TFB1M Human

Transcription Factor B1, Mitochondrial Human Recombinant
Shipped with Ice Packs
In Stock

Description

Overview of TFB1M Human

TFB1M (Transcription Factor B1, Mitochondrial) is a mitochondrial enzyme encoded by the TFB1M gene located on human chromosome 6. It belongs to the methyltransferase superfamily and plays a dual role:

  • rRNA Dimethylation: Catalyzes the dimethylation of two conserved adenine residues (m⁶²A) in helix 45 (h45) of mitochondrial 12S rRNA, essential for ribosome assembly and translation of mitochondrial genes .

  • Transcriptional Regulation: Interacts with mitochondrial RNA polymerase (POLRMT) and transcription factor A (TFAM) to modulate basal transcription, though this activity is less pronounced compared to its paralog, TFB2M .

Table 2: Disease Implications of TFB1M Dysregulation

DiseasePathogenic MechanismEvidence Source
Type 2 DiabetesLoss of m⁶²A in h45 → Ribosome dysfunction → β-cell failure
HCCTFB1M overexpression → Glycolytic shift → Tumor growth

Therapeutic Targets

  • m⁶²A Modification: Targeting TFB1M activity could modulate mitochondrial translation in metabolic disorders.

  • Mitochondrial Biogenesis: Restoring TFB1M function may improve OXPHOS efficiency in diseases like neurodegeneration.

Diagnostic Biomarkers

  • Genetic Variants: TFB1M risk alleles (e.g., rs12536695) predict diabetes susceptibility .

  • Protein Levels: Elevated TFB1M in HCC tissues may serve as a prognostic marker .

Product Specs

Introduction

As a member of the methyltransferase superfamily, TFB1M functions as a dimethyltransferase that specifically methylates mitochondrial 12S rRNA at its conserved stem loop. This protein is an integral component of the basal mitochondrial transcription complex and plays a critical role in mitochondrial gene expression. Importantly, TFB1M exhibits the ability to stimulate transcription independent of its methyltransferase activity.

Description

Recombinant TFB1M Human, produced in E. coli, is a single, non-glycosylated polypeptide chain consisting of 340 amino acids (28-346 a.a.). With a molecular weight of 38.8 kDa, it features a 21 amino acid His-tag fused at the N-terminus. Purification is achieved through proprietary chromatographic techniques.

Physical Appearance
A clear solution that has undergone sterile filtration.
Formulation

The TFB1M protein solution is provided at a concentration of 0.5 mg/ml. It is formulated in a buffer containing 20 mM Tris-HCl (pH 8.0), 20% glycerol, 0.1 M NaCl, and 1 mM DTT.

Stability

For short-term storage (2-4 weeks), the product should be kept at 4°C. For extended storage, freezing at -20°C is recommended. To ensure optimal stability during long-term storage, the addition of a carrier protein (0.1% HSA or BSA) is advised. It is important to avoid repeated freeze-thaw cycles to maintain product integrity.

Purity
The purity of the TFB1M protein is determined to be greater than 90.0% based on SDS-PAGE analysis.
Synonyms

Dimethyladenosine transferase 1 mitochondrial, Mitochondrial 12S rRNA dimethylase 1, Mitochondrial transcription factor B1, h-mtTFB, h-mtTFB1, hTFB1M, mtTFB1, S-adenosylmethionine-6-N'-N'-adenosyl(rRNA) dimethyltransferase 1, TFB1M, CGI-75, CGI75, mtTFB.

Source
Escherichia Coli.
Amino Acid Sequence

MGSSHHHHHH SSGLVPRGSH MQAAKQLSQN FLLDLRLTDK IVRKAGNLTN AYVYEVGPGP GGITRSILNA DVAELLVVEK DTRFIPGLQM LSDAAPGKLR IVHGDVLTFK VEKAFSESLK RPWEDDPPNV HIIGNLPFSV STPLIIKWLE NISCRDGPFV YGRTQMTLTF QKEVAERLAA NTGSKQRSRL SVMAQYLCNV RHIFTIPGQA FVPKPEVDVG VVHFTPLIQP KIEQPFKLVE KVVQNVFQFR RKYCHRGLRM LFPEAQRLES TGRLLELADI DPTLRPRQLS ISHFKSLCDV YRKMCDEDPQ LFAYNFREEL KRRKSKNEEK EEDDAENYRL.

Q&A

What is TFB1M and what is its primary function in human mitochondria?

TFB1M (Transcription Factor B1 Mitochondrial) is a dual-function protein that serves as both a mitochondrial transcription specificity factor and a dimethyltransferase. Its primary biochemical function is catalyzing the dimethylation of two adjacent adenines located in helix 45 (h45) of the 12S ribosomal RNA (rRNA) . This m62A modification is indispensable for the assembly and maturation of human mitochondrial ribosomes.

As a transcription factor, TFB1M works in concert with mitochondrial RNA polymerase and Tfam (a DNA binding stimulatory factor) to enhance mitochondrial DNA (mtDNA) transcription . It belongs to a family that includes TFB2M, with both proteins markedly enhancing mtDNA transcription, though TFB1M demonstrates approximately 1/10 the transcriptional activity of TFB2M .

The dual role of TFB1M highlights its critical importance in mitochondrial biogenesis, as it connects transcriptional regulation with translational control through ribosome assembly.

How is TFB1M expression regulated in human cells?

TFB1M expression is governed by a sophisticated regulatory network that integrates mitochondrial biogenesis with cellular energy demands. Key components of this regulatory pathway include:

  • Nuclear Respiratory Factors (NRFs): The expression of human TFB1M promoters is directly governed by NRF-1 and NRF-2, key transcription factors implicated in mitochondrial biogenesis . These NRF recognition sites within the TFB1M promoter are required for maximal expression.

  • PGC-1 Family Coactivators: TFB1M promoters are trans-activated by PGC-1 family coactivators, including PGC-1α and PRC . The physiological induction of these coactivators has been associated with the integration of NRFs and other transcription factors in a program of mitochondrial biogenesis.

  • Coordinate Regulation: TFB1M is up-regulated alongside Tfam and either PGC-1α or PRC in cellular systems where mitochondrial biogenesis is induced . This coordinate regulation ensures synchronized expression of all components necessary for mitochondrial function.

  • Promoter Structure: The human TFB1M promoter (accession number AL139101) contains specific binding sites for transcriptional regulators. The following oligonucleotide sequences are important for regulation:

    • TFB1M/NRF-1: GATCCGGACTTAGCGCATGCGCTCTCAGCA

    • TFB1M/NRF-2: GATCCGCCGGGAATTTCCTGTCCGCGGTCATCGCTTCCGGTGGGA

This regulatory framework demonstrates how nuclear factors coordinate the expression of nucleus-encoded mitochondrial transcription factors essential to the control of mitochondrial biogenesis.

What is the structural basis for TFB1M function?

Recent structural studies have provided critical insights into the mechanism of TFB1M-mediated rRNA modification. The crystal structures of both a ternary complex (hsTFB1M–h45–SAM) and a binary complex (hsTFB1M–h45) at 3.0 Å resolution have revealed several key features:

  • RNA Recognition Mechanism: The 'GGAA' tetraloop of 12S h45 flips out to interact with human TFB1M, revealing a distinct mode of interaction between the enzyme and its rRNA substrate .

  • Substrate Preference: The terminal adenosine in the 'GGAA' tetraloop, specifically m.937A, is favorably positioned for modification by TFB1M because it enters the enzyme active site in the initial state .

  • Active Site Architecture: The enzyme active site contains specific residues that determine binding affinity and catalytic activity, as confirmed through binding affinity and enzyme activity assays .

These structural insights have significantly advanced our understanding of how TFB1M recognizes its substrate and performs the dimethylation reaction essential for mitochondrial ribosome assembly and function.

What are the methodological approaches to study TFB1M dimethyltransferase activity?

Studying TFB1M dimethyltransferase activity requires a multidisciplinary approach combining biochemical, structural, and cellular techniques:

Structural Analysis Methodology:

  • X-ray Crystallography: Crystal structures of TFB1M complexes (such as hsTFB1M–h45–SAM) provide atomic-level insights into substrate recognition and catalytic mechanism .

  • NMR Spectroscopy: Solution structures of h45 with and without modifications can be compared to understand structural changes induced by TFB1M-mediated methylation .

Biochemical Assays:

  • Methyltransferase Activity Assays: Using radiolabeled S-adenosylmethionine (SAM) or SAM analogs to measure the transfer of methyl groups to target rRNA substrates.

  • Binding Affinity Measurements: Techniques such as surface plasmon resonance or isothermal titration calorimetry can quantify TFB1M-substrate interactions and identify critical residues .

Cellular and Molecular Approaches:

  • Site-Directed Mutagenesis: Creating TFB1M mutants affecting residues in the active site or RNA-binding interface to evaluate their impact on enzyme function .

  • Mass Spectrometry: Analysis of methylation patterns in isolated mitochondrial ribosomes to identify and quantify m62A modifications.

  • Suppression Techniques: Reducing TFB1M expression through RNAi or CRISPR to evaluate downstream effects on mitochondrial function and ribosome assembly .

Researchers should consider combining these approaches to comprehensively characterize TFB1M activity and its biological consequences.

How does TFB1M deficiency affect mitochondrial protein synthesis and function?

Effects on Mitochondrial Translation and Structure:

  • Reduced Protein Synthesis: Loss of TFB1M results in decreased levels of mitochondrially encoded proteins, despite normal transcription of the corresponding genes .

  • Aberrant Mitochondrial Morphology: Mitochondria in TFB1M-deficient cells are more abundant but display disrupted architecture .

  • Ribosome Assembly Defects: The m62A modification catalyzed by TFB1M is indispensable for the assembly and maturation of human mitochondrial ribosomes .

Functional Consequences:

  • ATP Production: TFB1M-deficient cells show reduced ATP production and oxygen consumption rates .

  • Oxidative Stress Response: Levels of reactive oxygen species in response to cellular stress are increased while induction of defense mechanisms is attenuated in TFB1M-deficient cells .

  • Cell Death Pathways: Increased apoptosis and necrosis have been observed, along with infiltration of immune cells (macrophages and CD4+ cells) in affected tissues .

Tissue-Specific Effects:
The β-cell specific knockout (β-Tfb1m-/-) mouse model demonstrates that:

  • TFB1M deficiency in pancreatic β-cells leads to impaired glucose-stimulated insulin secretion

  • Islets contain less insulin and fewer secretory granules

  • Reduced β-cell mass occurs over time

  • These cellular defects ultimately progress to diabetes

What are the implications of TFB1M variants in metabolic diseases like Type 2 Diabetes?

TFB1M has emerged as a significant gene in the pathogenesis of Type 2 Diabetes (T2D), with both human genetic studies and experimental models providing compelling evidence:

Human Genetic Evidence:

  • Risk Variant Identification: TFB1M has been identified as a T2D risk gene through human genetic studies .

  • Gene-Dosage Effect: The TFB1M risk variant exhibits a negative gene-dosage effect on islet TFB1M mRNA levels and insulin secretion . This indicates that the variant affects TFB1M expression levels, which directly impacts β-cell function.

Mechanistic Insights from Animal Models:

  • Progressive Diabetes Development: β-cell specific knockout of Tfb1m in mice (β-Tfb1m-/- mice) leads to gradual development of diabetes .

  • Impaired Glucose Metabolism: Prior to the onset of diabetes, β-Tfb1m-/- mice exhibit retarded glucose clearance due to impaired insulin secretion .

  • Molecular Basis: The pathway from TFB1M deficiency to diabetes involves:

    • Reduced mitochondrial 12S rRNA methylation

    • Decreased ATP production

    • Impaired oxygen consumption

    • Compromised insulin secretion in response to metabolic fuels

Clinical Implications:

  • Predictive Value: The risk variant can predict the occurrence of T2D in future, as revealed by analysis of a prospective cohort .

  • Therapeutic Target Potential: Understanding TFB1M's role in mitochondrial function offers new avenues for therapeutic interventions targeting mitochondrial dysfunction in diabetes.

  • Biomarker Development: TFB1M expression levels or activity could potentially serve as biomarkers for mitochondrial dysfunction in at-risk individuals.

How do researchers resolve discrepancies in TFB1M knockout models across different species?

Researchers face significant challenges when interpreting seemingly contradictory results from TFB1M knockout models across different species and experimental systems:

Observed Discrepancies:

  • Lethal vs. Viable Phenotypes: Homozygous Tfb1m-/- knockout mice show a lethal embryonic phenotype , while tissue-specific knockouts demonstrate varying degrees of dysfunction .

  • Overexpression Effects: Overexpression of TFB1M in mice showed no impact on mitoribosomal methylation status or hearing , suggesting potential compensatory mechanisms or context-dependent functions.

  • Species Differences: The functional consequences of TFB1M deficiency may vary between model organisms due to differences in mitochondrial genome organization and expression.

Methodological Approaches to Resolve Discrepancies:

  • Conditional Knockout Systems: Using tissue-specific and inducible knockout models to bypass embryonic lethality and study TFB1M function in adult tissues .

  • Dose-Dependency Analysis: Examining the effects of partial reduction versus complete elimination of TFB1M to establish threshold effects on mitochondrial function.

  • Comprehensive Phenotyping: Detailed characterization of molecular, cellular, and physiological parameters across multiple tissues in knockout models.

  • Cross-Species Comparison: Systematic comparison of TFB1M function, expression patterns, and protein interactions across species to identify conserved and divergent aspects.

  • Integration with Human Data: Correlating findings from model organisms with human genetic studies and ex vivo analyses of human tissues to validate physiological relevance .

By applying these approaches, researchers can better understand the context-dependent roles of TFB1M and resolve apparent contradictions in experimental findings, ultimately generating more accurate models of how TFB1M dysfunction contributes to human disease.

What experimental approaches can identify novel interacting partners of TFB1M in the mitochondrial gene expression machinery?

Elucidating the complete interactome of TFB1M is essential for understanding its roles beyond its established functions. Several complementary experimental approaches can be employed:

Protein-Protein Interaction Discovery Methods:

  • Proximity-Based Labeling: BioID or APEX2 fusion proteins can identify proteins in close proximity to TFB1M within the mitochondrial environment.

  • Co-Immunoprecipitation with Mass Spectrometry: Pull-down of endogenous or tagged TFB1M followed by mass spectrometry analysis to identify interacting proteins. This technique has already revealed interaction between TFB1M and the carboxy-terminal domain of Tfam .

  • Yeast Two-Hybrid Screening: Using mitochondrial protein libraries to identify direct binding partners, though this approach needs to account for the mitochondrial localization of TFB1M.

Protein-RNA Interaction Methods:

  • CLIP-Seq Variants: Cross-linking immunoprecipitation followed by sequencing to identify RNA targets beyond the known 12S rRNA substrate.

  • RNA Electrophoretic Mobility Shift Assays (EMSA): To characterize binding specificity and affinity for various RNA structures .

  • RNA Pull-Down Assays: Using tagged RNA substrates to identify proteins that co-bind with TFB1M to the same RNA regions.

Functional Validation Approaches:

  • Genetic Interaction Screens: RNAi or CRISPR-based screens to identify genes that show synthetic lethality or rescue effects with TFB1M depletion.

  • Fluorescence Microscopy: Using techniques like Förster resonance energy transfer (FRET) or bimolecular fluorescence complementation (BiFC) to validate interactions in live cells.

  • Mitochondrial Ribosome Profiling: Assessing how TFB1M and its partners affect the landscape of mitochondrial translation.

Data Integration Framework:
The most comprehensive approach would integrate data from multiple experimental platforms with computational predictions to build a high-confidence interactome map of TFB1M within the mitochondrial gene expression machinery.

These methods collectively provide a powerful toolkit for discovering novel TFB1M interactions that may reveal unexpected functions and regulatory mechanisms in mitochondrial biology.

How can targeting TFB1M function be utilized in developing therapeutic strategies for mitochondrial dysfunction?

Given the critical role of TFB1M in mitochondrial function, several therapeutic approaches targeting this pathway could be developed:

Potential Therapeutic Strategies:

  • Gene Therapy Approaches: Delivery of functional TFB1M to tissues with deficient expression or function, particularly relevant for metabolic diseases like T2D where reduced TFB1M activity has been implicated .

  • Small Molecule Modulators: Development of compounds that can enhance TFB1M methyltransferase activity or stabilize the methylated 12S rRNA structure to improve mitochondrial translation efficiency.

  • RNA-Based Therapies: Antisense oligonucleotides or RNA aptamers designed to stabilize the interaction between TFB1M and its RNA substrates in cases where binding is compromised.

Therapeutic Target Validation Methodology:

  • High-Throughput Screening Platforms: Development of assays that measure TFB1M methyltransferase activity or mitochondrial translation efficiency in disease models.

  • Structure-Based Drug Design: Utilizing the crystal structures of hsTFB1M complexes to identify potential binding pockets for small molecule modulators .

  • Organoid and Patient-Derived Cell Models: Testing therapeutic approaches in complex tissue systems that better recapitulate human disease conditions.

Challenges and Considerations:

  • Mitochondrial Targeting: Ensuring therapeutic agents reach mitochondria efficiently.

  • Tissue Specificity: Developing delivery systems that target tissues most affected by TFB1M dysfunction, such as pancreatic β-cells in T2D .

  • Safety Assessment: Careful evaluation of potential off-target effects, given that complete loss of TFB1M is embryonically lethal in mice .

Future research in this area holds promise for addressing mitochondrial dysfunction in various diseases, particularly metabolic disorders where TFB1M deficiency has been directly implicated in pathogenesis.

What models best replicate human TFB1M dysfunction for translational research?

Selecting appropriate models to study TFB1M dysfunction is critical for translational relevance. Various models offer complementary advantages:

Cellular Models:

  • Patient-Derived Fibroblasts: Cells from individuals with TFB1M variants or mutations provide directly relevant human cellular contexts.

  • CRISPR-Modified Human Cell Lines: Engineered cell lines with specific TFB1M mutations or expression levels can model discrete aspects of dysfunction.

  • iPSC-Derived Specialized Cells: Induced pluripotent stem cells differentiated into relevant cell types (e.g., β-cells, neurons) can model tissue-specific effects of TFB1M dysfunction .

Animal Models:

  • Tissue-Specific Conditional Knockouts: The β-cell specific knockout (β-Tfb1m-/-) mouse has proven valuable for studying TFB1M's role in diabetes pathogenesis .

  • Hypomorphic Alleles: Models with reduced rather than absent TFB1M may better replicate human variants with partial loss of function.

  • Humanized Mice: Replacing mouse Tfb1m with human TFB1M could provide better translational relevance for testing therapeutics.

Ex Vivo Human Tissue:

  • Islet Preparations: Human pancreatic islets can be used to study the effects of TFB1M variants on insulin secretion and mitochondrial function .

  • Tissue Microarrays: Can evaluate TFB1M expression patterns across multiple human tissues and disease states.

Comparative Model Evaluation Table:

Model TypeAdvantagesLimitationsBest Applications
β-Tfb1m-/- miceProgressive diabetes development; permits in vivo studiesSpecies differences in metabolismDiabetes pathogenesis studies
Human islets with TFB1M variantsDirect human relevanceLimited availability; short lifespan in cultureValidation of mechanisms found in animal models
CRISPR-edited cell linesPrecise genetic manipulation; high throughputSimplified cellular environmentMolecular mechanism studies
iPSC-derived β-cellsHuman genetics; differentiated cell typeImmature phenotype compared to adult cellsDrug screening; personalized medicine approaches

The ideal approach involves using multiple complementary models, with findings in simpler systems validated in more complex ones, ultimately translating to human tissues when possible.

What are the challenges in analyzing TFB1M-mediated rRNA modifications in human samples?

Analyzing TFB1M-mediated m62A modifications in human samples presents several technical challenges that researchers must address:

Analytical Challenges:

  • Low Abundance of Modification: The m62A modification occurs at specific positions in mitochondrial 12S rRNA, making detection challenging due to the relatively low abundance of modified nucleotides in total cellular RNA.

  • Sample Preparation Issues: Isolation of intact mitochondrial ribosomes from limited human samples without contamination from cytosolic ribosomes requires careful fractionation techniques.

  • Methylation Stability: The dimethyladenosine modification can be unstable under certain experimental conditions, potentially leading to underestimation of modification levels.

Methodological Solutions:

  • Mass Spectrometry Approaches: Liquid chromatography-tandem mass spectrometry (LC-MS/MS) provides sensitive and specific detection of m62A modifications, but requires specialized equipment and expertise .

  • Antibody-Based Detection: Development of antibodies specific to m62A can enable immunoprecipitation of modified rRNA fragments, though cross-reactivity must be carefully controlled.

  • Next-Generation Sequencing Methods:

    • Direct RNA sequencing technologies that can detect modifications

    • Specialized library preparation methods that capture modification-induced signatures

  • Single-Cell Analysis: Emerging technologies for analyzing RNA modifications at the single-cell level could reveal cell-to-cell variation in TFB1M activity.

Quality Control Considerations:

  • Reference Standards: Use of synthetic RNA oligonucleotides containing m62A modifications as positive controls.

  • Validation Across Methods: Confirmation of results using orthogonal techniques to ensure reliability.

  • Quantification Accuracy: Development of standard curves and internal controls to enable accurate quantification of modification levels.

By addressing these technical challenges, researchers can more accurately assess TFB1M activity in various human tissues and disease states, enhancing our understanding of how this modification affects mitochondrial function in health and disease.

How can researchers effectively measure the impact of TFB1M on mitochondrial protein synthesis?

Assessing the impact of TFB1M on mitochondrial protein synthesis requires a combination of approaches that capture both direct and indirect effects:

Direct Translation Measurement Methods:

  • Mitochondrial Ribosome Profiling: This technique provides a genome-wide snapshot of active translation by capturing ribosome-protected mRNA fragments specifically from mitochondrial ribosomes .

  • Metabolic Labeling: Using radioactive amino acids (35S-methionine/cysteine) or non-canonical amino acids (like azidohomoalanine) combined with mitochondrial isolation to specifically detect newly synthesized mitochondrial proteins.

  • Polysome Analysis: Sucrose gradient fractionation of mitochondrial ribosomes to assess changes in translation initiation and elongation efficiency with altered TFB1M function.

Functional Readouts:

  • Respiratory Complex Assembly and Activity: Blue native polyacrylamide gel electrophoresis (BN-PAGE) combined with in-gel activity assays or spectrophotometric measurements of individual respiratory complexes .

  • ATP Production Measurement: Both cellular ATP levels and mitochondrial ATP synthesis capacity can be quantified using luciferase-based assays .

  • Oxygen Consumption Analysis: High-resolution respirometry to measure oxygen consumption rates as an indicator of oxidative phosphorylation efficiency .

Methodological Workflow for Comprehensive Analysis:

  • Experimental Design Considerations:

    • Use of appropriate controls (wild-type, heterozygous, and homozygous mutants)

    • Time-course experiments to capture progressive effects

    • Tissue-specific analyses to identify differential sensitivities

  • Data Integration Framework:

    • Correlating translational efficiency with protein abundance

    • Linking protein synthesis defects to functional outcomes

    • Computational modeling to predict threshold effects

This multi-faceted approach allows researchers to establish clear connections between TFB1M-mediated rRNA modification, mitochondrial translation efficiency, and downstream functional consequences in various experimental systems and disease models.

How might TFB1M function be affected by environmental factors and stress conditions?

The regulation and function of TFB1M may be significantly modulated by environmental factors and cellular stress conditions, representing an important frontier in mitochondrial research:

Stress Responses and TFB1M Regulation:

  • Oxidative Stress: TFB1M-deficient cells show increased levels of reactive oxygen species (ROS) in response to cellular stress, suggesting a potential regulatory feedback loop . Research should investigate whether oxidative stress itself alters TFB1M expression or activity.

  • Nutrient Availability: Given TFB1M's role in mitochondrial biogenesis and its regulation by PGC-1 family coactivators , nutrient-sensing pathways likely influence TFB1M function. Studies examining TFB1M response to caloric restriction or excess could reveal important regulatory mechanisms.

  • Inflammatory Signals: The observation of macrophage and CD4+ T-cell infiltration in TFB1M-deficient tissues suggests potential interactions between inflammatory pathways and TFB1M function that warrant further investigation.

Experimental Approaches to Study Environmental Influences:

  • Exposure Models: Exposing cells or organisms with varying TFB1M levels to different stressors (oxidants, nutrient deprivation, inflammatory cytokines) to assess changes in TFB1M expression, localization, and activity.

  • Epigenetic Analysis: Investigating whether environmental factors induce epigenetic modifications at the TFB1M locus that affect its expression.

  • Post-Translational Modification Profiling: Mass spectrometry-based approaches to identify stress-induced modifications of TFB1M protein that might alter its function.

  • Transcriptional Network Analysis: Examining how stress-responsive transcription factors interact with the regulatory elements in the TFB1M promoter, particularly focusing on the NRF-1 and NRF-2 binding sites .

Understanding how environmental factors influence TFB1M function could provide crucial insights into mitochondrial adaptations to stress and potentially reveal new therapeutic targets for conditions associated with mitochondrial dysfunction.

What is the evolutionary significance of TFB1M's dual role in transcription and rRNA modification?

The evolutionary history of TFB1M provides important context for understanding its functional significance in human mitochondria:

Evolutionary Origins:

  • Ancestral Functions: TFB1M is related to rRNA methyltransferases , suggesting that its rRNA modification function may be the ancestral role from which its transcriptional function evolved.

  • Functional Divergence: In mammals, TFB1M has lower transcriptional activity compared to TFB2M (approximately 1/10) , potentially indicating evolutionary specialization toward its methyltransferase function.

Comparative Analysis Across Species:

  • Functional Conservation: Studying the relative importance of transcriptional versus methyltransferase activities across evolutionary distant species could reveal selective pressures that shaped TFB1M function.

  • Structural Adaptations: Comparing the structures of TFB1M orthologs across species could identify conserved domains essential for each function and reveal how the dual functionality is maintained.

  • Genetic Compensation: Examining species with single versus multiple TFB homologs to understand how functional redundancy and specialization evolved.

Evolutionary Hypotheses:

  • Co-regulatory Advantage: The dual function may provide an evolutionary advantage by coordinating transcription with translation through direct regulation of ribosome assembly.

  • Metabolic Adaptation: Changes in TFB1M function across species may reflect adaptations to different metabolic demands and mitochondrial genome organizations.

  • Constraint-Based Evolution: The essential nature of both functions may have constrained evolutionary divergence, maintaining the dual role despite potential efficiency trade-offs.

Product Science Overview

Gene and Protein Structure

TFB1M is encoded by the TFB1M gene, which is located on chromosome 6q25.3 . The protein consists of 346 amino acids and shares homology with bacterial rRNA dimethyltransferases . This homology suggests that TFB1M has evolved from an RNA-modifying enzyme, which is reflected in its dual functionality.

Function and Mechanism

TFB1M is a part of the basal mitochondrial transcription complex. It works in conjunction with mitochondrial RNA polymerase (POLRMT) and mitochondrial transcription factor A (TFAM) to initiate and regulate the transcription of mtDNA . TFB1M binds to the mitochondrial light strand promoter (LSP) and enhances transcription from LSP by TFAM .

In addition to its role in transcription, TFB1M also functions as a dimethyltransferase. It methylates the conserved stem loop of mitochondrial 12S rRNA, which is necessary for the assembly or stability of the small subunit of the mitochondrial ribosome . This methylation is crucial for normal mitochondrial translation, metabolism, and cell growth .

Expression and Localization

TFB1M is ubiquitously expressed in human tissues and localizes to the mitochondria . When transfected into HeLa cells, fluorescence-labeled TFB1M colocalizes with mitochondrial markers, indicating its specific localization within the mitochondria .

Clinical Significance

Mutations or dysregulation of TFB1M can lead to mitochondrial dysfunction, which is associated with various human diseases. For example, TFB1M has been linked to Bardet-Biedl Syndrome 18 and progressive leukoencephalopathy with ovarian failure . Understanding the function and regulation of TFB1M is therefore critical for developing therapeutic interventions for mitochondrial-related diseases.

Research and Therapeutic Potential

Research on TFB1M continues to uncover its multifaceted roles in mitochondrial biology. The protein’s ability to act both as a transcription factor and an rRNA modification enzyme makes it a unique target for therapeutic interventions aimed at correcting mitochondrial dysfunctions .

In conclusion, Transcription Factor B1, Mitochondrial (Human Recombinant) is a vital component of the mitochondrial transcription machinery. Its dual role in transcription and rRNA modification underscores its importance in maintaining mitochondrial function and cellular energy metabolism.

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.