SIRT6 Human

Sirtuin-6 Human Recombinant
Shipped with Ice Packs
In Stock

Description

DNA Repair and Genomic Stability

  • Double-Strand Break Repair: SIRT6 stabilizes DNA-PKcs at damage sites, enhancing non-homologous end joining (NHEJ) and homologous recombination (HRR) .

  • LINE1 Suppression: Centenarian-associated SIRT6 variants (e.g., N308K/A313S) exhibit 40–50% greater efficiency in suppressing LINE1 retrotransposons, reducing genomic instability .

  • Senescence Rescue: Overexpression in pre-senescent human fibroblasts restores homologous recombination repair (HRR) capacity by 70–80% through PARP1 activation .

Metabolic Regulation

  • Hepatic Function: SIRT6 enhances gluconeogenesis and fatty acid β-oxidation in mice, maintaining NAD⁺/NADH ratios and energy homeostasis during fasting .

  • Oxidative Stress: SIRT6-knockout human mesenchymal stem cells (hMSCs) show 2.5× higher ROS levels, reversed by HO-1 overexpression .

Lifespan Extension

  • Mouse Models: SIRT6 overexpression extends median lifespan by 15–27% in C57BL/6JOlaHsd mice, with improved physical activity (30–40% longer running distances) and reduced frailty .

  • Centenarian Variants: The centSIRT6 allele (rs183444295/rs201141490) is enriched in Ashkenazi Jewish centenarians (1.0% vs. 0.5% in controls) and enhances DNA repair efficiency .

Genetic Associations with Longevity

VariantPopulationAssociationFunctional Impact
rs350845 (eQTL)Ashkenazi JewsIncreased SIRT6 expression (P = 0.009)Enhanced DNA repair, metabolic regulation
centSIRT6 (missense)Ashkenazi centenarians2× higher allele frequency vs. controlsImproved LINE1 suppression, cancer resistance

Anti-Aging Effects

  • Cellular Level: SIRT6-deficient hMSCs exhibit accelerated senescence (50% reduction in proliferation) and impaired differentiation into osteoblasts/chondrocytes .

  • Organismal Level: Transgenic mice show delayed anemia (RBC counts: 9.5 vs. 8.1 ×10⁶/μL in aged controls) and preserved albumin levels (3.8 vs. 3.2 g/dL) .

Disease Implications

  • Cancer: SIRT6 suppresses tumor growth by ribosylating LMNA (lamin A/C) and enhancing p53 activity, reducing cancer cell viability by 40–60% .

  • Inflammation: SIRT6 deacetylates H3K9 at NF-κB target genes, reducing pro-inflammatory cytokines (e.g., TNF-α) by 60–70% in murine models .

Future Directions

  • Therapeutic Targeting: Small-molecule activators (e.g., UBCS039) show promise in boosting SIRT6 activity by 3–5 fold in preclinical models .

  • Clinical Translation: Human trials are needed to validate the safety and efficacy of SIRT6 modulation in age-related diseases.

Product Specs

Introduction
Belonging to the sirtuin family of proteins (Class IV), SIRT6 is homologous to the yeast Sir2 protein and possesses a sirtuin core domain. Yeast sirtuin proteins are known for their ability to regulate epigenetic gene silencing and suppress rDNA recombination. As a chromatin-associated protein, SIRT6 plays a role in DNA repair. Human sirtuins act as intracellular regulatory proteins with mono-ADP-ribosyltransferase activity.
Description
Recombinant SIRT6 Human, produced in E. coli, is a single, non-glycosylated polypeptide chain comprising 375 amino acids (1-355 a.a.). With a molecular mass of 41 kDa, it features a 20-amino acid His-tag fused at the N-terminus. Purification is achieved through standard chromatographic techniques.
Physical Appearance
Clear, colorless, and sterile-filtered solution.
Formulation
The SIRT6 protein solution (1 mg/ml) is formulated in 20 mM Tris-HCl buffer at pH 8 with 10% glycerol.
Stability
For short-term storage (2-4 weeks), the product should be kept at 4°C. For extended storage, it is recommended to freeze at -20°C. The addition of a carrier protein (0.1% HSA or BSA) is advisable for long-term storage. Repeated freeze-thaw cycles should be avoided.
Purity
Purity exceeds 95.0% as determined by SDS-PAGE analysis.
Synonyms
Mono-ADP-ribosyltransferase sirtuin-6, SIR2-like protein 6, SIRT6, SIR2L6, Sirtuin-6.
Source
Escherichia Coli.
Amino Acid Sequence
MGSSHHHHHH SSGLVPRGSH MSVNYAAGLS PYADKGKCGL PEIFDPPEEL ERKVWELARL VWQSSSVVFH TGAGISTASG IPDFRGPHGV WTMEERGLAP KFDTTFESAR PTQTHMALVQ LERVGLLRFL VSQNVDGLHV RSGFPRDKLA ELHGNMFVEE CAKCKTQYVR DTVVGTMGLK ATGRLCTVAK ARGLRACRGE LRDTILDWED SLPDRDLALA DEASRNADLS ITLGTSLQIR PSGNLPLATK RRGGRLVIVN LQPTKHDRHA DLRIHGYVDE VMTRLMEHLG LEIPAWDGPR
VLERALPPLP RPPTPKLEPK EESPTRINGS IPAGPKQEPC AQHNGSEPAS PKRERPTSPA PHRPPKRVKA KAVPS.

Q&A

What is SIRT6 and what are its primary cellular functions?

SIRT6 is a member of the sirtuin family of NAD+-dependent deacylases that plays crucial roles in regulating various cellular processes. It functions primarily as a deacetylase, mono-ADP-ribosyltransferase, and long fatty deacylase . SIRT6 is involved in multiple pathways including DNA repair (particularly double-strand breaks), chromatin regulation, metabolism, and inflammation control . One of its key mechanisms is causing DNA wrapped around histones to tighten, leading to more heterochromatin regions of the genome . SIRT6 has been strongly correlated with longevity across species and in genetically modified mice, raising questions about whether higher SIRT6 activity may be associated with longer lifespan in humans .

Where is the SIRT6 gene located in the human genome and how is it structured?

The human SIRT6 gene is located on chromosome 19p13.3, a region identified as a mutation hotspot for many tumor diseases . Both coding and non-coding genetic polymorphisms in the SIRT6 gene region have been associated with human longevity in candidate SNP analyses . Targeted sequencing has identified specific SNPs associated with exceptional longevity, such as rs350845, which was associated with living beyond 100 years in an Ashkenazi Jewish population study (P = 0.049) . Interestingly, this SNP lies within a SIRT6 intron and functions as an eQTL (expression quantitative trait locus) for SIRT6 upregulation across 18 tissue types . This SNP is in high linkage disequilibrium (r2 > 0.98) with two other eQTLs, rs350843 and rs350846, which also upregulate SIRT6 .

How does SIRT6 contribute to DNA damage repair mechanisms?

SIRT6 plays a critical role in DNA double-strand break (DSB) repair across species. In longer-lived rodent species, SIRT6 has been optimized to repair double-strand breaks more efficiently, indicating its evolutionary importance in the study of longevity . The protein acts as a deacetylase on histones, particularly H3K9, which helps regulate chromatin structure around damaged DNA sites .

Research demonstrates that SIRT6 deficiency leads to genomic instability and increased sensitivity to DNA damage . In human mesenchymal stem cells (hMSCs), SIRT6 knockout results in accelerated functional decay . Interestingly, this decay wasn't due to compromised chromosomal stability (as might be expected) but was instead characterized by dysregulated redox metabolism and increased sensitivity to oxidative stress . This suggests SIRT6's DNA repair functions may work in concert with its role in regulating cellular metabolism and stress response pathways.

What experimental approaches best demonstrate SIRT6's role in chromatin regulation?

To investigate SIRT6's role in chromatin regulation, researchers should employ a multi-method approach:

  • Immunofluorescence microscopy: This technique effectively visualizes SIRT6 localization and its effects on chromatin structure. Experiments have shown differential staining patterns when comparing SIRT6 knockout cells with those expressing SIRT6 from different species . For example, immunofluorescence staining showing protein DAPI (blue) and SIRT6 transfection (green) can reveal how SIRT6 affects hybrid RNA presence after transfection .

  • ChIP-seq analysis: This method maps SIRT6 binding sites genome-wide and correlates them with specific histone modifications. This is particularly important since SIRT6 acts as a histone deacetylase.

  • DNA-RNA hybrid immunoprecipitation: This approach can detect how SIRT6 affects DNA-RNA hybrid formation, as knockout studies show increased prevalence of hybrids in SIRT6-deficient cells .

  • Co-immunoprecipitation studies: These reveal SIRT6's protein binding partners in chromatin regulation. Research has found SIRT6 in a protein complex with both nuclear factor erythroid 2-related factor 2 (NRF2) and RNA polymerase II, which was required for the transactivation of NRF2-regulated antioxidant genes .

  • Clustering analysis: Co-citation network analysis has identified "chromatin localization" as a key research cluster, examining SIRT6's biological functions including telomere chromatin, longevity, DNA damage, and metabolic regulation .

How can researchers effectively generate and validate SIRT6 knockout models?

Creating reliable SIRT6 knockout models requires specific methodological approaches:

  • Cell culture techniques: For in vitro models, researchers should use established cell lines such as mouse embryonic fibroblasts (MEFs). These can be cultured using Dulbecco's Modified Eagle Medium (DMEM) and standard passaging techniques, including trypsinization, centrifugation to create cell pellets, and cell counting .

  • CRISPR-Cas9 gene editing: This has proven effective for generating SIRT6 knockout human mesenchymal stem cells (hMSCs) . Validation should include confirming the absence of SIRT6 protein through Western blotting and immunofluorescence.

  • Functional validation: Beyond confirming genetic deletion, researchers should verify functional consequences. In hMSCs, SIRT6 deficiency results in accelerated functional decay and increased sensitivity to oxidative stress, providing biological confirmation of knockout effectiveness .

  • Rescue experiments: These are critical for validating the specificity of observed phenotypes. Researchers can transfect knockout cells with SIRT6 plasmids using lipofectamine reagent . For cross-species studies, using the same endogenous CMV promoter ensures consistent expression levels, as it induces very strong expression .

  • Appropriate controls: Always include wild-type cells alongside knockout models. When conducting rescue experiments, use empty vector controls for comparison .

What techniques best measure SIRT6's interaction with other proteins?

To investigate SIRT6's protein interactions, researchers should employ:

  • Co-immunoprecipitation: This technique has successfully identified SIRT6 in protein complexes with NRF2 and RNA polymerase II . These interactions were shown to be required for the transactivation of NRF2-regulated antioxidant genes, including heme oxygenase 1 (HO-1) .

  • Immunofluorescence co-localization: This method visually confirms protein interactions in cellular contexts. Double staining for SIRT6 and potential binding partners can reveal spatial relationships within cells .

  • Proximity ligation assays: These provide higher sensitivity for detecting protein-protein interactions in situ with single-molecule resolution.

  • Functional validation: Demonstrating functional relationships strengthens interaction claims. For example, overexpression of HO-1 in SIRT6-null hMSCs rescued premature cellular attrition, confirming the functional significance of the SIRT6-NRF2-HO-1 pathway .

  • Domain mapping: Using truncation or point mutations of SIRT6 helps identify specific interaction domains. This approach can distinguish between SIRT6's enzymatic and structural roles in protein complexes.

How should researchers interpret contradictory findings about SIRT6's role in disease?

When faced with contradictory findings about SIRT6 in disease contexts, researchers should:

  • Consider context-dependency: SIRT6 function may vary by tissue type, disease stage, or microenvironmental conditions. For instance, SIRT6's role may differ between cancer types or stages.

  • Examine methodology differences: Contradictions may arise from using different experimental approaches (knockout vs. knockdown) or model systems (cell lines vs. primary tissues).

  • Apply co-citation network analysis: Scientometric analysis has organized SIRT6 literature into distinct clusters, such as "SIRT6 deficiency," "DNA repair," "poor prognosis," and "myocardial infarction" . Understanding which research cluster contradictory findings belong to can help contextualize them.

  • Consider temporal dynamics: Timeline views of SIRT6 research show the evolution of different focus areas. Early studies focused on DNA repair (cluster 2), while later research explored disease implications (clusters 4 and 5) . This temporal context may explain apparent contradictions.

  • Examine species differences: Cross-species comparisons reveal that SIRT6 function varies between species. For example, beaver SIRT6 shows higher hyperphosphorylation than mouse SIRT6 and is better at repressing L1s . Similarly, porcupine SIRT6 shows higher prevalence of SIRT6 protein than hamster SIRT6 in rescue experiments .

What statistical approaches are most appropriate for analyzing SIRT6 genetic association studies?

When analyzing SIRT6 genetic associations, researchers should employ:

  • Case-control study design: The approach used to identify rs350845 association with centenarians (P = 0.049) provides a methodological template . Researchers should compare SIRT6 variants between individuals with exceptional longevity and appropriate controls.

  • Linkage disequilibrium analysis: It's crucial to examine SNPs in high linkage disequilibrium with identified variants. For example, rs350845 is in high LD (r2 > 0.98) with two other eQTLs, rs350843 and rs350846, which also upregulate SIRT6 .

  • eQTL analysis: For non-coding variants, determine if they function as expression quantitative trait loci (eQTLs). The SNP rs350845 is an eQTL for SIRT6 upregulation across 18 tissue types, explaining its potential mechanism in longevity .

  • Multiple testing correction: When examining numerous SNPs, appropriate statistical corrections should be applied to minimize false positives.

  • Meta-analysis approaches: For contradictory findings, systematic reviews and meta-analyses can help determine consensus effects across studies .

How can researchers effectively compare SIRT6 function across different species?

Cross-species SIRT6 analysis requires specific methodological approaches:

  • Rescue experiments: Transfect SIRT6 knockout cells with SIRT6 plasmids from different species. This approach has successfully demonstrated functional differences between species' SIRT6 proteins .

  • Consistent expression systems: Use the same endogenous promoter (such as CMV) across species to ensure comparable expression levels .

  • Immunofluorescence visualization: This technique effectively demonstrates differences in SIRT6 function between species. For instance, immunofluorescence has shown that beaver SIRT6 is more hyperphosphorylated than mouse SIRT6 and better at repressing L1s . Similarly, porcupine SIRT6 shows higher prevalence of SIRT6 protein compared to hamster SIRT6 .

  • Correlate with maximum lifespan: Compare SIRT6 function with species lifespan data. Rodent species have drastically different lifespans not specifically correlated to size (rats: up to 4 years; naked mole rats: up to 30 years), making them excellent candidates for such comparisons .

  • Hybrid formation analysis: Measure DNA-RNA hybrid presence after SIRT6 transfection from different species. S6KO hamster MEFs show higher presence of hybrids compared to S6KO porcupine MEFs, indicating species-specific functional differences .

What experimental strategies best demonstrate SIRT6's role in mesenchymal stem cell homeostasis?

To investigate SIRT6 in mesenchymal stem cell (MSC) homeostasis:

  • Generate SIRT6 knockout hMSCs: Using targeted gene editing such as CRISPR-Cas9 allows direct examination of SIRT6's role in these cells .

  • Measure functional decay parameters: Rather than focusing solely on senescence markers, assess multiple functional aspects of MSCs including proliferation, differentiation capacity, and response to stress .

  • Examine redox metabolism: SIRT6-deficient hMSCs exhibit dysregulated redox metabolism rather than compromised chromosomal stability. Researchers should measure oxidative stress markers and antioxidant pathway components .

  • Analyze protein complexes: Co-immunoprecipitation reveals that SIRT6 forms complexes with NRF2 and RNA polymerase II, critical for antioxidant gene transactivation .

  • Conduct rescue experiments: Overexpression of downstream targets like heme oxygenase 1 (HO-1) in SIRT6-null hMSCs can rescue premature cellular attrition, confirming pathway specificity .

  • Perform aging-related analyses: Since stem cell exhaustion is considered a hallmark of aging, researchers should connect SIRT6's role in hMSCs to broader aging processes .

How can researchers effectively study SIRT6's role in oxidative stress regulation?

To investigate SIRT6's function in oxidative stress regulation:

  • Examine NRF2 pathway interactions: Research has uncovered that SIRT6 functions as a NRF2 coactivator, representing a novel regulatory layer for oxidative stress-associated stem cell decay . Researchers should analyze how SIRT6 influences NRF2 target gene expression.

  • Measure antioxidant gene expression: SIRT6 is required for the transactivation of NRF2-regulated antioxidant genes, including heme oxygenase 1 (HO-1) . qPCR and Western blot analysis of these targets provides functional readouts of SIRT6 activity.

  • Test cellular sensitivity to oxidative stressors: SIRT6-null cells show increased sensitivity to oxidative stress. Researchers should challenge cells with oxidative agents and measure survival, damage markers, and recovery .

  • Perform rescue experiments: Overexpression of downstream targets like HO-1 in SIRT6-null cells can rescue oxidative stress phenotypes, confirming pathway specificity .

  • Use immunofluorescence techniques: These can visualize the co-localization of SIRT6 with oxidative stress response factors and reveal changes in nuclear structure following oxidative challenge .

  • Employ redox-sensitive probes: These allow real-time monitoring of reactive oxygen species in living cells with and without functional SIRT6.

What methodological approaches best demonstrate SIRT6's connection to human longevity?

To investigate SIRT6's role in human longevity:

  • Targeted sequencing of longevity populations: The approach used to sequence the SIRT6 locus in 496 Ashkenazi Jewish centenarians and 572 controls provides a methodological template . This identified SNP rs350845 associated with living beyond 100 years (P = 0.049) .

  • eQTL analysis: For non-coding variants, determine if they function as expression quantitative trait loci. The SNP rs350845 is an eQTL for SIRT6 upregulation across 18 tissue types .

  • Linkage disequilibrium mapping: Examine SNPs in high linkage disequilibrium with identified variants. rs350845 is in high LD (r2 > 0.98) with other eQTLs that upregulate SIRT6 .

  • Cross-species comparative studies: Compare SIRT6 function across species with different lifespans. Rodent models are particularly useful as they show dramatically different lifespans not correlated with body size .

  • Rescue experiments with different species' SIRT6: Transfecting knockout cells with SIRT6 from different species can demonstrate functional conservation or divergence, correlating with lifespan differences .

  • Scientometric analysis: Review the literature using approaches like co-word and co-citation network analysis to identify how SIRT6's regulation of chromatin, lifespan, DNA damage, and metabolism forms its intellectual basis .

What emerging technologies might enhance SIRT6 research in the coming years?

Several emerging technologies show promise for advancing SIRT6 research:

  • Single-cell multi-omics: These approaches will allow researchers to examine SIRT6 function with unprecedented resolution, revealing cell-specific roles and heterogeneity.

  • CRISPR base editing and prime editing: These precise genome editing technologies will enable subtle modifications to SIRT6 and its regulatory elements without double-strand breaks.

  • Spatial transcriptomics: This will help map SIRT6 expression and its effects in tissue contexts, preserving spatial relationships that may be critical to function.

  • Protein-protein interaction mapping at scale: Technologies like proximity labeling combined with mass spectrometry will reveal the complete SIRT6 interactome across different cellular conditions.

  • In situ chromatin profiling: These methods will allow direct visualization of SIRT6's effects on chromatin in intact cells and tissues.

  • Longitudinal aging cohorts with multi-omics profiling: These will connect SIRT6 variants and expression patterns with health outcomes and lifespan.

  • AI-driven literature analysis: Expanding on current scientometric approaches, these will help synthesize the growing body of SIRT6 research and identify patterns not obvious to human researchers .

How might SIRT6 research translate into therapeutic applications?

SIRT6 research offers several promising translational avenues:

  • Small molecule SIRT6 activators: These could mimic the effects of SIRT6 upregulation seen in centenarians with specific SIRT6 variants .

  • Antioxidant pathway targeting: Since SIRT6 functions as a NRF2 coactivator in regulating antioxidant genes, this pathway represents a promising therapeutic target .

  • Stem cell therapies: SIRT6 enhancement could improve the function and resilience of mesenchymal stem cells used in regenerative medicine . Research shows that SIRT6 safeguards human mesenchymal stem cells from oxidative stress-associated functional decline .

  • Anti-aging interventions: The strong correlation between SIRT6 activity and longevity across species suggests potential for human lifespan extension .

  • Cancer-specific approaches: Understanding SIRT6's complex roles in cancer through approaches like the clustering analysis of co-citation networks could lead to context-specific cancer therapies .

  • Cardiovascular disease treatments: Research clusters involving "myocardial infarction" and SIRT6's regulation of the cardiovascular system suggest specific applications in heart disease .

What key unresolved questions about SIRT6 should guide future research priorities?

Critical unresolved questions that should guide SIRT6 research include:

  • Tissue-specific functions: How does SIRT6 function vary across different tissues, and what are the implications for targeted therapies?

  • Variant effects in diverse populations: Are there population-specific SIRT6 variants with functional consequences for longevity or disease risk beyond those identified in Ashkenazi Jewish centenarians ?

  • Interaction with environmental factors: How do diet, exercise, and environmental exposures modify SIRT6 function?

  • Downstream targets prioritization: Among SIRT6's many targets, which are most critical for its effects on longevity, metabolism, and disease resistance?

  • Species-specific optimization: What structural and functional differences explain why SIRT6 from longer-lived species (like beaver and porcupine) shows enhanced function compared to shorter-lived species (like mouse and hamster) ?

  • Therapeutic window: What is the optimal level of SIRT6 activity for health benefits without adverse effects?

  • Integration with other sirtuins: How does SIRT6 function coordinate with other sirtuin family members, particularly SIRT1 and SIRT3, which have shown connections to SIRT6 in co-citation analysis ?

Product Science Overview

Structure and Function

SIRT6 is characterized by a sirtuin core domain and is involved in DNA repair, genomic stability, and telomere integrity . It catalyzes the transfer of ADP-ribose moiety from NAD to itself and histones . This enzyme is also implicated in the regulation of gene expression and metabolism .

Biological Processes

SIRT6 is involved in several biological processes, including:

  • DNA Repair: It plays a significant role in the repair of double-strand breaks via homologous recombination .
  • Telomere Maintenance: SIRT6 is essential for maintaining telomere integrity, which is crucial for cellular aging and longevity .
  • Glycolysis Regulation: It negatively regulates the glycolytic process, thereby influencing glucose homeostasis .
  • Inflammation: SIRT6 has anti-inflammatory properties, which help in reducing age-related disorders and premature aging .
Therapeutic Potential

Human sirtuins, including SIRT6, are considered potential therapeutic targets for a broad spectrum of diseases such as cancer, neurodegenerative diseases, and metabolic disorders . Despite the challenges in developing isoform-selective and potent molecules with good drug-like properties, small molecule sirtuin regulators have advanced into clinical trials .

Research and Development

The last two decades have witnessed significant advancements in the research and development of sirtuin regulators . These regulators have shown promise in maintaining genome integrity, regulating metabolism homeostasis, and promoting organismal longevity .

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.