SIRT3 Human

Sirtuin 3 Human Recombinant
Shipped with Ice Packs
In Stock

Description

Molecular Structure

  • Domain Architecture: SIRT3 contains a catalytic sirtuin core domain and a mitochondrial targeting sequence at its N-terminus .

  • Crystal Structures: Structural studies reveal conformational changes during substrate binding. For example, SIRT3 interacts with acetyl-CoA synthetase 2 (AceCS2) via a substrate-induced "open-to-closed" transition .

Key Biological Roles

FunctionMechanismSubstrates/Pathways
Energy MetabolismDeacetylates enzymes in the TCA cycle and electron transport chain (ETC)AceCS2, IDH2, Complexes I–IV
ROS RegulationActivates antioxidant enzymes (e.g., MnSOD) to neutralize superoxide radicalsMnSOD, FOXO3a
Mitochondrial DynamicsModulates fission/fusion via deacetylation of OPA1 and MFN2OPA1, MFN2
ApoptosisInhibits mitochondrial permeability transition pore (mPTP) openingCyclophilin D

Disease Associations and Mechanisms

Liver Diseases

  • Non-Alcoholic Fatty Liver Disease (NAFLD): Reduced SIRT3 levels correlate with hepatic lipid accumulation. Overexpression restores BNIP3-mediated mitophagy and inhibits fibrosis .

  • Viral Hepatitis: SIRT3 deacetylates HBV cccDNA-bound histone H3, suppressing viral replication .

Neurodegenerative Disorders

  • Alzheimer’s and Parkinson’s: SIRT3 deficiency exacerbates oxidative stress and mitochondrial dysfunction. Caloric restriction upregulates SIRT3, enhancing neuronal survival .

  • Amyotrophic Lateral Sclerosis: SIRT3 rescues SOD1-induced mitochondrial fragmentation .

Cancer

  • Tumor Suppression: SIRT3 inhibits glycolysis (Warburg effect) by deacetylating pyruvate dehydrogenase complex (PDC), promoting apoptosis .

  • Oncogenic Role: Context-dependent overexpression in certain cancers may enhance stress adaptation .

Metabolic and Cardiovascular Diseases

  • Insulin Resistance: SIRT3 knockout mice exhibit hyperacetylation of mitochondrial proteins and impaired glucose metabolism .

  • Cardiac Hypertrophy: SIRT3 deficiency increases ROS, leading to pathological remodeling .

Key Research Findings

Substrate-Specific Mechanisms

  • AceCS2 Activation: Deacetylation of AceCS2 at Lys-642 enhances acetyl-CoA synthesis, linking SIRT3 to fatty acid oxidation .

  • IDH2 and MnSOD: Deacetylation boosts NADPH and glutathione production, mitigating oxidative damage .

Therapeutic Targets

CompoundEffect on SIRT3Therapeutic PotentialReferences
ResveratrolActivatorEnhances mitochondrial biogenesis in neurodegeneration
HonokiolActivatorReduces cardiac hypertrophy and fibrosis
AGK2InhibitorSuppresses SIRT3-dependent cancer cell survival

Product Specs

Introduction
SIRT3, a member of the Sirtuin protein family, plays a crucial role in aging, stress resistance, and metabolic regulation. Characterized by a Sirtuin core domain, SIRT3 exhibits NAD+-dependent deacetylase activity within mitochondria. Its overexpression elevates mitochondrial uncoupling protein 1 levels. Notably, SIRT3 protein levels are significantly elevated in specific breast cancer types.
Description
Produced in E. coli, our SIRT3 is a single, non-glycosylated polypeptide chain encompassing amino acids 118-399. With a molecular mass of 33.5 kDa, this protein is fused to a 21 amino acid His-tag at the N-terminus and purified using proprietary chromatographic techniques.
Physical Appearance
Clear, sterile-filtered solution.
Formulation
The SIRT3 protein solution (1mg/ml) is supplied in 20mM Tris-HCl buffer (pH 8.0) containing 1mM DTT, 0.1M NaCl, and 10% glycerol.
Purity
Purity exceeds 95% as determined by SDS-PAGE.
Stability
For short-term storage (2-4 weeks), keep at 4°C. For extended periods, store frozen at -20°C. Adding a carrier protein (0.1% HSA or BSA) is recommended for long-term storage. Minimize freeze-thaw cycles.
Synonyms
Sirtuin 3, SIR2-like protein 3, Sir2-like 3, NAD-dependent deacetylase sirtuin-3, mitochondrial.
Source
Escherichia Coli.
Amino Acid Sequence
MGSSHHHHHH SSGLVPRGSH MSDKGKLSLQ DVAELIRARA CQRVVVMVGA GISTPSGIPD FRSPGSGLYS NLQQYDLPYP EAIFELPFFF
HNPKPFFTLA KELYPGNYKP NVTHYFLRLL HDKGLLLRLY TQNIDGLERV SGIPASKLVE AHGTFASATC TVCQRPFPGE DIRADVMADR
VPRCPVCTGV VKPDIVFFGE PLPQRFLLHV VDFPMADLLL ILGTSLEVEP FASLTEAVRS SVPRLLINRD LVGPLAWHPR SRDVAQLGDV
VHGVESLVEL LGWTEEMRDL VQRETGKLDG PDK.

Q&A

What is the subcellular localization of SIRT3 in human cells?

SIRT3 is predominantly localized in the mitochondria, where it functions as an NAD⁺-dependent deacetylase. While initially synthesized as a longer protein in the cytoplasm, SIRT3 undergoes proteolytic processing during import into the mitochondria, resulting in the removal of its N-terminal localization sequence. Research methodologies to study SIRT3 localization typically involve subcellular fractionation followed by Western blotting, immunofluorescence microscopy with mitochondrial co-staining, or expression of SIRT3-GFP fusion proteins. Recent super-resolution microscopy techniques have further refined our understanding of SIRT3's precise submitochondrial distribution, showing enrichment in the mitochondrial matrix where it can access numerous protein substrates involved in metabolic regulation .

How does the structure of SIRT3 relate to its deacetylase function?

SIRT3 consists of two distinct domains: a larger domain containing the catalytic core and a smaller zinc-binding domain. The catalytic mechanism involves a cleft formed between these domains where substrates bind. Structurally, SIRT3 contains a flexible loop that changes conformation during catalysis. Crystal structure analyses reveal that substrate binding promotes a productive conformation that facilitates NAD⁺ binding, which is essential for deacetylase activity . Mutations at key conserved residues (such as H248) abolish catalytic activity. Research approaches typically employ site-directed mutagenesis of specific residues followed by enzymatic activity assays to correlate structure with function. X-ray crystallography and molecular dynamics simulations have been instrumental in elucidating how substrate binding induces conformational changes necessary for catalysis .

What are the major protein targets of SIRT3 deacetylation in human cells?

SIRT3 deacetylates numerous mitochondrial proteins involved in metabolism, antioxidant defense, and mitochondrial dynamics. Key substrates include:

Protein TargetFunctionConsequence of SIRT3 Deacetylation
SOD2 (MnSOD)Antioxidant enzymeActivation, increased ROS detoxification
LCADFatty acid oxidationEnhanced metabolic efficiency
IDH2TCA cycleIncreased NADPH production, redox balance
Cyclophilin DmPTP componentInhibition of mitochondrial permeability transition
FOXO3aTranscription factorEnhanced antioxidant gene expression
ParkinMitophagy regulatorPromotion of mitochondrial quality control

Research methodologies to identify SIRT3 targets typically include mass spectrometry-based acetylomics comparing wild-type and SIRT3-deficient cells, followed by validation through in vitro deacetylation assays with recombinant SIRT3 and site-specific acetylation antibodies .

How does SIRT3 deficiency contribute to cardiac fibrosis and inflammation?

SIRT3 deficiency promotes cardiac fibrosis and inflammation through multiple interconnected mechanisms. Studies using SIRT3 knockout mice have demonstrated augmented transcriptional activity of AP-1, a key regulator of inflammatory and fibrotic responses. Mechanistically, SIRT3 inhibits FOS transcription (a component of AP-1) through specific histone H3 lysine K27 deacetylation at its promoter . The absence of this epigenetic regulation results in sustained proinflammatory signaling. Additionally, SIRT3 deficiency leads to mitochondrial dysfunction, increased ROS production, and activation of redox-sensitive proinflammatory pathways including NF-κB. Research methodologies include histological assessment of fibrosis (Masson's trichrome staining), qPCR analysis of inflammatory cytokines, chromatin immunoprecipitation (ChIP) assays to assess histone modifications, and electrophoretic mobility shift assays (EMSA) to measure transcription factor binding activity .

What experimental approaches can evaluate SIRT3's role in pressure overload-induced cardiac hypertrophy?

To evaluate SIRT3's role in pressure overload-induced cardiac hypertrophy, researchers employ:

  • Animal Models: Transverse aortic constriction (TAC) in SIRT3 knockout and overexpressing mice compared to wild-type controls.

  • Cellular Models: Cardiomyocytes treated with phenylephrine or angiotensin II to induce hypertrophy, with SIRT3 levels manipulated through overexpression or siRNA knockdown.

  • Functional Assessment:

    • Echocardiography for in vivo cardiac function and dimensions

    • Pressure-volume loops for hemodynamic parameters

    • Heart weight to body weight or tibia length ratios

  • Molecular Analysis:

    • Cardiomyocyte cross-sectional area measurement

    • qPCR for hypertrophic markers (ANP, BNP, β-MHC)

    • Western blotting for signaling pathways (mTOR-p70S6K, JNK, TGF-β/Smad3)

    • Mitochondrial function assays (oxygen consumption, ATP production)

  • Pharmacological Intervention: Testing SIRT3 activators (like 2-APQC) to assess their ability to prevent or reverse hypertrophy .

This multifaceted approach enables comprehensive assessment of how SIRT3 modulates the hypertrophic response at molecular, cellular, and whole-organ levels .

How do SIRT3 levels correlate with vascular function in hypertensive patients?

SIRT3 levels show significant negative correlation with vascular dysfunction in hypertensive patients. Clinical research demonstrates that arterioles from human subjects with essential hypertension have decreased SIRT3 levels compared to normotensive controls . This reduction correlates with alterations in vascular metabolic, inflammatory, and cell-senescence pathways, mirroring findings in SIRT3-depleted mice. Methodologically, these studies involve:

  • Collection of resistance arterioles from subcutaneous fat biopsies of hypertensive and normotensive patients

  • Quantification of SIRT3 protein and mRNA levels

  • Assessment of endothelial function through wire myography measuring endothelium-dependent and independent vasodilation

  • Analysis of oxidative stress markers (DHE staining, protein carbonylation)

  • Evaluation of inflammatory markers (IL-6, MCP-1)

  • Transcriptomic profiling to identify altered pathways

The consistent correlation between reduced SIRT3 expression and vascular dysfunction across both human samples and animal models suggests SIRT3 as a potential therapeutic target for vascular complications in hypertension .

How does SIRT3 expression vary across different regions and cell types in the human brain?

SIRT3 expression varies considerably across brain regions and neural cell types. Studies indicate that SIRT3 is the third highest expressed sirtuin in the adult brain (behind SIRT2 and SIRT5). Expression analysis shows fairly consistent SIRT3 levels from embryonic day 18 through advanced age (24 months) in rat cortex, cerebellum, and hippocampus, with developmental fluctuations observed between postnatal day 7 and 21 . This temporal pattern suggests critical periods during neural development when SIRT3 function may be particularly important.

In terms of cell-type distribution, SIRT3 is not restricted to neurons; significant expression has been documented in astrocytes and microglia, indicating diverse roles across neural cell populations. Research methodologies to map SIRT3 expression include:

  • Cell-type specific isolation followed by qPCR and Western blotting

  • Immunohistochemistry with co-staining for neuronal, astrocytic, and microglial markers

  • Single-cell RNA sequencing to precisely quantify SIRT3 expression across neural cell subtypes

  • In situ hybridization for region-specific expression patterns

This heterogeneous expression pattern suggests that SIRT3 may have cell type-specific functions in the brain, potentially explaining its diverse roles in neuroprotection, metabolism, and stress response .

What methodologies can assess SIRT3's role in protecting against neurodegenerative disease models?

To assess SIRT3's neuroprotective role in neurodegenerative disease models, researchers employ multi-level investigative approaches:

  • In vitro models:

    • Primary neuron cultures from SIRT3 knockout or overexpressing mice

    • Application of disease-specific stressors (Aβ for Alzheimer's, MPP+ for Parkinson's)

    • Assessment of neuronal viability, mitochondrial function, and oxidative stress

    • Mechanistic pathway analysis through pharmacological inhibitors or genetic manipulation

  • Animal models:

    • Crossing SIRT3 knockout or transgenic mice with neurodegenerative disease models

    • Behavioral testing (cognitive function, motor coordination)

    • Neuroimaging (MRI for brain volume, PET for metabolism)

    • Histological analysis of pathological hallmarks (protein aggregation, neuroinflammation)

  • Biochemical analyses:

    • Mitochondrial function (respirometry, membrane potential)

    • Oxidative stress markers (protein carbonylation, lipid peroxidation)

    • ATP production and NAD+/NADH ratio measurements

    • Acetylome analysis to identify disease-relevant SIRT3 substrates

  • Therapeutic intervention testing:

    • Administration of SIRT3 activators at various disease stages

    • Dose-response studies and brain penetration analysis

    • Comparison with established treatments

    • Combined therapy approaches

These methodologies help establish whether SIRT3 activation represents a viable therapeutic strategy for neurodegenerative conditions .

How does SIRT3 regulation of mitochondrial metabolism impact human metabolic diseases?

SIRT3 regulation of mitochondrial metabolism has profound implications for human metabolic diseases through its deacetylation of key enzymes involved in energy production and substrate utilization. Research methodologies examining this relationship typically include:

  • Metabolic flux analysis: Using isotope-labeled substrates (13C-glucose, 13C-palmitate) to trace metabolic pathways in SIRT3-manipulated systems, revealing how SIRT3 influences substrate preference and utilization efficiency.

  • Respirometry: Measuring oxygen consumption rates and extracellular acidification in tissues and cells with altered SIRT3 expression, demonstrating SIRT3's impact on oxidative phosphorylation and glycolysis.

  • Metabolomics: Comprehensive profiling of metabolites in SIRT3-deficient versus normal samples to identify altered metabolic signatures.

  • Patient-derived samples: Analysis of SIRT3 expression, activity, and target acetylation in samples from individuals with metabolic disorders compared to healthy controls.

These approaches have revealed that SIRT3 deficiency contributes to metabolic dysfunction through multiple mechanisms:

Metabolic PathwaySIRT3 EffectConsequence of Dysregulation
Fatty acid oxidationActivates LCAD, MCADImpaired fat utilization, lipid accumulation
TCA cycleActivates IDH2, SDHReduced energy production, altered redox state
KetogenesisActivates HMGCS2Decreased ketone body production during fasting
Urea cycleRegulates OTCAmmonia detoxification impairment
Amino acid metabolismActivates GDHAltered glutamine utilization

These findings establish SIRT3 as a central regulator of mitochondrial metabolism whose dysfunction contributes to the pathogenesis of metabolic disorders including obesity, insulin resistance, and non-alcoholic fatty liver disease .

What experimental designs best demonstrate SIRT3's role in oxidative stress resistance?

Robust experimental designs to demonstrate SIRT3's role in oxidative stress resistance incorporate multiple complementary approaches:

  • Loss-of-function and gain-of-function models:

    • SIRT3 knockout cells/mice exposed to oxidative challenges (H₂O₂, paraquat)

    • SIRT3 overexpression systems to assess protective capabilities

    • Inducible or tissue-specific SIRT3 manipulation to avoid developmental compensation

  • Oxidative stress assessment panel:

    • Direct ROS measurement (DCF-DA, MitoSOX, EPR spectroscopy)

    • Oxidative damage markers (protein carbonylation, 8-OHdG, lipid peroxidation)

    • Antioxidant enzyme activity assays (SOD2, catalase, GPx)

    • Cellular GSH/GSSG ratio determination

  • Mechanistic dissection:

    • Site-specific mutagenesis of key SOD2 acetylation sites (K68, K122)

    • ChIP analysis of FOXO3a binding to antioxidant gene promoters

    • NAD⁺ manipulation to alter SIRT3 activity

    • Mitochondrial isolation to distinguish compartment-specific effects

  • Translational models:

    • Ischemia-reperfusion injury models

    • Aging studies comparing young vs. old tissues with SIRT3 manipulation

    • Disease models where oxidative stress is pathogenic (neurodegenerative, cardiovascular)

  • Rescue experiments:

    • Administration of mitochondria-targeted antioxidants (mitoTEMPO)

    • Reconstitution with acetylation-deficient SOD2 mutants

    • NAD⁺ precursor supplementation

These comprehensive approaches have established that SIRT3 enhances oxidative stress resistance primarily through deacetylation and activation of SOD2, the major mitochondrial antioxidant enzyme, and by indirectly supporting NADPH production via IDH2 activation .

What are the most promising approaches for pharmacological activation of SIRT3?

Pharmacological activation of SIRT3 represents a rapidly evolving field with several promising approaches:

  • Direct SIRT3 Activators:

    • Small molecule activators like 2-APQC that bind directly to SIRT3 and enhance its catalytic activity

    • Structure-based drug design targeting the SIRT3 catalytic domain

    • Allosteric modulators that stabilize active conformations

  • NAD⁺ Boosters:

    • NAD⁺ precursors (NMN, NR) that increase mitochondrial NAD⁺ pools

    • CD38 or PARP inhibitors that prevent NAD⁺ consumption

    • NAMPT activators that enhance NAD⁺ biosynthesis

  • Upstream Regulators:

    • AMPK activators that induce SIRT3 expression

    • PGC-1α activators that promote SIRT3 transcription

    • Compounds that stabilize SIRT3 protein or prevent its degradation

  • Natural Compounds:

    • Polyphenols with SIRT3-activating properties

    • Plant extracts identified through bioactivity-guided fractionation

    • Dietary compounds that increase SIRT3 expression or activity

Pharmacological characterization typically involves:

  • In vitro deacetylation assays with recombinant SIRT3

  • Cellular acetylation status of known SIRT3 targets

  • Mitochondrial function assessments

  • Target engagement studies

  • Pharmacokinetic/pharmacodynamic analysis

  • Tissue-specific distribution and blood-brain barrier penetration

The small molecule 2-APQC has emerged as particularly promising, demonstrating efficacy in alleviating cardiac hypertrophy and myocardial fibrosis in animal models through SIRT3-dependent mechanisms .

How do SIRT3 activators and inhibitors show different therapeutic potential in cardiovascular versus cancer contexts?

SIRT3 activators and inhibitors demonstrate context-dependent therapeutic effects that highlight the complex role of SIRT3 in disease pathophysiology:

Disease ContextSIRT3 ActivatorsSIRT3 Inhibitors
Cardiovascular Disease- Prevent cardiac hypertrophy and fibrosis
- Reduce vascular oxidative stress
- Improve endothelial function
- Attenuate hypertension
- Protect against ischemia-reperfusion injury
- Generally detrimental
- May exacerbate oxidative damage
- Promote endothelial dysfunction
- Accelerate cardiac aging
Cancer- Suppress certain cancer types (breast, ovarian)
- Reduce metabolic adaptation in some tumors
- Enhance stress sensitivity
- May improve chemotherapy efficacy
- Induce apoptosis in certain cancer cells
- Disrupt cancer metabolism
- Compounds MI-44 and MI-217 show strong anti-cancer effects
- May prevent adaptive responses to therapies

This dichotomy arises from SIRT3's dual nature:

  • In normal tissues, particularly cardiovascular system: SIRT3 maintains mitochondrial homeostasis, reduces oxidative stress, and prevents pathologic remodeling. Activators like 2-APQC demonstrate cardioprotective effects by enhancing these beneficial functions, inhibiting pathways like mTOR-p70S6K, JNK, and TGF-β/Smad3 to prevent hypertrophy and fibrosis .

  • In cancer contexts: SIRT3 can promote survival and metabolic adaptation in certain tumors. Inhibitors like MI-44 and MI-217 demonstrate significant anti-cancer effects, inducing apoptosis in breast cancer cells (MDA-MB-231) with apoptotic cell percentages increasing from 1.94% in control to 79.37% for MI-44 and 85.37% for MI-217 at 15 μM .

Research methodologies to evaluate this context-dependence include:

  • Parallel testing of compounds in normal versus cancer cells

  • Tissue-specific genetic manipulation in animal models

  • Combined therapy approaches (e.g., SIRT3 modulators with standard treatments)

  • Biomarker development to identify contexts where activation versus inhibition is beneficial

This context-dependence highlights the importance of developing tissue-specific or targeted delivery strategies for SIRT3 modulators to achieve desired therapeutic effects while minimizing adverse outcomes .

What role might SIRT3 play in acute lung injury and sepsis, and how could it be therapeutically targeted?

SIRT3 plays a critical protective role in acute lung injury (ALI) and sepsis through maintenance of mitochondrial homeostasis and modulation of inflammatory responses. Research using SIRT3-deficient models has revealed multiple mechanisms:

  • Mitophagy Regulation: SIRT3 deficiency impairs mitophagy through downregulation of Parkin, a key mitophagy regulator. This leads to accumulation of damaged mitochondria in lung endothelial cells during sepsis .

  • Inflammasome Activation: Impaired mitophagy in SIRT3-deficient conditions results in:

    • Increased mitochondrial ROS (mtROS) production

    • Enhanced release of mitochondrial DNA (mtDNA) into the cytosol

    • Activation of the NLRP3 inflammasome

    • Promotion of endothelial cell pyroptosis, a highly inflammatory form of cell death

  • Endothelial Barrier Function: SIRT3 maintains endothelial integrity through:

    • Preservation of adherens junctions

    • Prevention of excessive vascular permeability

    • Maintenance of endothelial metabolic homeostasis

Therapeutic targeting approaches include:

ApproachMechanismExperimental Evidence
SIRT3 restorationGene therapy or viral vectors to restore SIRT3 expressionAttenuated sepsis-induced ALI and sepsis severity in animal models
NAD⁺ boostersIncrease SIRT3 activity through NAD⁺ supplementationImproved outcomes in preclinical sepsis models
Mitochondria-targeted antioxidantsMitigate consequences of SIRT3 deficiencyMitoTEMPO reduced lung injury in septic models
NLRP3 inhibitorsBlock downstream effects of SIRT3 deficiencyPharmacological NLRP3 inhibition attenuated sepsis-induced ALI
Mitophagy inducersBypass SIRT3 deficiency to restore mitochondrial quality controlEnhanced clearance of damaged mitochondria reduced inflammation

Research methodologies to evaluate SIRT3-targeted therapies in ALI/sepsis include:

  • Cecal ligation and puncture models in SIRT3 KO and WT mice

  • LPS-induced endotoxemia with SIRT3 modulation

  • Ex vivo lung perfusion systems

  • Lung endothelial barrier function assays

  • Assessment of inflammatory mediators and cell death markers

These findings establish SIRT3 as a promising therapeutic target for sepsis-induced ALI, with restoration of SIRT3 function potentially limiting both mitochondrial dysfunction and excessive inflammatory responses .

What high-throughput screening approaches are most effective for identifying novel SIRT3 modulators?

High-throughput screening (HTS) for SIRT3 modulators requires specialized approaches due to SIRT3's unique properties and mitochondrial localization. The most effective screening strategies include:

  • Biochemical Activity-Based Screens:

    • Fluorescence-based deacetylation assays using Fluor-de-Lys technology

    • FRET-based assays monitoring proximity of SIRT3 and substrate

    • Mass spectrometry-based assays to directly measure deacetylated products

    • Bioluminescence assays coupling NAD⁺ consumption to luciferin production

  • Virtual Screening Approaches:

    • Structure-based molecular docking targeting the SIRT3 catalytic domain

    • Pharmacophore-based screening using known SIRT3 modulators as templates

    • Molecular dynamics simulations to assess ligand interactions with dynamic protein states

    • Machine learning algorithms trained on known SIRT3 modulators

  • Cell-Based Phenotypic Screens:

    • Reporter systems with SIRT3 target acetylation sites linked to fluorescent proteins

    • Mitochondrial function readouts (membrane potential, respiration)

    • Acetylation status of endogenous SIRT3 targets

    • Stress resistance assays in SIRT3-dependent models

  • Target Engagement Validation:

    • Cellular thermal shift assays (CETSA) to confirm direct binding

    • Microscale thermophoresis for binding affinity determination

    • Hydrogen-deuterium exchange mass spectrometry to map binding sites

    • Competition assays with known SIRT3 ligands

The effectiveness of virtual screening approaches has been demonstrated by the identification of eight active compounds with favorable binding affinities to SIRT3 from a library of approximately 800 compounds. Subsequent validation through docking studies confirmed stable interactions with SIRT3's catalytic domain . Two compounds, MI-44 and MI-217, showed particularly high binding free energies (-45.61 ± 0.064 kcal/mol and -41.65 ± 0.089 kcal/mol, respectively), indicating strong affinity for SIRT3 . These computational predictions were validated through functional assays, demonstrating the efficacy of integrated virtual and experimental screening approaches for SIRT3 modulator discovery.

How can researcher's best measure SIRT3 activity in human clinical samples?

  • Direct Activity Measurement:

    • Isolation of mitochondria from clinical samples (tissue biopsies, blood cells)

    • In vitro deacetylation assays using synthetic SIRT3-specific peptide substrates

    • Measurement of NAD⁺-dependent activity with SIRT3-selective inhibitor controls

    • Normalization to mitochondrial content markers (citrate synthase, COX IV)

  • Surrogate Marker Assessment:

    • Western blotting for acetylation status of well-validated SIRT3 targets:

      • Ac-SOD2 (K68)

      • Ac-LCAD (K42)

      • Ac-IDH2 (K413)

    • Quantitative immunohistochemistry for acetylated targets in tissue sections

    • Activity assays of SIRT3-regulated enzymes (SOD2, LCAD)

    • Correlation of target acetylation with clinical parameters

  • Comprehensive Profiling:

    • Acetylome analysis of mitochondrial proteins using mass spectrometry

    • Integration of acetylation data with proteomic and metabolomic profiles

    • SIRT3 protein and mRNA quantification alongside activity measures

    • NAD⁺/NADH ratio determination as a cofactor availability indicator

  • Technical Considerations:

    • Rapid sample processing to prevent ex vivo changes in acetylation

    • Preservation of native NAD⁺ levels during mitochondrial isolation

    • Inclusion of deacetylase inhibitors for baseline preservation

    • Parallel processing of control samples for standardization

  • Validation Approaches:

    • Correlation between multiple SIRT3 targets

    • Genetic validation using SIRT3 variants in population studies

    • Intervention studies with NAD⁺ precursors to dynamically assess SIRT3 responsiveness

    • Longitudinal sampling to account for temporal variations

This multi-faceted approach provides a more comprehensive assessment of SIRT3 activity than any single measure, enabling more reliable correlation with clinical outcomes and therapeutic responses .

What cutting-edge techniques are advancing our understanding of SIRT3's role in epigenetic regulation?

Cutting-edge techniques are revolutionizing our understanding of SIRT3's epigenetic regulatory functions, particularly its unexpected roles beyond mitochondria:

  • Chromatin Immunoprecipitation Sequencing (ChIP-seq) Innovations:

    • CUT&RUN (Cleavage Under Targets and Release Using Nuclease) for higher resolution mapping of SIRT3 associations with chromatin

    • CUT&Tag (Cleavage Under Targets and Tagmentation) with SIRT3 antibodies to identify genomic binding sites

    • HiChIP approaches to connect SIRT3-associated chromatin with three-dimensional genome organization

    • Integration with ATAC-seq to correlate SIRT3 binding with chromatin accessibility

  • Histone Modification Analysis:

    • Mass spectrometry-based acetylomics specifically targeting histone modifications

    • Multiplexed imaging of histone acetylation states in single cells

    • Site-specific acetyl-histone antibodies for SIRT3-regulated marks

    • ChIP-seq for H3K27ac and other marks regulated by SIRT3

  • Nuclear-Mitochondrial Crosstalk Assessment:

    • Proximity labeling techniques (BioID, APEX) to identify SIRT3-interacting proteins in different cellular compartments

    • Live-cell imaging of SIRT3 translocation between mitochondria and nucleus

    • Single-cell multi-omics to correlate SIRT3 activity with gene expression patterns

    • Mitochondrial-derived peptide influence on nuclear gene expression

  • Mechanistic Studies:

    • CRISPR-based epigenome editing to manipulate SIRT3-regulated histone modifications

    • In vitro reconstitution of SIRT3-mediated histone deacetylation

    • Nuclear vs. mitochondrial SIRT3 isoform-specific function analysis

    • Delineation of direct vs. indirect effects through metabolite-mediated regulation

Research has demonstrated that SIRT3 inhibits FOS transcription through histone H3 lysine K27 deacetylation at its promoter, revealing a novel mechanism by which SIRT3 influences inflammatory and fibrotic responses in cardiac cells . This nuclear function of SIRT3 in modulating specific histone modifications represents a paradigm shift in understanding SIRT3's regulatory roles beyond its canonical mitochondrial protein deacetylation functions.

These advanced techniques are helping to establish SIRT3 as a critical link between mitochondrial metabolism and nuclear epigenetic regulation, potentially explaining the wide-ranging effects of SIRT3 modulation on cellular physiology and disease processes .

Product Science Overview

Structure and Localization

SIRT3 is primarily localized in the mitochondrial matrix . It contains a mitochondrial processing peptide at the N-terminus, which is essential for its import into mitochondria . The protein has been crystallized in several forms, revealing its interaction with substrates and cofactors .

Function

SIRT3 is involved in the deacetylation of key mitochondrial proteins, thereby regulating their activity. Some of its known targets include:

  • Acetyl-CoA synthetase 2 (ACSS2)
  • Isocitrate dehydrogenase 2 (IDH2)
  • Glutamate dehydrogenase (GDH)

By deacetylating these proteins, SIRT3 enhances their activity, which is crucial for mitochondrial function and energy production .

Role in Metabolism and Aging

SIRT3 has emerged as an important regulator of mitochondrial metabolism. Overexpression of SIRT3 in cultured cells has been shown to increase respiration and decrease the production of reactive oxygen species (ROS) . This suggests that SIRT3 plays a protective role against oxidative stress, which is a key factor in aging and various diseases .

Recombinant SIRT3

Recombinant human SIRT3 is typically expressed in E. coli and purified for research purposes . It is used to study the enzyme’s activity, structure, and potential therapeutic applications. The recombinant protein is often tagged with a 6-His tag to facilitate purification .

Applications in Research

Recombinant SIRT3 is widely used in biochemical assays to understand its enzymatic activity and interaction with substrates. It is also employed in structural studies to elucidate the conformational changes upon substrate binding .

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.