Resource

Online Inquiry

Tandem Mass Tag (TMT) Technology in Proteomics

What is tandem mass tag (TMT)?

Tandem Mass Tag (TMT) is a technology developed for proteomics analysis. The concept was first introduced and patented by Proteome Sciences PLC, a leading provider of protein biomarker discovery and validation services, in the early 2000s. The main objective of TMT is to enable the simultaneous identification and quantification of proteins in multiple samples in a single experiment, increasing accuracy and reducing experimental variability. This is typically used in biological and biomedical research.

The development of TMT has been characterized by constant enhancements to increase its multiplexing capabilities, thereby facilitating the comparison of protein expression levels across multiple samples. Early versions of TMT supported the analysis of two samples, but rapid advancements have expanded this to 11 samples with the latest TMT kits, a major breakthrough in the field of quantitative proteomics. Further evolution of TMT has been driven by improvements in mass spectrometry technology.

A significant milestone in the history of TMT was its increased usage and application in clinical research and biomarker discovery. Its ability to unbiasedly quantify thousands of proteins in multiple samples has revolutionized the field. Various improvements and novel applications of TMT have been published in many high-impact journals, further proving the impact of TMT on the scientific community.

Over the years, TMT has paved the way for significant advancements in proteomics, bioinformatics, and translational research. Its ability to provide more precise, accurate, and comprehensive analysis makes it a powerful tool in biological and biomedical research, especially in the era of big data and precision medicine.

Purpose of TMT Labeling

The purpose of TMT labeling in proteomics research is multifaceted and encompasses several key objectives aimed at enhancing the quantitative analysis of protein expression levels across multiple samples.

First and foremost, TMT labeling facilitates the accurate quantification of proteins present in complex biological samples. In many biological studies, researchers are interested in comparing protein expression levels between different experimental conditions, such as diseased versus healthy tissues or treated versus untreated cells. TMT labeling allows for the simultaneous analysis of multiple samples within a single mass spectrometry experiment, thereby enabling robust and high-throughput quantitative proteomics analysis. By incorporating isobaric chemical tags into peptides derived from protein digests, TMT labeling enables the differentiation and quantification of peptides from different samples based on their mass-to-charge ratios during mass spectrometry analysis.

Additionally, TMT labeling offers the advantage of multiplexing, allowing researchers to simultaneously analyze multiple samples in a single experiment. This multiplexing capability significantly reduces experimental time and sample consumption compared to traditional label-free or single-labeling methods. By labeling samples with distinct TMT reagents, researchers can pool them together for simultaneous analysis, thereby minimizing experimental variability and improving data reproducibility.

Moreover, TMT labeling enables the accurate quantification of protein abundance changes in response to various experimental perturbations or treatments. By quantifying the relative abundance of proteins across different conditions, researchers can gain insights into biological processes, identify potential biomarkers of disease, and elucidate the mechanisms underlying cellular responses to stimuli. TMT labeling can be applied to a wide range of biological samples, including cell lysates, tissue extracts, and biological fluids, making it a versatile tool for quantitative proteomics research in diverse biological contexts.

Tandem mass tag (TMT) labeling strategies and experimental workflowTandem mass tag (TMT) labeling strategies and experimental workflow (Deng et al., 2019).

What is the TMT method for proteomics?

The Tandem Mass Tag (TMT) method for proteomics involves a series of steps designed to facilitate the accurate quantification of proteins in complex biological samples. The experimental workflow typically includes sample preparation, TMT labeling, sample mixing, mass spectrometry (MS) analysis, and data interpretation.

1. Sample Preparation: The first step in the TMT workflow involves the preparation of biological samples for proteomic analysis. This may include cell lysis, protein extraction, and protein digestion to generate peptides suitable for mass spectrometry analysis. Sample preparation protocols may vary depending on the nature of the sample and the research question being addressed.

2. TMT Labeling: After sample preparation, peptides are labeled with isobaric TMT reagents. TMT reagents consist of a mass-balanced structure comprising three main components: a reporter group, a mass normalization group, and a reactive group. The reporter group imparts mass differences to the labeled peptides, allowing for their differentiation during mass spectrometry analysis. The mass normalization group ensures that all labeled peptides have similar overall mass, while the reactive group facilitates covalent attachment of the TMT reagent to peptide amino groups. Each TMT reagent contains a unique mass reporter ion that is released upon fragmentation during MS analysis, enabling quantification of the corresponding peptide. The labeling reaction is typically carried out under mild conditions to minimize side reactions and ensure efficient labeling of peptide amino groups.

3. Sample Mixing: Labeled peptide samples are then combined and mixed together in equal proportions. The multiplexing capability of TMT allows for the simultaneous analysis of multiple samples within a single mass spectrometry run. Sample mixing helps to reduce experimental variability and ensure accurate quantification of protein abundance changes across different conditions.

4. Mass Spectrometry Analysis: The mixed peptide samples are subjected to liquid chromatography-tandem mass spectrometry (LC-MS/MS) analysis. During LC-MS/MS, peptides are separated based on their physicochemical properties using liquid chromatography and subsequently ionized and fragmented in the mass spectrometer. The resulting fragment ions are then detected and analyzed to determine the sequence and abundance of peptides. TMT-labeled peptides produce characteristic reporter ions upon fragmentation, allowing for their quantification relative to internal standards.

5. Data Interpretation: Finally, the acquired MS data are processed and analyzed using bioinformatics tools to identify and quantify proteins. Quantitative information obtained from TMT reporter ions is used to compare protein expression levels across different samples and conditions. Statistical methods may be employed to identify significant differences in protein abundance and infer biological insights from the data.

Advantages and disadvantages of tandem mass tags

Advantages of Tandem Mass Tags (TMT)Disadvantages of Tandem Mass Tags (TMT)
1. Multiplexing capability: TMT allows for simultaneous analysis of multiple samples within a single experiment, reducing experimental time and sample consumption.1. Cost: TMT reagents can be expensive, especially for large-scale experiments involving numerous samples.
2. Accurate quantification: TMT enables precise and accurate quantification of protein abundance changes across different experimental conditions.2. Isobaric interference: Isobaric TMT reagents may exhibit interference from co-eluting peptides, leading to inaccuracies in quantification.
3. High-throughput analysis: TMT facilitates high-throughput proteomics analysis, enabling the comparison of protein expression levels in complex biological samples.3. Limited dynamic range: TMT labeling may have a limited dynamic range, particularly for low abundance proteins, leading to decreased sensitivity in quantification.
4. Reduced experimental variability: TMT minimizes experimental variability by labeling samples with distinct isotopic tags, improving data reproducibility.4. Sample complexity: TMT analysis of highly complex samples may result in increased sample complexity and data processing challenges.
5. Versatility: TMT can be applied to various biological samples, including cell lysates, tissue extracts, and biological fluids, making it a versatile tool for proteomics research.5. Technical expertise: TMT labeling requires technical expertise and optimization to achieve optimal labeling efficiency and quantification accuracy.
6. Insights into biological processes: TMT facilitates the identification of protein abundance changes associated with biological processes, disease states, and drug treatments, providing valuable insights into cellular pathways and mechanisms.6. Data interpretation: TMT data analysis may be complex and require sophisticated bioinformatics tools for accurate interpretation and biological insights.
7. Compatibility with mass spectrometry: TMT-labeled peptides are compatible with mass spectrometry analysis, allowing for comprehensive proteomic profiling and characterization.7. Compatibility with certain experimental conditions: TMT labeling may not be suitable for all experimental conditions or sample types, limiting its applicability in certain research areas.
8. Integration with other omics technologies: TMT can be integrated with other omics technologies, such as genomics and transcriptomics, to provide a holistic view of biological systems.

Applications of TMT technology

Biomarker Discovery:

TMT technology has revolutionized biomarker discovery by enabling the simultaneous quantification of protein expression levels across multiple samples. This is particularly valuable in the identification of disease-specific biomarkers for early diagnosis, prognosis, and monitoring of various conditions such as cancer, cardiovascular diseases, and neurological disorders. By comparing protein profiles between diseased and healthy tissues or bodily fluids, researchers can pinpoint proteins that are differentially expressed, providing valuable insights into disease mechanisms and potential therapeutic targets.

Drug Discovery and Development:

TMT technology plays a crucial role in drug discovery and development by elucidating the mechanisms of drug action, identifying off-target effects, and evaluating drug efficacy and toxicity. By quantifying changes in protein expression in response to drug treatment, researchers can uncover molecular pathways affected by drugs and identify novel drug targets. TMT-based proteomics can also be used to assess drug-induced changes in protein phosphorylation, protein-protein interactions, and post-translational modifications, providing comprehensive insights into drug mechanisms of action.

Systems Biology and Functional Proteomics:

TMT technology facilitates systems-level analysis of complex biological systems by quantifying changes in protein abundance and dynamics in response to physiological stimuli, environmental perturbations, or genetic alterations. Integrating TMT-based proteomics with other omics technologies, such as genomics, transcriptomics, and metabolomics, allows for a comprehensive understanding of cellular pathways and regulatory networks. This holistic approach is essential for unraveling the complexities of biological systems and identifying key players involved in health and disease.

Clinical Proteomics and Personalized Medicine:

TMT technology holds promise for advancing clinical proteomics and personalized medicine by enabling the identification of patient-specific biomarkers, disease subtypes, and therapeutic targets. TMT-based proteomics can be applied to clinical samples, such as blood, urine, and tissue biopsies, for diagnostic purposes, treatment selection, and monitoring of treatment response. By profiling protein expression patterns in individual patients, clinicians can tailor therapeutic interventions to optimize outcomes and minimize adverse effects, leading to more personalized and effective patient care.

Functional Genomics and Protein Interaction Networks:

TMT technology facilitates the mapping of protein interaction networks and functional pathways involved in various biological processes. By quantifying changes in protein abundance across different cellular states or experimental conditions, researchers can infer protein-protein interactions, regulatory networks, and signaling pathways. This information is invaluable for understanding the underlying mechanisms of cellular processes, disease pathogenesis, and drug responses.

Reference

  1. Deng, Liting, et al. "Amyloid β induces early changes in the ribosomal machinery, cytoskeletal organization and oxidative phosphorylation in retinal photoreceptor cells." Frontiers in Molecular Neuroscience 12 (2019): 24.
* For Research Use Only. Not for use in diagnostic procedures.
Our customer service representatives are available 24 hours a day, 7 days a week. Inquiry

Online Inquiry

Please submit a detailed description of your project. We will provide you with a customized project plan to meet your research requests. You can also send emails directly to for inquiries.

* Email
Phone
* Service & Products of Interest
Services Required and Project Description
* Verification Code
Verification Code