algorithms to detect altered splicing events

    • Comparison of splice isoform ratios between LSM4-high and LSM4-low samples

    • Minigene assays to validate specific splicing changes

  • Mechanistic investigations:

    • RNA immunoprecipitation followed by sequencing (RIP-seq) to identify direct LSM4 RNA targets

    • CLIP-seq (Crosslinking and Immunoprecipitation) to map LSM4 binding sites at nucleotide resolution

    • In vitro splicing assays with purified components to assess biochemical effects of LSM4 alterations

  • Functional consequences assessment:

    • Proteomic analysis to identify altered protein isoforms resulting from LSM4-mediated splicing changes

    • Pathway analysis of genes with LSM4-dependent alternative splicing

    • Correlation of specific splicing events with phenotypic outcomes in cell models

Dysregulated splicing resulting from abnormal LSM4 activity could affect cancer-related genes, potentially promoting oncogenic isoforms that drive proliferation, survival, or metastasis.

What experimental approaches are most appropriate for studying LSM4 function in cancer models?

When designing experiments to study LSM4 function in cancer, researchers should implement a multi-faceted approach:

  • Gene expression modulation:

    • CRISPR-Cas9 knockout or knockdown to eliminate or reduce LSM4 expression

    • Inducible expression systems for controlled overexpression

    • Rescue experiments with wild-type and mutant LSM4 constructs

  • Phenotypic assessments:

    • Proliferation, migration, and invasion assays

    • Colony formation and anchorage-independent growth

    • In vivo xenograft models with LSM4-modulated cells

    • Patient-derived xenografts stratified by LSM4 expression

  • Molecular characterization:

    • RNA-seq to identify global transcriptomic changes

    • Alternative splicing analysis using junction-centric algorithms

    • Proteomics to identify altered protein expression and post-translational modifications

    • ChIP-seq or CUT&RUN to identify potential chromatin interactions

  • Pathway analysis:

    • Assessment of signaling pathways previously associated with LSM4, including "Cell cycle regulation" and "Immune response IL-15 signaling via MAPK and PI3K cascade"

    • Phosphoproteomic analysis to identify altered signaling nodes

    • Drug sensitivity profiling following LSM4 modulation

  • Translational research approaches:

    • Correlation of experimental findings with patient data

    • Development of biomarkers based on LSM4 expression or associated signatures

    • Testing combination treatments targeting LSM4-related vulnerabilities

These approaches should be implemented with appropriate controls and replicated across multiple cell lines representing different cancer subtypes to establish the generalizability of findings.

How can single-case experimental designs be applied to study LSM4-targeted interventions?

Single-case experimental designs (SCEDs) offer valuable approaches for developing personalized LSM4-targeted interventions, particularly in translational research. Based on methodological principles, the following SCED approaches can be applied :

  • Reversal designs (A-B-A-B):

    • Phase A1: Baseline measurement of LSM4 expression and related outcomes

    • Phase B1: Administration of an LSM4-targeting intervention

    • Phase A2: Withdrawal period to assess return to baseline

    • Phase B2: Reintroduction of intervention to confirm causality

    This design requires continuous monitoring with a minimum of 5 data points per phase, with stability established within each phase (data points falling within a 15% range of the median) .

  • Multiple baseline designs:

    • Implementation of LSM4-targeting interventions across different patient-derived samples

    • Staggered introduction of treatment to establish experimental control

    • Continuous measurement across all samples regardless of treatment status

    • Demonstration of change only following intervention initiation

  • Combined designs:

    • Integration of reversal and multiple baseline approaches

    • Randomization of intervention order when possible to reduce bias

    • Blinding of intervention and data collection phases when feasible

For N-of-1 trials with individual patients, LSM4 expression could be monitored through serial biopsies or liquid biopsies, with treatment adjusted based on response. Results from multiple N-of-1 trials could then be aggregated using meta-analytic techniques to establish generalizable findings .

What statistical approaches should be used when analyzing LSM4 expression data across different patient cohorts?

When analyzing LSM4 expression data across different patient cohorts, researchers should employ robust statistical approaches that account for cohort heterogeneity and potential confounding factors:

How should researchers interpret seemingly contradictory findings about LSM4 expression or function?

When confronting contradictory findings regarding LSM4, researchers should implement a systematic approach to interpretation:

  • Methodological differences assessment:

    • Examine expression measurement platforms (microarray vs. RNA-seq vs. qPCR)

    • Compare antibody specificity and protocols for protein detection

    • Evaluate data normalization approaches

    • Consider cutoff thresholds used to define "high" vs. "low" expression

  • Sample and cohort characteristic analysis:

    • Assess patient demographics and clinical characteristics

    • Compare cancer subtypes representation across studies

    • Evaluate treatment history of included patients

    • Consider sample collection, preservation, and processing methods

  • Biological context consideration:

    • Examine cancer type and molecular subtype specificity

    • Consider tumor microenvironment differences

    • Assess temporal variations during disease progression

    • Evaluate potential splice variant or isoform-specific effects

  • Integration strategies:

    • Perform meta-analysis with random-effects models to account for heterogeneity

    • Stratify results by relevant clinical or molecular features

    • Employ Bayesian methods to integrate evidence from multiple sources

    • Conduct sensitivity analyses using varying thresholds

  • Validation approaches:

    • Design experiments to directly test competing hypotheses

    • Use orthogonal measurement techniques on the same samples

    • Employ functional studies to determine biological relevance

Researchers should view discrepancies as opportunities to generate refined hypotheses about context-dependent functions of LSM4 rather than simply contradictions requiring resolution in favor of one interpretation.

What techniques can help identify the molecular mechanisms through which LSM4 influences cancer progression?

To elucidate the molecular mechanisms by which LSM4 influences cancer progression, researchers should employ a comprehensive toolkit of molecular and cellular techniques:

  • Transcriptome analysis:

    • RNA-seq to identify genes differentially expressed after LSM4 modulation

    • rMATS or similar tools to detect alterations in alternative splicing

    • GSEA for pathway enrichment analysis

    • Time-course experiments to identify primary vs. secondary effects

  • Protein-RNA interaction mapping:

    • CLIP-seq (Cross-Linking Immunoprecipitation) to identify direct RNA targets

    • RNA immunoprecipitation (RIP) to isolate LSM4-bound transcripts

    • RNA-protein interaction prediction algorithms combined with experimental validation

    • In vitro binding assays to determine binding specificity and affinity

  • Protein interaction network analysis:

    • Immunoprecipitation followed by mass spectrometry

    • Proximity labeling techniques (BioID, APEX) to identify neighboring proteins

    • Yeast two-hybrid screening for binary interactions

    • Co-localization studies using super-resolution microscopy

  • Functional genomics:

    • CRISPR screens to identify synthetic lethal interactions with LSM4

    • Rescue experiments with wild-type vs. mutant LSM4

    • Domain mapping to identify crucial regions for LSM4 function

    • Epistasis experiments to position LSM4 within signaling pathways

  • Signaling pathway investigation:

    • Phosphoproteomic analysis following LSM4 modulation

    • Western blotting for key pathway components

    • Reporter assays for pathway activation

    • Small molecule inhibitors to test pathway dependencies

Based on existing data, key pathways to investigate include "Cell cycle role of APC in cell cycle regulation" and "Immune response IL-15 signaling via MAPK and PI3K cascade," which have been associated with LSM4 through computational analyses .

How can multi-omics approaches advance our understanding of LSM4 biology?

Multi-omics approaches offer powerful strategies to comprehensively understand LSM4 biology across multiple molecular levels:

  • Integrated genomics and transcriptomics:

    • Correlation of LSM4 copy number alterations with expression changes

    • Identification of cis and trans genetic modulators of LSM4 expression

    • eQTL analysis to map genetic variants affecting LSM4 regulation

    • Integration of DNA methylation data to assess epigenetic regulation

  • Transcriptomics and proteomics integration:

    • Correlation of LSM4 mRNA levels with protein abundance

    • Identification of post-transcriptional regulation mechanisms

    • Analysis of altered protein isoforms resulting from LSM4-dependent splicing

    • Assessment of changes in protein complex formation

  • Proteomics and metabolomics coordination:

    • Mapping metabolic pathway alterations following LSM4 modulation

    • Identification of post-translational modifications affected by metabolic changes

    • Characterization of energy metabolism shifts in LSM4-high vs. LSM4-low cells

  • Spatial multi-omics approaches:

    • Integration of spatial transcriptomics with protein imaging

    • Mapping of LSM4 expression patterns in relation to tumor architecture

    • Assessment of tumor-stroma interactions in regions with varying LSM4 levels

    • Correlation with immune cell infiltration patterns

  • Temporal multi-omics:

    • Time-resolved analyses after LSM4 perturbation

    • Determination of immediate vs. delayed effects

    • Inference of causal relationships between molecular changes

    • Modeling of dynamic responses to therapy in relation to LSM4 status

  • Computational integration frameworks:

    • Network-based approaches to integrate multiple data types

    • Machine learning models to predict LSM4-dependent phenotypes

    • Systems biology modeling of pathways influenced by LSM4

    • Causal inference methods to establish directionality of effects

A comprehensive multi-omics approach would allow researchers to bridge the gap between LSM4's molecular function in RNA splicing and its clinical associations with cancer progression and immune infiltration, potentially revealing novel intervention points for therapeutic development.

What are promising therapeutic strategies targeting LSM4 in cancer?

Based on current understanding of LSM4 biology, several therapeutic strategies show promise for cancer treatment:

  • Direct LSM4 inhibition:

    • Small molecule inhibitors targeting LSM4's RNA-binding domains

    • Antisense oligonucleotides or siRNAs for LSM4 knockdown

    • Proteolysis-targeting chimeras (PROTACs) for selective LSM4 degradation

    • Peptide inhibitors disrupting LSM4 interactions with other spliceosome components

  • Targeting LSM4-dependent splicing events:

    • Splice-switching oligonucleotides to modulate specific events downstream of LSM4

    • Small molecules targeting LSM4-regulated exons or introns

    • Development of synthetic lethal approaches with other splicing factors

  • Exploiting immune correlations:

    • Combination therapies with immune checkpoint inhibitors, given LSM4's association with immune cell infiltration in multiple breast cancer subtypes

    • Vaccines targeting LSM4-derived neoantigens

    • Adoptive cell therapies directed against LSM4-expressing cells

  • Biomarker-guided approaches:

    • Patient stratification based on LSM4 expression levels

    • Monitoring of LSM4-dependent splicing events as response biomarkers

    • Development of companion diagnostics for LSM4-targeting therapies

  • Precision medicine implementation:

    • Single-case experimental designs to optimize LSM4-targeted treatments for individual patients

    • N-of-1 trials with LSM4 inhibitors in patients with LSM4-overexpressing tumors

    • Adaptive trial designs incorporating LSM4 status and related biomarkers

These approaches are particularly promising for breast cancer, where LSM4 overexpression correlates with poor survival outcomes , but could potentially extend to other cancer types where LSM4 dysregulation is observed.

What emerging technologies could advance LSM4 research in the next five years?

Several cutting-edge technologies are poised to significantly advance LSM4 research:

  • Advanced RNA-protein interaction technologies:

    • Enhanced CLIP-seq variants for single-nucleotide resolution of binding sites

    • RNA-BioID for comprehensive mapping of the RNA neighborhood of LSM4

    • Live-cell imaging of LSM4-RNA interactions using MS2 or similar systems

    • Nanopore direct RNA sequencing to identify LSM4-dependent RNA modifications

  • Structural biology innovations:

    • Cryo-electron microscopy to visualize LSM4 within spliceosome complexes

    • Integrative structural approaches combining multiple data types

    • Time-resolved structural studies to capture dynamic conformational changes

    • AlphaFold and similar AI platforms for structure prediction of LSM4 complexes

  • Spatial biology advances:

    • Multiplexed ion beam imaging (MIBI) to visualize LSM4 and dozens of other proteins simultaneously

    • Spatial transcriptomics at single-cell resolution

    • 3D tissue mapping of LSM4 expression patterns

    • Correlative light and electron microscopy for ultrastructural context

  • Single-cell multi-omics:

    • Integrated scRNA-seq, scATAC-seq, and scProteomics

    • Single-cell splicing analysis to detect LSM4-dependent events

    • Trajectory inference to map LSM4's role in cellular state transitions

    • Spatial single-cell technologies to preserve tissue context

  • Advanced functional genomics:

    • Base editing or prime editing for precise LSM4 modification

    • CRISPR screens with single-cell readouts for high-resolution phenotyping

    • Perturb-seq to link genetic perturbations with transcriptional responses

    • Combinatorial genetic screens to identify synthetic interactions

  • Artificial intelligence applications:

    • Deep learning for prediction of LSM4-dependent splicing outcomes

    • Multi-modal data integration using graph neural networks

    • Automated image analysis for LSM4 localization studies

    • AI-driven drug discovery targeting LSM4 or its interaction partners

These technologies will enable researchers to move beyond correlative observations to mechanistic understanding of LSM4's roles in RNA metabolism and cancer biology, potentially accelerating therapeutic development.

What controls and validation steps are essential when studying LSM4 in experimental models?

Rigorous controls and validation steps are critical for LSM4 research integrity:

  • Gene expression modulation validation:

    • Confirmation of knockdown/knockout efficiency at both mRNA and protein levels

    • Rescue experiments to verify phenotype specificity

    • Use of multiple independent siRNAs/shRNAs to rule out off-target effects

    • Verification of CRISPR editing via sequencing

  • Antibody validation:

    • Confirmation of specificity using knockout/knockdown controls

    • Western blot demonstration of expected molecular weight band

    • Comparison of multiple antibodies targeting different epitopes

    • Peptide competition assays to verify binding specificity

  • Cell line authentication:

    • Regular STR profiling to confirm identity

    • Mycoplasma testing

    • Passage number tracking and limitation

    • Use of multiple cell lines to demonstrate reproducibility

  • Experimental design considerations:

    • Inclusion of appropriate positive and negative controls

    • Randomization procedures to minimize bias

    • Blinded assessment of outcomes when possible

    • Technical and biological replicates with appropriate statistical analysis

  • Phenotype validation:

    • Use of complementary assays measuring the same phenotype

    • Time course experiments to establish causality

    • Dose-response relationships for pharmacological interventions

    • In vivo validation of key in vitro findings

  • Clinical correlation validation:

    • Use of multiple independent patient cohorts

    • Stratification by relevant clinical and molecular factors

    • Application of appropriate statistical methods with multiple testing correction

    • External validation in prospective studies when possible

These validation steps ensure that findings regarding LSM4's functions and clinical relevance are robust and reproducible across different experimental contexts.

How should researchers address the challenges of studying LSM4 in heterogeneous tumor samples?

Tumor heterogeneity presents significant challenges for LSM4 research that require specialized approaches:

  • Sampling strategies:

    • Multi-region sampling to capture spatial heterogeneity

    • Longitudinal sampling to address temporal changes

    • Matched primary and metastatic samples for comparison

    • Integration of normal adjacent tissue controls

  • Single-cell approaches:

    • scRNA-seq to resolve expression patterns in distinct cell populations

    • Single-cell proteomics for protein-level assessment

    • Computational deconvolution methods for bulk samples

    • Cell type-specific markers to identify LSM4-expressing populations

  • Spatial analysis techniques:

    • Multiplexed immunofluorescence to visualize LSM4 alongside cell type markers

    • Digital spatial profiling for high-plex protein and RNA analysis

    • Laser capture microdissection for region-specific molecular profiling

    • In situ hybridization for LSM4 mRNA localization

  • Computational deconvolution:

    • Reference-based methods using known cell type signatures

    • Reference-free approaches to identify cell populations

    • Estimation of cellular proportions and their association with LSM4 levels

    • Integration with immune infiltration data, given LSM4's correlation with immune cells

  • Patient-derived models:

    • Organoid cultures preserving tumor heterogeneity

    • Patient-derived xenografts established from different tumor regions

    • Co-culture systems with multiple cell types

    • Ex vivo tissue slice cultures maintaining original architecture

  • Data integration approaches:

    • Correlation of bulk and single-cell data

    • Integration of genomic, transcriptomic, and proteomic profiles

    • Machine learning methods to identify patterns across heterogeneous samples

    • Network analysis to identify consistent LSM4-associated pathways despite heterogeneity

By implementing these strategies, researchers can obtain a more complete understanding of LSM4's varied roles across different cell populations within tumors, potentially identifying the specific contexts where targeting LSM4 would be most effective.

Product Science Overview

Structure and Function

LSM4 is part of the heptameric LSM2-8 complex, which binds specifically to the 3’-terminal oligo(U) tract of U6 small nuclear RNA (snRNA). This binding is essential for the stability and function of U6 snRNA, a critical component of the spliceosome . The spliceosome is a complex molecular machine responsible for removing introns from pre-mRNA, a process known as splicing .

The LSM2-8 complex, including LSM4, is involved in the formation of the U4/U6-U5 tri-snRNP complex, which is a key component of the spliceosome assembly. This complex plays a significant role in the pre-mRNA splicing process by mediating the formation of the spliceosome’s catalytic core .

Biological Pathways

LSM4 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 molecules that can be translated into proteins .
  • Deadenylation-Dependent mRNA Decay: This pathway is responsible for the degradation of mRNA molecules, which is crucial for regulating gene expression and maintaining cellular homeostasis .
Clinical Significance

Mutations or dysregulation of LSM4 have been linked to various diseases, including:

  • Spindle Cell Thymoma: A rare type of tumor that arises from the thymus gland .
  • Spinal Muscular Atrophy: A genetic disorder characterized by the loss of motor neurons, leading to muscle weakness and atrophy .
Research and Applications

The recombinant form of LSM4, known as “LSM4 Homolog, U6 Small Nuclear RNA Associated (Human Recombinant),” is used in various research applications to study its function and role in RNA metabolism. This recombinant protein is produced using recombinant DNA technology, which allows for the expression of the human LSM4 gene in a host organism, such as bacteria or yeast .

Researchers use this recombinant protein to investigate the molecular mechanisms underlying RNA processing and degradation, as well as to develop potential therapeutic strategies for diseases associated with LSM4 dysfunction .

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.