Methionine Sulfoxide Reductase B2 (MSRB2), encoded by the MSRB2 gene, is a mitochondrial matrix enzyme critical for maintaining protein integrity under oxidative stress. It catalyzes the reduction of methionine sulfoxide (MetO) to methionine, restoring function to oxidized proteins. MSRB2 is distinct from other methionine sulfoxide reductases (e.g., MsrA, MsrB1, MsrB3) due to its exclusive mitochondrial localization and specialized roles in mitophagy and cytokinesis .
MSRB2 is a critical regulator of mitochondrial quality control via mitophagy, particularly in oxidative stress conditions.
Parkin Activation:
LC3 Interaction:
Switch Mechanism:
MSRB2 regulates abscission timing by interacting with Aurora B and ANCHR:
Hyperglycemia: Induces oxidative stress → ↑ MSRB2 expression → ↑ mitophagy → ↓ platelet apoptosis .
Proteomic Data: Met192 oxidation in Parkin is prevalent in diabetic platelets, mitigated by MSRB2 .
Parkin Mutations: Mutations at Met192 (e.g., M192L/V) impair Parkin function, linked to disease progression .
Patient Studies: ↓ MSRB2 correlates with ↓ mitophagy and ↑ platelet apoptosis in PD patients .
MSRB2 primarily functions as a methionine sulfoxide reductase that reduces oxidized methionine residues (MetO) back to their native state. Unlike other methionine sulfoxide reductases that can be found in multiple cellular compartments, MSRB2 was originally characterized as being exclusively located in the mitochondrial matrix . Its enzymatic activity is particularly important in high oxidative stress environments where it can protect proteins from oxidative damage by restoring methionine residues. This redox function is critical for maintaining proper protein structure and function, especially in mitochondria where reactive oxygen species (ROS) production is high. Methodologically, researchers can assess MSRB2 activity using specific substrates containing methionine sulfoxide and measuring the reduction rate spectrophotometrically.
To effectively study MSRB2 in its dual localization patterns, researchers should employ a multi-method approach:
Immunofluorescence with confocal microscopy: Using specific anti-MSRB2 antibodies along with mitochondrial markers (e.g., MitoTracker) to visualize colocalization patterns. High-resolution confocal microscopy has successfully demonstrated MSRB2 and LC3 colocalization in diabetic platelets .
Immunoelectron microscopy: This technique provides ultrastructural resolution and has been used to confirm MSRB2 localization, using small (5 nm) gold particles for MSRB2 and larger (15 nm) particles for interaction partners like LC3 .
Subcellular fractionation with Western blotting: Separating mitochondrial, cytosolic, and nuclear fractions followed by immunoblotting.
Live-cell imaging: Using fluorescently tagged MSRB2 constructs to track its dynamic localization, particularly important for studying its translocation during mitochondrial damage or cell division.
Proximity ligation assays: To detect in situ protein-protein interactions between MSRB2 and its binding partners in different cellular compartments.
MSRB2 serves as both a switch and transducer for mitophagy through a sophisticated mechanism:
As a switch: MSRB2 is normally sequestered in the mitochondrial matrix, but upon mitochondrial damage, it is released from ruptured mitochondria. Once released, it reduces oxidized methionine residues on Parkin (specifically MetO192), restoring Parkin's function . This reduction prevents Parkin aggregation and enables its E3 ligase activity.
As a transducer: After activating Parkin, MSRB2 becomes ubiquitinated by Parkin and subsequently interacts with LC3 through its LC3-interacting motif (LIM), facilitating autophagosome formation around the damaged mitochondrion .
This dual functionality ensures that mitophagy proceeds selectively at severely damaged or ruptured mitochondria, providing a quality control mechanism. Methodologically, researchers can investigate this mechanism using mitochondrial damage models (e.g., CCCP treatment), followed by co-immunoprecipitation of MSRB2 with Parkin and LC3, and assessment of ubiquitination patterns.
To quantitatively assess MSRB2-mediated mitophagy, researchers should employ multiple complementary approaches:
LC3-II/LC3-I ratio analysis: Western blotting to measure the conversion of LC3-I to LC3-II, with a focus on the lower LC3-II band that increases during mitophagy .
Mitochondrial mass quantification: Using mitochondrial markers (e.g., TOMM20, ATPB) to measure mitochondrial content via immunoblotting or flow cytometry.
Mitochondrial membrane potential assessment: Using fluorescent dyes like TMRE to measure mitochondrial membrane potential, which decreases during mitophagy .
Co-immunoprecipitation assays: To detect interactions between MSRB2, Parkin, and LC3 during mitophagy.
mPTP opening assessment: Using cyclosporine A (CsA) to inhibit mitochondrial permeability transition pore (mPTP) opening and measuring MSRB2 release from mitochondria .
Genetic approaches: Comparing wild-type cells with MSRB2 knockout or knockdown models, assessing changes in mitophagy markers and mitochondrial function.
Two key pathophysiological conditions show significant alterations in MSRB2 expression and function:
Diabetes Mellitus (DM):
Parkinson's Disease (PD):
These contrasting patterns suggest that MSRB2 expression levels can serve as potential biomarkers for disease states. Methodologically, researchers can analyze MSRB2 expression in patient samples using Western blotting, qPCR, and immunofluorescence, comparing with age-matched controls and correlating with disease severity markers.
Recent research has revealed that MSRB2 plays a critical role in cytokinesis, specifically controlling abscission timing through several mechanisms:
F-actin regulation: MSRB2 counteracts MICAL1-mediated actin oxidation. Depletion of MSRB2 leads to diminished F-actin levels in intercellular bridges (ICBs), whereas MICAL1 depletion increases F-actin. Co-depletion of both proteins restores normal F-actin levels .
ESCRT-III recruitment: MSRB2 influences the localization of ESCRT-III components (specifically CHMP4B) at the abscission site. MSRB2 depletion increases the proportion of late ICBs with CHMP4B at both the midbody and abscission site .
Abscission timing: MSRB2 depletion accelerates abscission, while MICAL1 depletion delays it. Co-depletion of both proteins restores normal timing .
These findings indicate that MSRB2 controls the timing of abscission, F-actin levels, and intercellular bridge stability. Methodologically, researchers can study these effects using live-cell imaging of fluorescently labeled cells, measuring the time from anaphase onset to abscission.
MSRB2 functions as a component of the abscission checkpoint, with several lines of evidence supporting this role:
Colocalization with checkpoint components: MSRB2 colocalizes with active, phosphorylated Aurora B at the midbody when the checkpoint is activated by lagging chromatin .
Genetic interactions: The delay in abscission caused by MSRB2 overexpression is fully dependent on Aurora B activity. Aurora B inhibition abolishes the delay, suggesting they act in the same pathway .
Binucleation phenotype: MSRB2 depletion leads to the formation of binucleated cells in dividing cells with chromatin bridges, a phenotype observed when other core components of the abscission checkpoint (Aurora B, ALIX, and ANCHR) are inactivated .
Response to multiple checkpoint triggers: MSRB2 is involved in delaying abscission not only in the presence of chromatin bridges but also in response to nuclear pore defects, which activate the checkpoint independently of chromatin bridges .
Methodologically, researchers can investigate this relationship using immunofluorescence co-staining of MSRB2 with Aurora B and ANCHR, coupled with genetic manipulation experiments (depletion or overexpression) and specific inhibitors of checkpoint components.
Though MSRB2 was traditionally considered exclusively mitochondrial, research has revealed a distinct nonmitochondrial, cytosolic pool with unique properties:
Localization: While the canonical MSRB2 contains a mitochondrial targeting sequence (MTS) in its first 23 amino acids, the cytosolic pool lacks or bypasses this targeting sequence .
Function: Mitochondrial MSRB2 primarily functions in mitophagy regulation, while cytosolic MSRB2 controls cytokinesis and abscission timing .
Interaction partners: Mitochondrial MSRB2 interacts with Parkin and LC3 during mitophagy, whereas cytosolic MSRB2 interacts with Aurora B and ANCHR during cell division .
This dual localization makes MSRB2 unique among methionine sulfoxide reductases. Methodologically, researchers can distinguish between these pools using subcellular fractionation, localization-specific mutations, and compartment-specific interaction studies.
Several lines of evidence connect MSRB2 to Parkinson's disease (PD) pathophysiology:
Reduced expression: PD patients show significantly reduced MSRB2 levels compared to age-matched controls .
Mitophagy defects: This reduction in MSRB2 correlates with reduced LC3-II levels, indicating impaired mitophagy, which is a known contributor to PD pathogenesis .
Parkin mutations: Mutations at Met192 on Parkin (M192L and M192V), the same position that MSRB2 reduces from MetO in diabetes mellitus, are associated with Parkinson's disease .
Mechanistic connection: MSRB2 reduces oxidized Parkin, preventing its aggregation and allowing mitophagy to proceed. Without sufficient MSRB2, oxidized Parkin remains inactive, potentially contributing to the accumulation of damaged mitochondria characteristic of PD .
These findings suggest that enhancing MSRB2 function might be a novel treatment strategy for PD. Methodologically, researchers can investigate this connection using patient samples, PD animal models, and cell culture models with MSRB2 manipulation.
MSRB2 exhibits distinct expression patterns and functional characteristics in diabetic versus normal cellular environments:
This differential expression suggests an adaptive response to the high oxidative stress environment in diabetes. Methodologically, researchers can study these differences using diabetic animal models, high glucose treatment of cells, and patient samples, employing techniques like Western blotting, co-immunoprecipitation, and confocal microscopy.
To address the seemingly contradictory findings about MSRB2 localization (mitochondrial matrix versus cytosolic), researchers should consider these advanced methodological approaches:
Super-resolution microscopy: Techniques like STORM or PALM can provide nanoscale resolution to precisely localize MSRB2 in different cellular compartments.
Proximity-dependent biotinylation (BioID or APEX): These techniques can identify the protein neighbors of MSRB2 in different compartments, providing functional context.
Split-protein complementation assays: To verify protein-protein interactions in specific cellular locations.
Isoform-specific antibodies: Development of antibodies that can distinguish between potential MSRB2 isoforms that might have different localizations.
Mass spectrometry-based proteomics: To identify post-translational modifications that might affect localization or potential alternative translation start sites.
CRISPR-based tagging: Endogenous tagging of MSRB2 to track its native localization without overexpression artifacts.
These approaches, used in combination, can help reconcile the apparent contradiction between MSRB2's established mitochondrial localization and its newly discovered cytosolic functions.
Investigating MSRB2's enzymatic activity across different cellular compartments presents several methodological challenges:
Compartment-specific substrates: Identifying which methionine-containing proteins are MSRB2 substrates in mitochondria versus cytosol.
Redox environment differences: The distinct redox environments of mitochondria and cytosol may affect MSRB2 activity differently.
Cofactor availability: Different cellular compartments may have varying levels of cofactors needed for MSRB2 activity.
Temporal dynamics: MSRB2 may shuttle between compartments under specific conditions, making static analyses misleading.
Technical limitations: Preserving native redox states during cell fractionation and protein extraction can be challenging.
Researchers can address these challenges by developing compartment-specific activity assays, using genetically encoded redox sensors, and employing metabolic labeling techniques to track MSRB2 substrates in different cellular locations.
Based on the reduced MSRB2 expression observed in Parkinson's disease patients and its mechanistic connection to mitophagy, several therapeutic approaches could be developed:
Gene therapy approaches: Viral vector-mediated delivery of MSRB2 to affected tissues to restore mitophagy.
Small molecule enhancers: Compounds that increase MSRB2 expression or enhance its enzymatic activity.
Peptide mimetics: Development of peptides that mimic MSRB2's interaction with Parkin to prevent its aggregation.
Targeted protein degradation: Using proteolysis-targeting chimeras (PROTACs) to selectively degrade oxidized Parkin if MSRB2 cannot be effectively restored.
Metabolic modulators: Compounds that reduce oxidative stress specifically in mitochondria to decrease the burden on the MSRB2 system.
Methodologically, researchers should employ multiple model systems, including patient-derived iPSCs, organoids, and animal models, to test these approaches. Biomarker development to identify patients with MSRB2 deficiency would also be critical for clinical translation.
Methionine Sulfoxide Reductase B2 (MsrB2) is an enzyme that plays a crucial role in the repair of oxidatively damaged proteins. It is part of the methionine sulfoxide reductase (Msr) system, which is responsible for reducing methionine sulfoxide (MetO) back to methionine, thereby protecting cells from oxidative stress. MsrB2 specifically reduces the R-isomer of MetO, distinguishing it from MsrA, which reduces the S-isomer .
Oxidative stress is a condition characterized by excessive reactive oxygen species (ROS) that can damage cellular components, including proteins. Methionine residues in proteins are particularly susceptible to oxidation, forming MetO. The accumulation of MetO can impair protein function and contribute to various diseases, including neurodegenerative disorders, cardiovascular diseases, and diabetes .
MsrB2 is highly expressed in mitochondria, where it plays a vital role in maintaining mitochondrial function by repairing oxidized proteins. This is particularly important in tissues with high metabolic rates, such as the heart and brain .
Research has shown that MsrB2 has protective effects against several diseases. For instance, in diabetic cardiomyopathy, a condition characterized by heart damage due to diabetes, MsrB2 helps mitigate oxidative damage and improve cardiac function. Studies in diabetic mice have demonstrated that increased expression of MsrB2 in the heart can reduce ROS levels, decrease protein oxidation, and prevent cardiac fibrosis .
Additionally, MsrB2 has been implicated in neuroprotection. In models of neurodegenerative diseases, such as Alzheimer’s and Parkinson’s, MsrB2 helps protect neurons from oxidative damage, thereby preserving cognitive and motor functions .
Human recombinant MsrB2 is produced using recombinant DNA technology, which involves inserting the human MsrB2 gene into a suitable expression system, such as bacteria or yeast. This allows for the large-scale production of the enzyme for research and therapeutic purposes.
Recombinant MsrB2 is used in various studies to understand its function, regulation, and potential therapeutic applications. It is also employed in assays to measure the activity of the Msr system and to screen for compounds that can modulate its activity .