SRP19 Human

Signal Recognition Particle 19kDa Human Recombinant
Shipped with Ice Packs
In Stock

Description

Overview of SRP19 Human

SRP19 (Signal Recognition Particle 19 kDa) is a human protein encoded by the SRP19 gene (NCBI Gene ID: 6728) located on chromosome 5q21-q22 . It is a core component of the signal recognition particle (SRP), a ribonucleoprotein complex essential for co-translational targeting of secretory and membrane proteins to the endoplasmic reticulum (ER) . SRP19 facilitates SRP assembly by binding 7S RNA and mediating SRP54 subunit recruitment, ensuring proper protein trafficking . Genetic defects in SRP19 are linked to Severe Congenital Neutropenia (SCN) and Shwachman-Diamond Syndrome 1 (SDS1) .

Key Features of SRP19 Protein

PropertyDetailsSource
Molecular Weight18.5 kDa (recombinant form, His-tagged) Assay Genie
DomainsSingle domain with βαββα topology; binds GGAG tetraloop of SRP RNA helix 6 Frontiers
Binding Partners7S RNA, SRP54, ribosome GeneCards
Cellular LocalizationNucleolus, nucleoplasm, cytosol, signal recognition particle UniProt

SRP19 stabilizes the SRP S-domain by inducing a closed conformation between helices 6 and 8 of 7S RNA, enabling SRP54 recruitment . This interaction is critical for signal sequence recognition and GTP-dependent docking with the SRP receptor .

Disorders Linked to SRP19 Mutations

DiseasePathogenic MechanismKey Findings
Severe Congenital NeutropeniaImpaired granulocyte differentiation due to proteome dysregulationGlobal proteome aberrations in neutrophil granulocytes
Shwachman-Diamond Syndrome 1Defective protein targeting and ER stressAssociated with bone marrow failure and pancreatic dysfunction

Recent studies identified SRP19 variants in patients with SCN, revealing disrupted SRP-dependent protein homeostasis and neutrophil maturation defects . In vitro differentiation of human iPSCs and zebrafish models confirmed these findings .

Key Studies on SRP19

  1. CRISPR Screening

    • SRP19 is a core essential gene; knockout causes cell death in all 1,019 tested cancer cell lines .

    • Partial SRP19 suppression correlates with sensitivity in cancers exhibiting APC/SRP19 loss (e.g., colorectal cancer) .

  2. Proteomic Profiling

    • SRP19-deficient cells show dysregulation of ribosomal proteins and translation machinery .

    • SRP19 dysfunction alters stress granule dynamics, linking it to cellular stress responses .

  3. Therapeutic Targeting

    • siRNA-mediated SRP19 knockdown reduces proliferation in APC/SRP19-deficient cancers, validated via rescue experiments .

    • SRP19 expression levels predict sensitivity to ribosomal stress-inducing agents .

Recombinant SRP19 Protein (Human)

ParameterDetails
SourceEscherichia coli-expressed, His-tagged (N-terminus)
Purity>95% via proprietary chromatography
Buffer20 mM Tris-HCl (pH 8.0), 0.2 M NaCl, 50% glycerol, 2 mM DTT
ApplicationsStructural studies, SRP assembly assays, drug screening

Future Directions

  • Mechanistic Studies: Elucidate SRP19’s role in stress granule regulation and viral RNA packaging .

  • Therapeutic Development: Explore SRP19 inhibitors for cancers with APC/SRP19 co-deletions .

  • Diagnostic Biomarkers: Validate SRP19-associated proteome signatures for early detection of SCN/SDS1 .

Product Specs

Introduction
Signal recognition particle 19kDa protein (SRP19) is a protein belonging to the SRP19 family. It plays a crucial role in protein targeting by binding directly to 7S RNA. This interaction facilitates the binding of the 54 kDa subunit of the signal recognition particle (SRP).
Description
SRP19 Human Recombinant is a genetically engineered protein produced in E. coli. It consists of a single, non-glycosylated polypeptide chain containing 167 amino acids (with amino acids 1-144 representing the SRP19 sequence) and a 23 amino acid His tag at the N-terminus. This recombinant protein has a molecular weight of 18.5 kDa and is purified using proprietary chromatographic techniques.
Physical Appearance
Clear, colorless solution that has been sterilized through filtration.
Formulation
The SRP19 solution is provided at a concentration of 0.25 mg/ml in a buffer consisting of 20 mM Tris-HCl (pH 8.0), 0.2 M NaCl, 50% glycerol, and 2 mM DTT.
Stability
For short-term storage (up to 2-4 weeks), the SRP19 solution should be kept refrigerated at 4°C. For long-term storage, it is recommended to store the solution at -20°C. To ensure optimal stability during long-term storage, consider adding a carrier protein such as HSA or BSA at a concentration of 0.1%. Repeated freezing and thawing of the solution should be avoided.
Purity
The purity of the SRP19 Human Recombinant is greater than 90%, as determined by SDS-PAGE analysis.
Synonyms
Signal recognition particle 19kDa protein, SRP19.
Source
Escherichia Coli.
Amino Acid Sequence
MGSSHHHHHH SSGLVPRGSH MGSMACAAAR SPADQDRFIC IYPAYLNNKK TIAEGRRIPI SKAVENPTAT EIQDVCSAVG LNVFLEKNKM YSREWNRDVQ YRGRVRVQLK QEDGSLCLVQ FPSRKSVMLY AAEMIPKLKT RTQKTGGADQ SLQQGEGSKK GKGKKKK.

Q&A

What is SRP19 and what is its primary function in human cells?

SRP19 is a 19 kDa protein subunit of the Signal Recognition Particle (SRP), which is essential for the cotranslational targeting of secretory and membrane proteins to the endoplasmic reticulum (ER) . The primary function of SRP19 within this complex is to bind directly to 7SL RNA and mediate the binding of SRP54 to the SRP complex . This binding creates a crucial structural change in the 7SL RNA that enables SRP54 association, which is essential for signal sequence recognition and subsequent protein targeting .

Experimental evidence demonstrates that SRP19 functions as a rate-limiting component for the formation of the entire Signal Recognition Particle . This is evident from studies showing that SRP54 stability is dependent on SRP19 protein levels, and excess unbound SRP54 is degraded by the proteasome in cells with APC/SRP19 loss . Furthermore, proteasome inhibitor experiments with Bortezomib have confirmed that SRP54 protein not bound to the 7SL complex in APC/SRP19 loss cells undergoes proteasomal degradation .

How is SRP19 transported within human cells and what is its subcellular localization?

SRP19 exhibits a complex pattern of subcellular localization and transport. Despite the SRP complex functioning primarily in the cytoplasm, SRP19 is efficiently imported into the nucleus and nucleolus . Its nuclear import is mediated by two members of the importin beta superfamily of transport receptors: importin 8 and transportin . SRP19 is also imported less efficiently by several other members of the importin beta family .

To study SRP19 localization experimentally, researchers have used immunofluorescence staining techniques with specific antibodies against SRP proteins. In mouse cells (NIH 3T3), staining for SRP19 shows a strongly reticular pattern, consistent with its association with the SRP complex . Unlike SRP9/14, which localizes to stress granules under arsenite treatment, SRP19 does not enrich in these granular structures during cellular stress response . This differential behavior under stress conditions suggests distinct roles for various SRP components beyond their canonical function in the SRP complex.

What are the most effective methods for detecting and quantifying SRP19 in human tissue samples?

For detecting and quantifying SRP19 in human tissue samples, researchers commonly employ a combination of complementary techniques:

Immunohistochemistry and Immunofluorescence:
These techniques utilize specific antibodies against SRP19 for visualization in fixed tissues. For improved detection specificity, acetone/methanol fixation may yield better results than formaldehyde fixation, as evidenced by differential detection of SRP components in nucleoli depending on fixation method . Dual staining with markers for specific cellular compartments (e.g., nucleolar markers) can confirm subcellular localization.

Western Blotting:
For quantitative assessment of SRP19 protein levels, western blotting with specific antibodies is the standard approach. Researchers should note that SRP19 detection by antibodies may not be linear over a sufficiently large concentration range, which can complicate precise quantification .

PCR-Based Detection:
For genetic analysis of SRP19, PCR techniques have been successfully employed. For instance, the SRP19 variant described in research results in a PCR product of only 209bp (compared to the wild-type product) . RT-qPCR can be used for mRNA quantification.

Proteomics Approaches:
Mass spectrometry-based proteomics offers an unbiased approach to detect and quantify SRP19 along with associated proteins. This approach has been particularly valuable in studies examining proteome changes in neutrophil granulocytes from patients with variants in SRP genes .

How does SRP19 participate in the assembly of the Signal Recognition Particle complex?

SRP19 plays a critical role in the hierarchical assembly of the Signal Recognition Particle complex. The assembly process follows a specific sequence where SRP19 binding to the 7SL RNA induces a structural change that is necessary for the subsequent binding of SRP54 . This sequential assembly mechanism ensures proper formation of the functional SRP complex.

Experimentally, the importance of SRP19 in SRP assembly has been demonstrated through targeted knockdown studies. When SRP19 is depleted, there is a corresponding decrease in SRP54 protein levels (but not mRNA levels), indicating post-translational regulation . Further investigation revealed that excess SRP54 protein not bound to the 7SL complex in cells with reduced SRP19 is actively degraded by the proteasome . This was confirmed by treating cells with the proteasome inhibitor Bortezomib, which stabilized SRP54 protein levels specifically in APC/SRP19 loss cells .

The assembly sequence can be methodologically studied through in vitro reconstitution experiments, RNA-protein binding assays, and structural biology approaches such as cryo-electron microscopy that can capture intermediate states in the assembly process.

What experimental approaches can identify SRP19-dependent proteins in the secretory pathway?

Several experimental approaches can be employed to identify SRP19-dependent proteins in the secretory pathway:

Proteomics Analysis of SRP19 Depletion:
Comparative proteomics of cells with normal versus reduced SRP19 levels can identify proteins whose abundance is affected by SRP19 function. This approach has been successfully used to characterize proteome aberrations in neutrophil granulocytes from patients with variants in SRP genes . The methodology involves:

  • Isolating cellular fractions (membrane, secreted, cytosolic)

  • Quantitative mass spectrometry analysis (e.g., TMT labeling or SILAC)

  • Bioinformatic analysis to identify proteins with signal peptides affected by SRP19 depletion

Pulse-Chase Experiments:
To specifically track newly synthesized secretory proteins:

  • Metabolic labeling with radioactive amino acids (e.g., 35S-methionine)

  • SRP19 knockdown or mutation

  • Immunoprecipitation of specific secretory proteins

  • Analysis of protein maturation and trafficking kinetics

Ribosome Profiling:
This technique can identify transcripts whose translation is affected by SRP19 deficiency:

  • Cell treatment with translation elongation inhibitors

  • Isolation of ribosome-protected mRNA fragments

  • Deep sequencing to identify ribosome positioning on mRNAs encoding secretory proteins

  • Comparison between normal and SRP19-depleted conditions

What is the relationship between SRP19 expression levels and cellular stress responses?

The relationship between SRP19 expression levels and cellular stress responses appears multifaceted. While SRP19 itself does not localize to stress granules during arsenite-induced stress (unlike SRP9/14) , alterations in SRP19 levels can induce cellular stress through impaired protein secretion and ER stress pathways.

Research indicates that partial loss of SRP19 creates context-specific vulnerabilities, particularly in cancer cells that already have reduced levels of SRP19 . The heightened sensitivity of these cells suggests that lower baseline SRP19 expression reduces cellular capacity to handle additional stresses.

The methodology to study this relationship includes:

  • Controlled modulation of SRP19 expression using inducible shRNA or CRISPR systems

  • Assessment of stress pathway activation (UPR markers such as XBP1 splicing, ATF6 cleavage, and PERK phosphorylation)

  • Measurement of cell viability under various stress conditions (e.g., tunicamycin, thapsigargin, or glucose deprivation)

  • Evaluation of protein synthesis and secretion rates in response to SRP19 modulation

How do genetic defects in SRP19 contribute to severe congenital neutropenia?

Genetic defects in SRP19 have been identified as causative factors in severe congenital neutropenia through comprehensive genetic and functional studies . A specific homozygous mutation in SRP19 was identified in related pedigrees with 5 affected patients, establishing a clear genotype-phenotype correlation .

The pathogenic mechanism involves disruption of the SRP-dependent protein processing pathway, which is critical for neutrophil granulocyte differentiation . Experimental approaches to understand this mechanism include:

  • Proteome analysis: Studies of neutrophil granulocytes from patients with SRP19 variants revealed specific proteome aberrations . This approach identifies both global changes and alterations in specific proteins essential for neutrophil function.

  • In vitro differentiation models: Human induced pluripotent stem cell (iPSC) models allow for the study of neutrophil differentiation under SRP19 deficiency . These systems can recapitulate the developmental defects observed in patients.

  • In vivo zebrafish models: Zebrafish models of SRP deficiency enable the investigation of neutrophil development in a whole organism context . These models provide insights into how SRP19 dysfunction affects neutrophil development and function in vivo.

  • Electron microscopy studies: Analysis of neutrophil granulocytes from patients shows significant reduction of electron-dense granules , indicating defects in proper protein trafficking and organelle formation.

What are the molecular signatures of SRP19 dysfunction in patient-derived neutrophils?

The molecular signatures of SRP19 dysfunction in patient-derived neutrophils have been characterized through detailed proteomic analysis . These signatures include:

  • Reduced granule proteins: Patients with SRP19 defects show decreased levels of specific neutrophil granule proteins, reflecting defects in protein processing and trafficking required for granule formation .

  • Altered secretory pathway proteins: The proteome of affected neutrophils shows aberrations in components of the secretory pathway, consistent with the role of SRP19 in cotranslational targeting .

  • Compensatory changes: Some proteins show increased expression, potentially representing cellular attempts to compensate for SRP dysfunction .

  • Myeloid maturation arrest: Bone marrow analysis of patients with SRP19 defects shows characteristic myeloid maturation arrest , indicating a critical role for SRP19 in neutrophil differentiation.

These molecular signatures can be studied methodologically through:

  • Comparative proteomics of patient vs. healthy neutrophils using mass spectrometry

  • Immunoblotting for key neutrophil proteins

  • Flow cytometry to assess neutrophil maturation markers

  • Functional assays measuring neutrophil degranulation and respiratory burst capacity

How can SRP19 deficiency be modeled in experimental systems for therapeutic development?

Modeling SRP19 deficiency for therapeutic development requires a multi-system approach that captures the complexity of SRP-dependent protein processing. Successful experimental systems include:

1. Inducible knockdown cellular systems:

  • Doxycycline-inducible shRNA expression systems allow for controlled, partial suppression of SRP19 expression

  • This approach enables titration of SRP19 levels to mimic the partial deficiency seen in patients

  • Can be applied in relevant cell types including myeloid progenitor cells

2. Human iPSC-derived neutrophil models:

  • Patient-derived or genetically engineered iPSCs with SRP19 mutations

  • Differentiation protocols to generate neutrophil precursors and mature neutrophils

  • Assessment of differentiation efficiency, morphology, and functional characteristics

3. Zebrafish models:

  • CRISPR/Cas9-mediated mutation of srp19 in zebrafish

  • Analysis of neutrophil development using transgenic lines with fluorescently labeled neutrophils

  • In vivo assessment of neutrophil function and migration

4. Mouse xenograft models:

  • Immuno-deficient mice injected with cells containing inducible SRP19 knockdown

  • Allows for in vivo assessment of SRP19 function in a complex physiological context

  • Can be used to test therapeutic interventions

These models provide complementary information on different aspects of SRP19 function and can be used to screen for compounds that might rescue SRP19 deficiency phenotypes through:

  • High-throughput small molecule screens

  • Gene therapy approaches

  • Targeted protein degradation modulation

  • ER stress pathway modulation

How can evolutionary conservation of SRP19 inform structure-function studies?

Evolutionary conservation analysis of SRP19 provides valuable insights for structure-function studies. SRP19 is highly conserved across species, reflecting its essential role in the SRP complex. Methodological approaches to leverage this conservation include:

Comparative Sequence Analysis:

  • Multiple sequence alignment of SRP19 proteins from diverse species

  • Identification of invariant residues that may be critical for function

  • Detection of co-evolving residues that maintain structural or functional relationships

Structure-Guided Mutagenesis:
Based on conservation data, researchers can design targeted mutations to test specific hypotheses:

  • Mutations of highly conserved residues involved in RNA binding

  • Alterations to residues at protein-protein interfaces (particularly with SRP54)

  • Engineering of chimeric proteins with domains from different species to test functional compatibility

Heterologous Expression Systems:
To test functional conservation:

  • Expression of SRP19 from different species in human cells with SRP19 knockdown

  • Assessment of rescue efficiency for SRP complex formation and function

  • Identification of species-specific differences in SRP19 activity

These approaches can identify functional domains and critical residues that could serve as targets for therapeutic intervention or as sites for engineering SRP19 variants with enhanced or modified functions.

What are the specific roles of nuclear and cytoplasmic pools of SRP19 in human cells?

SRP19 exhibits a distinct subcellular distribution with significant pools in both the nucleus/nucleolus and the cytoplasm . Understanding the specific roles of these different pools requires sophisticated experimental approaches:

Subcellular Fractionation and Proteomics:

  • Separation of nuclear, nucleolar, and cytoplasmic fractions

  • Identification of SRP19-interacting partners in each compartment using immunoprecipitation followed by mass spectrometry

  • Comparison of post-translational modifications of SRP19 in different cellular compartments

Domain-Specific Targeting:

  • Engineering of SRP19 variants with modified nuclear localization or export signals

  • Creation of cell lines expressing these variants in an endogenous SRP19-depleted background

  • Assessment of SRP assembly, protein secretion, and cellular stress responses

Live-Cell Imaging:

  • Generation of fluorescently tagged SRP19 expressed at near-endogenous levels

  • Photoactivation or photobleaching experiments to track protein movement between compartments

  • Correlation of SRP19 dynamics with cell cycle phases or stress responses

Compartment-Specific Depletion:

  • Development of systems for induced protein degradation specifically in the nucleus or cytoplasm

  • Analysis of the immediate and long-term consequences of compartment-specific SRP19 depletion

  • Rescue experiments with compartment-restricted SRP19 variants

These methodologies can reveal whether nuclear SRP19 serves primarily in SRP assembly, has independent functions, or represents a storage pool for regulating cytoplasmic SRP19 levels.

How do post-translational modifications regulate SRP19 function in different cellular contexts?

Post-translational modifications (PTMs) likely play significant roles in regulating SRP19 function, although this area remains relatively unexplored compared to other aspects of SRP biology. A methodological framework for studying SRP19 PTMs includes:

PTM Mapping:

  • Immunoprecipitation of endogenous SRP19 from different cell types or conditions

  • Mass spectrometry analysis focusing on PTMs (phosphorylation, ubiquitination, acetylation, etc.)

  • Quantitative comparison of PTM patterns across cellular contexts (e.g., differentiation states, stress conditions)

Site-Directed Mutagenesis:

  • Generation of SRP19 variants with mutations at identified PTM sites (e.g., phospho-mimetic or phospho-null mutations)

  • Expression in SRP19-depleted backgrounds to assess functional consequences

  • Analysis of effects on SRP assembly, subcellular localization, and protein stability

PTM Enzyme Identification:

  • Candidate approach testing known kinases, acetylases, or other modifying enzymes

  • CRISPR screens for enzymes affecting SRP19 function or stability

  • In vitro modification assays with purified components

Context-Specific Analysis:

  • Comparison of SRP19 PTMs during neutrophil differentiation versus other cell types

  • Analysis of PTM changes during cell stress responses

  • Correlation of PTM patterns with SRP19 function in disease contexts

This research area holds particular promise for identifying regulatory mechanisms that could be exploited therapeutically in conditions with SRP19 dysfunction.

How might SRP19 be exploited as a therapeutic target in cancer?

Recent findings suggest SRP19 could serve as a promising therapeutic target, particularly in cancers with specific genetic backgrounds. Research has demonstrated that partial SRP19 inhibition creates a vulnerability in cancers with APC/SRP19 loss, while sparing normal cells . This therapeutic window can be exploited through several approaches:

Target Identification and Validation:

  • Comprehensive screening across cancer cell lines to identify those with SRP19 dependency

  • Correlation of SRP19 dependency with genetic markers (e.g., APC status) to define patient populations

  • In vivo validation using xenograft models with inducible SRP19 knockdown systems

Therapeutic Approaches:

  • RNA interference-based therapeutics targeting SRP19

  • Small molecule inhibitors of SRP19-7SL RNA interaction

  • Degrader technologies (PROTACs) targeting SRP19 protein

  • Combination strategies with proteasome inhibitors or ER stress inducers

Predictive Biomarkers:

  • Development of assays to measure SRP19 and SRP54 protein levels in tumors

  • Genetic testing for APC/SRP19 loss as companion diagnostics

  • Functional tests to assess SRP pathway activity in patient-derived samples

The experimental evidence supporting this approach includes the demonstration that DOX-inducible partial suppression of SRP19 in xenograft models significantly reduced tumor growth specifically in APC/SRP19 loss tumors (GP2D) but not in APC/SRP19 neutral tumors (RKO) . This context-specific vulnerability provides a strong rationale for therapeutic development.

What are the implications of SRP19 research for understanding other ribonucleoprotein complex-related diseases?

SRP19 research provides valuable insights into the broader category of ribonucleoprotein (RNP) complex-related diseases. The methodological approaches and conceptual frameworks can be applied to understanding similar disorders:

Comparative Disease Analysis:

  • Systematic comparison of neutrophil defects in SRP19-related neutropenia with other ribonucleoprotein-related disorders

  • Identification of common pathogenic mechanisms across different RNP complex diseases

  • Development of unified models for how RNP complex dysfunction leads to tissue-specific pathology

Translational Research Applications:

  • Development of patient-derived cellular models for various RNP-related diseases

  • High-throughput screening platforms adaptable to different RNP complexes

  • Biomarker discovery strategies applicable across RNP disorders

Therapeutic Strategy Development:

  • RNA stabilization approaches that might be applicable to multiple RNP diseases

  • Protein replacement strategies for various RNP complex components

  • Small molecule screening methods to identify compounds that enhance RNP assembly or function

The specific insights from SRP19 research that are most relevant to other RNP diseases include:

  • Understanding how partial loss of function creates context-specific vulnerabilities

  • Approaches to modulate RNP assembly through targeting rate-limiting components

  • Methods to characterize proteome changes resulting from RNP dysfunction

  • Strategies for modeling complex RNP disorders in various experimental systems

How might advances in protein trafficking research influence our understanding of SRP19 function?

Emerging technologies and concepts in protein trafficking research offer new opportunities to deepen our understanding of SRP19 function:

Single-Molecule Imaging Approaches:

  • Development of methods to visualize individual SRP complexes during protein targeting

  • Real-time tracking of SRP19 during the targeting cycle

  • Analysis of the kinetics and dynamics of SRP assembly in living cells

Organelle-Specific Proteomics:

  • Proximity labeling techniques to identify proteins in the vicinity of SRP19 in different cellular compartments

  • Targeted analysis of the ER translocon neighborhood during active translation

  • Temporal analysis of proteome changes during SRP complex assembly and disassembly

Cryo-Electron Tomography:

  • Visualization of SRP complexes in their native cellular environment

  • Structural analysis of SRP-ribosome interactions at different stages of translation

  • Identification of conformational changes associated with SRP19 binding and complex assembly

Synthetic Biology Approaches:

  • Engineering of minimal SRP systems to determine essential components

  • Creation of orthogonal SRP systems with altered specificity

  • Development of optogenetic tools to control SRP function with spatial and temporal precision

These advanced approaches can reveal new aspects of SRP19 function, potentially identifying previously unrecognized roles beyond its canonical function in protein targeting, and opening new avenues for therapeutic intervention in SRP19-related disorders.

What are the most critical unresolved questions about SRP19 that require immediate research attention?

Based on the current state of knowledge, several critical questions about SRP19 require urgent research attention:

  • Mechanistic understanding of neutropenia: While genetic evidence clearly links SRP19 mutations to congenital neutropenia , the precise mechanisms by which SRP19 dysfunction leads to neutrophil-specific pathology remain unclear. Is this related to specific cargo proteins that are particularly sensitive to SRP19 levels, or to unique aspects of neutrophil development?

  • Nuclear functions of SRP19: Given the significant nuclear localization of SRP19 , does it have functions beyond SRP assembly in the nucleus? Does it interact with other nuclear RNA-protein complexes or influence gene expression?

  • Regulation of SRP19 expression: What factors control SRP19 levels, and how is its expression coordinated with other SRP components? Are there tissue-specific regulatory mechanisms that might explain the tissue-specific manifestations of SRP19 deficiency?

  • Therapeutic approaches: Can SRP19 function be enhanced or replaced in congenital neutropenia patients? Are there bypass mechanisms that can compensate for SRP19 deficiency?

These questions represent significant knowledge gaps that limit our ability to fully understand SRP19 biology and develop effective interventions for SRP19-related diseases.

What new methodologies or technologies would accelerate progress in SRP19 research?

Several emerging methodologies and technologies could significantly accelerate progress in SRP19 research:

CRISPR-Based Genetic Screens:

  • Genome-wide synthetic lethal screens in SRP19-deficient backgrounds

  • CRISPRa/CRISPRi screens to identify modifiers of SRP19 phenotypes

  • Base editing approaches for precise modeling of patient mutations

Advanced Structural Biology:

  • Cryo-EM studies of the full SRP complex with various cargoes

  • Structural analysis of disease-causing SRP19 variants

  • Time-resolved structural studies of SRP assembly

Spatial Transcriptomics and Proteomics:

  • Single-cell analysis of SRP19 expression across tissues and cell states

  • Spatial mapping of SRP19 and its interacting partners within cells

  • Subcellular proteomics to identify compartment-specific SRP19 functions

Organoid and Advanced Cell Culture Models:

  • Patient-derived organoids to study tissue-specific effects of SRP19 mutations

  • Microfluidic systems to study neutrophil development and function

  • Co-culture systems to model the bone marrow niche in SRP19 deficiency

In Vivo Imaging:

  • Development of SRP19 biosensors to monitor its activity in living organisms

  • Intravital microscopy to track neutrophil development in SRP19 mutant models

  • Whole-animal protein synthesis imaging to visualize SRP-dependent translation

These methodological advances would provide new insights into the multiple facets of SRP19 biology and accelerate the development of therapeutic strategies for SRP19-related disorders.

How might integrative multi-omics approaches enhance our understanding of SRP19 biology?

Integrative multi-omics approaches offer powerful strategies to comprehensively understand SRP19 biology across multiple dimensions:

Integrated Data Collection:

  • Parallel analysis of the genome, transcriptome, proteome, and metabolome in SRP19-deficient systems

  • Time-course studies capturing dynamic changes during cellular differentiation or stress responses

  • Single-cell multi-omics to capture cellular heterogeneity in response to SRP19 deficiency

Computational Integration:

  • Network analysis to identify key nodes connecting SRP19 to various cellular processes

  • Machine learning approaches to predict functional impacts of SRP19 variants

  • Systems biology modeling of SRP-dependent protein targeting and its downstream effects

Validation and Mechanistic Studies:

  • Targeted perturbation of key nodes identified through multi-omics integration

  • Engineering of reporter systems to monitor predicted pathway alterations

  • Therapeutic targeting based on integrated network vulnerabilities

This integrative approach would provide a comprehensive understanding of how SRP19 dysfunction propagates through cellular systems to cause disease phenotypes, potentially revealing unexpected connections and therapeutic opportunities that would not be apparent from more focused studies.

Product Science Overview

Introduction

The Signal Recognition Particle (SRP) is a ribonucleoprotein complex that plays a crucial role in targeting nascent secretory and membrane proteins to the endoplasmic reticulum (ER) in eukaryotic cells and to the plasma membrane in prokaryotic cells . The SRP complex is composed of a 300-nucleotide RNA molecule (SRP RNA) and six polypeptides: SRP9, SRP14, SRP19, SRP54, SRP68, and SRP72 .

Structure and Function

SRP19 is one of the essential protein components of the SRP complex. It is a 19 kDa protein that binds directly to the 7SL RNA, a critical component of the SRP RNA . SRP19 is responsible for mediating the binding of SRP54 to the SRP complex, which is essential for the SRP’s function in protein targeting .

Nuclear Import and Assembly

Interestingly, SRP19 is imported into the nucleus by members of the importin β superfamily of transport receptors, specifically importin 8 and transportin . This nuclear import is crucial for the assembly of the SRP complex, as SRP RNA is synthesized in the nucleus and the SRP functions in the cytoplasm . The assembly process involves SRP19 binding to the SRP RNA in the nucleus before the complex is exported to the cytoplasm .

Recombinant SRP19

Recombinant human SRP19 is produced using techniques that involve expressing the protein in a host organism, such as Escherichia coli, and purifying it for research and therapeutic purposes . This recombinant protein retains the functional properties of the native SRP19, making it valuable for studying the SRP complex’s structure and function.

Applications and Research

Research on SRP19 has provided significant insights into the mechanisms of protein targeting and the role of SRP in cellular processes. Understanding SRP19’s function and its interactions within the SRP complex can lead to advancements in biotechnology and medicine, particularly in the development of therapeutic proteins and the study of diseases related to protein targeting and transport.

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.