SQSTM1 Human

Sequestosome 1 Human Recombinant
Shipped with Ice Packs
In Stock

Description

Molecular Structure and Domains

SQSTM1 is a 440-amino-acid protein with distinct functional domains:

DomainFunctionKey Interactions
UBA (Ubiquitin-Binding)Recognizes polyubiquitin chains, enabling cargo recognition for autophagy/proteasomal degradationUbiquitinated proteins
LIR (LC3-Interacting Region)Binds ATG8 family proteins (e.g., LC3), facilitating autophagosome targetingLC3-I/II, GABARAP
PB1 (Phox and Bem1p)Mediates oligomerization and phase separation for aggrephagyPB1 domain-containing proteins (e.g., NBR1)
KEAP1-InteractingSequesters KEAP1, activating Nrf2 for antioxidant responsesKEAP1, Nrf2

The protein’s structural flexibility allows it to act as a scaffold in diverse pathways, including NF-κB signaling and mTORC1 regulation .

Core Functional Roles

SQSTM1 is pivotal in maintaining proteostasis and cellular stress responses:

Selective Autophagy

  • Aggrephagy: Binds ubiquitinated protein aggregates, forming phase-separated "p62 bodies" for autophagosome delivery .

  • Mitophagy: Modulates early PINK1-dependent mitophagy but is dispensable for mitochondrial clearance .

  • Pexophagy: Targets peroxisomes for degradation under oxidative stress via PEX5 interaction .

Proteasomal Degradation

  • DNA Repair: Promotes proteasomal degradation of FLNA and RAD51, altering homologous recombination (HR) and nonhomologous end joining (NHEJ) ratios .

Product Specs

Introduction
SQSTM1, also known as p62, is an adapter protein that plays a crucial role in various cellular processes, including ubiquitin binding, NFKB1 activation, and signaling cascades regulation. It mediates the activation of NFKB1 by TNF-a, NGF, and IL-1 and is involved in titin/TTN downstream signaling in muscle cells. SQSTM1 controls signaling cascades through ubiquitination and participates in cell differentiation, apoptosis, immune response, and regulation of K(+) channels. Mutations in the UBA domain of SQSTM1 can lead to Paget's disease due to its essential role in aggregate sequestration and cell survival.
Description
This product consists of a recombinant human SQSTM1 protein produced in E. coli. It is a single, non-glycosylated polypeptide chain containing 364 amino acids (specifically, amino acids 1-356). The protein has a molecular mass of 39.7 kDa. For purification and detection purposes, an 8 amino acid His-Tag is fused to the C-terminus of the SQSTM1 protein. The protein is purified using standard chromatography techniques.
Physical Appearance
A clear and colorless solution that has been sterilized through filtration.
Formulation
The SQSTM1 Human protein solution has a concentration of 0.5mg/ml and is supplied in a buffer containing 20mM Tris-HCl (pH 8), 1mM DTT, and 10% glycerol.
Stability
For short-term storage (up to 2-4 weeks), the product can be stored at 4°C. For long-term storage, it is recommended to store the protein at -20°C. To further enhance stability during long-term storage, consider adding a carrier protein such as HSA or BSA at a concentration of 0.1%. It's important to avoid repeated freeze-thaw cycles to maintain protein integrity.
Purity
The purity of the SQSTM1 Human recombinant protein is greater than 85%, as determined by SDS-PAGE analysis.
Synonyms
A170, OSIL, p60, p62, p62B, PDB3, ZIP3, EBIAP, ORCA, OSIL, SQSTM1.
Source
Escherichia Coli.
Amino Acid Sequence
MAMSYVKDDI FRIYIKEKKE CRRDHRPPCA QEAPRNMVHP NVICDGCNGP VVGTRYKCSV CPDYDLCSVC EGKGLHRGHT KLAFPSPFGH LSEGFSHSRW LRKVKHGHFG WPGWEMGPPG NWSPRPPRAG EARPGPTAES ASGPSEDPSV NFLKNVGESV AAALSPLGIE VDIDVEHGGK RSRLTPVSPE SSSTEEKSSS QPSSCCSDPS KPGGNVEGAT QSLAEQMRKI ALESEGRPEE QMESDNCSGG DDDWTHLSSK EVDPSTGELQ SLQMPESEGP SSLDPSQEGP TGLKEAALYP HLPPEADPRL IESLSQMLSM GFSDEGGWLT RLLQTKNYDI GAALDTIQYS KHPPPLLEHH HHHH.

Q&A

What is SQSTM1 and what are its primary functions in human cells?

SQSTM1 (Sequestosome 1), also known as p62, is a multifunctional scaffold protein that participates in numerous cellular pathways crucial for maintaining cellular homeostasis. The protein functions as a key receptor for selective autophagy by shuttling ubiquitinated cargo toward autophagic degradation, making it an essential marker for monitoring autophagic flux . Beyond autophagy, SQSTM1 participates in various signal transduction pathways, including nuclear factor-kappaB signaling, apoptosis, and oxidative stress response . It also plays significant roles in metabolic reprogramming and serves as a stress response protein that helps maintain cellular health under challenging conditions .

How is SQSTM1 structured and which domains are critical for its function?

SQSTM1 contains several functional domains that facilitate its diverse cellular roles:

  • N-terminal PB1 domain: Mediates self-oligomerization and interaction with other proteins

  • ZZ-type zinc finger domain: Involved in binding to receptor-interacting protein (RIP)

  • TRAF6-binding domain: Interacts with TRAF6, a critical component of the NF-κB pathway

  • LC3-interacting region (LIR): Binds to LC3 for autophagic processes

  • KEAP1-interacting region (KIR): Regulates antioxidant responses

  • C-terminal ubiquitin-associated (UBA) domain: Binds ubiquitinated proteins, crucial for autophagy

The C-terminal UBA domain is particularly important, as most disease-causing mutations in SQSTM1 are located in this region, highlighting its critical functional significance .

What experimental methods are most effective for detecting SQSTM1 in human samples?

Several methodological approaches can be used to detect and quantify SQSTM1 in human samples:

  • Flow cytometry: Provides rapid quantitative measurement of total cellular SQSTM1 levels with improved sensitivity compared to conventional immunoblotting. This method requires fewer starting materials and offers higher throughput capabilities .

  • Immunoblotting (Western blot): Traditional approach for detecting SQSTM1 protein levels and monitoring changes in response to treatments.

  • Immunocytochemistry/Immunofluorescence: Enables visualization of SQSTM1 distribution within cells and colocalization with other proteins of interest .

  • RT-qPCR: Allows quantification of SQSTM1 mRNA expression levels.

  • Whole-exome sequencing: Employed for identifying genetic variants in SQSTM1, particularly useful in clinical research .

For optimal results, researchers should select methods based on their specific experimental questions, considering factors such as required sensitivity, cellular localization interest, and availability of research materials.

How are SQSTM1 variants linked to neurodegenerative disorders?

SQSTM1 variants have been identified in multiple neurodegenerative conditions, suggesting its crucial role in maintaining neuronal health. Studies have found:

  • SQSTM1 mutations occur in approximately 1% of amyotrophic lateral sclerosis (ALS) cases and up to 3% of frontotemporal dementia (FTD) cases .

  • Rare missense variants in SQSTM1 were identified in 4% of sporadic inclusion body myositis (sIBM) patients, with variants such as p.G194R significantly overrepresented compared to controls .

  • The SQSTM1 p.P392L mutation, located in the C-terminal ubiquitin-associated domain, is the most frequent mutation observed across different clinical phenotypes .

  • Loss-of-function mutations in SQSTM1 can lead to childhood- or adolescence-onset neurodegenerative disorders with characteristics similar to adult-onset conditions .

These genetic associations indicate that defects in SQSTM1-mediated cellular pathways, particularly those involving protein degradation and autophagy, may confer genetic susceptibility to various neurodegenerative diseases, reinforcing the mechanistic overlap in these disorders .

What methodological approaches are recommended for studying the effects of SQSTM1 variants in neurons?

When investigating SQSTM1 variants in neuronal contexts, researchers should consider these methodological approaches:

  • iPSC-derived neuronal models: Human induced pluripotent stem cells (iPSCs) can be genetically modified using CRISPR/Cas9 to generate SQSTM1 knockout or variant lines, which can then be differentiated into specific neuronal subtypes, such as cortical neurons .

  • Genomic integrity verification: After genetic modification, confirm the integrity of edited iPSC clones through karyotype G-banding analysis and array comparative genome hybridization .

  • Functional assessments:

    • Mitochondrial functionality using live-cell bioenergetics assays (e.g., Seahorse)

    • MitoTracker staining to assess mitochondrial distribution and morphology

    • Immunocytochemistry with mitochondrial markers (e.g., TOMM20)

    • Expression analysis of oxidative phosphorylation genes

  • Cellular phenotyping:

    • Autophagy flux monitoring through LC3-II/I ratios and SQSTM1 accumulation

    • Assessment of neuronal morphology, synaptic density, and electrophysiological properties

    • Stress response evaluations under conditions relevant to neurodegeneration

These approaches enable comprehensive analysis of how SQSTM1 variants affect neuronal function, providing insights into potential pathogenic mechanisms in neurodegenerative diseases.

How can researchers accurately measure autophagic flux using SQSTM1 as a marker?

SQSTM1 serves as a key marker for monitoring autophagic flux, but proper experimental design is essential for accurate interpretation:

  • Combined marker approach: Always use SQSTM1 in conjunction with other autophagy markers, particularly LC3-II, as changes in SQSTM1 levels alone can be misleading due to its transcriptional regulation .

  • Flux analysis protocol:

    • Measure baseline SQSTM1 levels

    • Add autophagy inhibitors (e.g., bafilomycin A1 or chloroquine) to block lysosomal degradation

    • Compare accumulation rates between experimental conditions

    • Include appropriate controls (e.g., starvation to induce autophagy)

  • Flow cytometry method:

    • Fix cells with 4% paraformaldehyde

    • Permeabilize with 0.1% Triton X-100

    • Incubate with anti-SQSTM1 primary antibody followed by fluorescently-labeled secondary antibody

    • Analyze using standard flow cytometry equipment, measuring geometric mean or median fluorescence intensity

  • Experimental considerations:

    • Account for transcriptional changes in SQSTM1 through parallel mRNA measurements

    • Include genetic controls (e.g., ATG5 or ATG7 knockout cells) to validate autophagy dependence

    • Consider cell type-specific variations in baseline autophagy levels

When interpreting results, remember that increased SQSTM1 can indicate either autophagy inhibition or upregulated expression, while decreased levels may reflect enhanced autophagy or reduced expression . Therefore, kinetic analyses and multiple methodological approaches provide more reliable assessments of autophagic flux.

What are the most significant confounding factors when studying SQSTM1 in autophagy experiments?

Several confounding factors can complicate the interpretation of SQSTM1-based autophagy studies:

  • Transcriptional regulation: SQSTM1 expression is regulated by various stress responses, including oxidative stress and proteasome inhibition, potentially leading to increased protein levels independent of autophagy changes .

  • Protein stability issues: SQSTM1 can be degraded through multiple pathways, including proteasomal degradation, not just autophagy.

  • Cell type variability: Different cell types exhibit varying baseline levels of SQSTM1 and autophagy flux rates, making direct comparisons between cell types challenging .

  • Subcellular localization: SQSTM1 can shuttle between different cellular compartments, affecting its detection depending on the experimental method used.

  • Methodological limitations: Technical aspects like antibody specificity, fixation protocols, and normalization methods can significantly impact results.

To mitigate these confounding factors, researchers should:

  • Include appropriate positive and negative controls

  • Use multiple complementary techniques to measure autophagy

  • Monitor SQSTM1 mRNA levels alongside protein measurements

  • Perform time-course experiments rather than single time-point analyses

  • Validate findings using genetic manipulations of core autophagy components

How does SQSTM1 deficiency affect mitochondrial function in human neurons?

SQSTM1 deficiency has significant impacts on mitochondrial function in human neurons, though with some surprising nuances:

  • Respiratory capacity: SQSTM1 knockout neurons show reduced spare respiratory capacity, indicating compromised ability to increase energy production under stress conditions. This suggests SQSTM1 plays a regulatory role in mitochondrial bioenergetics .

  • Gene expression alterations: Loss of SQSTM1 leads to dysregulation of genes involved in oxidative phosphorylation and mitochondrial function, particularly affecting components of the electron transport chain .

  • Mitochondrial distribution: Interestingly, despite functional deficits, SQSTM1-deficient neurons maintain normal mitochondrial distribution throughout cellular compartments, as demonstrated through MitoTracker staining and TOMM20 immunocytochemistry .

  • Mitophagy dynamics: SQSTM1 affects early processes of PINK1-dependent mitophagy but appears dispensable for complete mitochondrial clearance, suggesting compensatory mechanisms exist for mitochondrial quality control in neurons .

  • Developmental impact: SQSTM1 is not required for cortical neuron differentiation from iPSCs, indicating its role in mitochondrial function emerges primarily in mature neurons rather than during neuronal development .

These findings highlight the complex relationship between SQSTM1 and mitochondrial health in neurons, with implications for understanding neurodegenerative diseases associated with SQSTM1 mutations.

What experimental designs are optimal for investigating SQSTM1-mediated mitophagy in human cells?

To effectively investigate SQSTM1-mediated mitophagy in human cells, researchers should consider the following experimental approaches:

  • Genetic manipulation strategies:

    • CRISPR/Cas9-mediated SQSTM1 knockout in relevant cell types (e.g., iPSCs that can be differentiated into neurons)

    • Generation of point mutations mimicking disease-associated variants

    • Rescue experiments with wild-type or mutant SQSTM1 to confirm specificity

  • Mitophagy induction methods:

    • Mitochondrial uncouplers (e.g., CCCP, FCCP)

    • Antimycin A/oligomycin combination treatment

    • Hypoxia/reoxygenation

    • Parkin overexpression in combination with mitochondrial stressors

  • Mitophagy assessment techniques:

    • Live-cell imaging with mitochondrial reporters and pH-sensitive fluorophores

    • Colocalization analysis of mitochondrial markers with autophagosomal/lysosomal proteins

    • Biochemical fractionation to quantify mitochondrial mass

    • Mitochondrial DNA quantification

    • Flow cytometry with mitochondrial dyes

  • Time-course considerations:

    • Short-term (minutes to hours): For capturing early events in mitophagy initiation

    • Medium-term (hours to days): For monitoring complete mitochondrial clearance

    • Long-term (days to weeks): For assessing consequences on cellular health and adaptation

  • Validation controls:

    • PINK1 or Parkin knockout cells as pathway-specific controls

    • ATG5/ATG7 knockout cells as general autophagy controls

    • Lysosomal inhibitors to confirm flux

When designing these experiments, it's important to consider cell type-specific variations in mitophagy mechanisms, as SQSTM1's role may differ between cell types. The role of SQSTM1 in neuronal mitophagy appears complex, as it influences early mitophagic processes but may be dispensable for complete mitochondrial clearance, suggesting context-dependent functions .

How can researchers effectively model SQSTM1-related diseases using iPSC technology?

To effectively model SQSTM1-related diseases using iPSC technology, researchers should follow these methodological steps:

  • iPSC source selection:

    • Patient-derived iPSCs harboring natural SQSTM1 mutations

    • CRISPR/Cas9-engineered iPSCs with specific SQSTM1 variants

    • Isogenic control lines to minimize genetic background effects

  • Quality control measures:

    • Confirm genomic integrity post-editing via karyotyping and array comparative genome hybridization

    • Verify pluripotency marker expression

    • Validate mutation status through sequencing

  • Differentiation protocols:

    • Select cell types relevant to the disease (e.g., cortical neurons for neurodegenerative conditions)

    • Employ well-established, reproducible differentiation protocols

    • Characterize resulting cell populations using lineage-specific markers

  • Phenotypic assessments:

    • Mitochondrial function: Seahorse assays, mitochondrial membrane potential measurements

    • Autophagy dynamics: Flux assays, selective substrate degradation

    • Proteostasis: Protein aggregation, ubiquitination patterns

    • Stress responses: Oxidative stress vulnerability, ER stress markers

    • Cell-type specific functions: For neurons—electrophysiology, neurite outgrowth, synaptic density

  • Disease-relevant challenges:

    • Apply stressors that mimic disease conditions (e.g., oxidative stress, proteasome inhibition)

    • Conduct long-term cultures to model age-related aspects of disease

    • Consider co-culture systems to examine cell-cell interactions

This approach has successfully revealed that while SQSTM1 is dispensable for cortical neuron differentiation, its loss significantly impacts mitochondrial functionality and gene expression in mature neurons—findings that may explain aspects of SQSTM1-related neurodegeneration .

What are the most promising therapeutic targets in the SQSTM1 pathway for neurodegenerative diseases?

Based on current research, several promising therapeutic targets within the SQSTM1 pathway show potential for neurodegenerative disease interventions:

  • Autophagy enhancement strategies:

    • mTOR inhibitors to upregulate general autophagy

    • TFEB activators to increase lysosomal biogenesis and function

    • Small molecules that enhance selective autophagy without affecting general autophagy

  • Mitochondrial function modulators:

    • Compounds targeting bioenergetic deficits observed in SQSTM1-deficient neurons

    • Mitochondrial antioxidants to reduce oxidative stress burden

    • Enhancers of mitochondrial biogenesis to compensate for clearance defects

  • SQSTM1 structural and functional targets:

    • Stabilizers of SQSTM1 protein preventing degradation of functional variants

    • Small molecules enhancing SQSTM1's binding to ubiquitinated substrates

    • Compounds targeting specific domains affected by disease mutations, particularly the UBA domain

  • Inflammatory pathway modulation:

    • NF-κB pathway modulators, given SQSTM1's role in this signaling cascade

    • Regulators of major histocompatibility complex (MHC) genes, which show upregulation in SQSTM1-related disorders

  • Gene therapy approaches:

    • AAV-mediated delivery of functional SQSTM1 to affected tissues

    • CRISPR-based correction of disease-causing mutations

    • Antisense oligonucleotides to modulate SQSTM1 expression or splicing

The therapeutic potential of these targets is supported by findings that SQSTM1 variants contribute to 1-3.5% of ALS/FTD cases and are overrepresented in sIBM patients . Additionally, research demonstrating that SQSTM1 affects mitochondrial function but is dispensable for complete mitophagy suggests that targeting downstream pathways might be particularly effective .

How can researchers resolve contradictory findings in SQSTM1 research literature?

The SQSTM1 research field contains several apparently contradictory findings that require careful methodological approaches to resolve:

  • Systematic comparison of experimental models:

    • Directly compare different model systems (cell lines, primary cells, iPSC-derived neurons, animal models) using standardized protocols

    • Account for species-specific differences in SQSTM1 function and regulation

    • Consider cell type-specific roles of SQSTM1, as its function may vary significantly between neurons, muscle cells, and other cell types

  • Methodological standardization:

    • Develop consensus protocols for key assays (autophagy flux, mitophagy, etc.)

    • Use multiple complementary techniques to assess the same biological process

    • Report detailed experimental conditions that may affect outcomes (cell density, passage number, etc.)

  • Genetic background considerations:

    • Use isogenic controls whenever possible to minimize confounding genetic variables

    • When studying patient-derived samples, collect detailed genetic information beyond the SQSTM1 locus

    • Consider interaction effects between SQSTM1 variants and other genetic modifiers

  • Temporal dynamics analysis:

    • Conduct time-course experiments rather than single time-point analyses

    • Consider acute versus chronic effects of SQSTM1 manipulation

    • Account for compensatory mechanisms that may emerge over time

  • Context-dependent function evaluation:

    • Assess SQSTM1 function under different cellular stress conditions

    • Consider tissue-specific expression patterns and post-translational modifications

    • Evaluate the impact of aging and disease-relevant stressors on SQSTM1 function

These approaches can help reconcile seemingly contradictory findings, such as observations that SQSTM1 affects early mitophagy processes but is dispensable for complete mitochondrial clearance , or that SQSTM1 mutations can lead to diverse clinical presentations ranging from bone disorders to various neurodegenerative conditions .

Product Science Overview

Structure and Function

Sequestosome 1 is a ubiquitin-binding protein that acts as a scaffold/adaptor protein. It is involved in the selective degradation of ubiquitinated proteins through autophagy, a process known as aggrephagy . The protein contains multiple domains that facilitate its interaction with various proteins and organelles, enabling it to bridge polyubiquitinated proteins and autophagosomes .

Role in Cellular Processes
  1. Autophagy: Sequestosome 1 is essential for the recruitment of ubiquitinated cargo proteins to autophagosomes. It interacts with microtubule-associated protein 1 light chain 3 (MAP1LC3), which is crucial for the formation of autophagosomes .
  2. Signal Transduction: SQSTM1 regulates the activation of the NF-kB signaling pathway by acting as a scaffold for the assembly of signaling complexes. It interacts with tumor necrosis factor receptor-associated factor 6 (TRAF6) to mediate NF-kB activation in response to upstream signals .
  3. Oxidative Stress Response: The protein is involved in the cellular response to oxidative stress by regulating the degradation of damaged proteins and organelles .
Clinical Significance

Mutations in the SQSTM1 gene are associated with several diseases, including:

  • Paget’s Disease of Bone: A disorder characterized by abnormal bone remodeling, leading to bone pain, deformities, and fractures .
  • Frontotemporal Dementia and/or Amyotrophic Lateral Sclerosis (FTD/ALS): Neurodegenerative disorders that affect cognitive and motor functions .
Recombinant Sequestosome 1

Recombinant Sequestosome 1 (Human) is produced using recombinant DNA technology, which involves the insertion of the human SQSTM1 gene into an expression system, such as bacteria or yeast, to produce the protein in large quantities. This recombinant protein is used in various research applications to study its structure, function, and role in disease processes.

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.