Translatome analysis helps researchers identify novel proteins and uncover how gene regulation drives disease at the protein-production level.
More importantly, it explains why RNA abundance often fails to predict protein output.
In recent years, translatome approaches—especially when integrated with transcriptomics—have gained strong traction.
They have delivered valuable insights across cancer biology, developmental regulation, neuroscience, and metabolic disease research.
From our experience supporting translational profiling projects across oncology and developmental biology, integrating Ribo-seq with RNA-seq consistently reveals regulatory layers missed by transcriptomics alone.
Below, we highlight a representative oncology study demonstrating how Ribo-seq and RNA-seq integration clarifies disease mechanisms that remain hidden at the transcriptional level alone.
PM2.5 and Non-Small Cell Lung Cancer
Title:
PM2.5 Promotes NSCLC Carcinogenesis via Translational and Transcriptional Activation of DLAT-Driven Glycolytic Reprogramming
Journal: Journal of Experimental & Clinical Cancer Research
Publication date: July 2022
Impact factor: 12.66
Fine particulate matter (PM2.5) is a major contributor to global air pollution and haze exposure. Epidemiological evidence links PM2.5 to lung cancer development, including cases observed in non-smokers.
Despite this association, the molecular mechanisms underlying PM2.5-induced carcinogenesis remain poorly defined. Traditional cancer studies have largely focused on identifying differentially expressed mRNAs, assuming that transcriptional changes directly translate into protein-level effects.
However, transcriptomes contain substantial pools of untranslated RNA. As a result, correlations between mRNA abundance and protein expression are often weak. Protein output is instead more strongly shaped by translation efficiency.
Ribo-seq captures ribosome-protected RNA fragments, providing a direct snapshot of active protein synthesis. When combined with RNA-seq, this approach delivers a more complete and biologically meaningful gene expression landscape.
To dissect how PM2.5 drives NSCLC progression, the study employed a multi-layered experimental framework.
Ribo-seq and RNA-seq were first performed to identify PM2.5-responsive genes and metabolic pathways. Candidate targets were prioritised for downstream validation.
Gene and protein expression levels were quantified using quantitative RT-PCR, Western blotting, and immunohistochemistry. Functional consequences were assessed through gain- and loss-of-function assays, biochemical analyses, and glycolytic stress tests.
Clinical relevance was evaluated using human tissue microarrays and 18F-FDG PET/CT imaging data from NSCLC patients. Finally, molecular mechanisms were explored through polysome profiling, chromatin immunoprecipitation (ChIP), and dual-luciferase reporter assays.
PM2.5 exposure induced a selective translational shift in glycolysis-related genes rather than a global increase in metabolic activity. Among these targets, DLAT showed the most pronounced increase in translation efficiency.
Elevated DLAT expression enhanced glycolytic flux while suppressing acetyl-CoA production. This metabolic imbalance significantly intensified the malignant phenotype of NSCLC cells.
Figure 1. Mechanistic role of DLAT-mediated glycolytic reprogramming in PM2.5-induced NSCLC carcinogenesis.
Patient-based analyses supported the experimental findings. In NSCLC patients, high DLAT expression was positively correlated with larger tumour size and poorer clinical prognosis.
Notably, DLAT levels also showed a strong positive association with SUVmax values from 18F-FDG PET/CT imaging, indicating increased glucose uptake and metabolic activity in vivo.
PM2.5 amplified DLAT expression through coordinated transcriptional and translational regulation.
At the translational level, PM2.5 increased expression of eIF4E, a key translation initiation factor. This shift promoted DLAT mRNA recruitment to polysomes, directly enhancing DLAT protein synthesis.
At the transcriptional level, PM2.5 activated the transcription factor Sp1. Sp1 binding increased DLAT promoter activity, further elevating DLAT transcription.
Together, these mechanisms synergistically reinforced DLAT expression and glycolytic metabolism.
This study identifies DLAT-mediated glycolytic reprogramming as a central driver of PM2.5-induced NSCLC progression. By simultaneously enhancing DLAT transcription and translation, PM2.5 reshapes cancer cell metabolism toward aggressive growth and survival.
These findings suggest that targeting DLAT-driven glycolysis may represent a promising metabolic intervention strategy for non-small cell lung cancer.
Ultrasensitive Ribo-seq Reveals Translational Landscapes During Mammalian Oocyte-to-Embryo Transition
Title
Ultrasensitive Ribo-seq Reveals Translational Landscapes During Mammalian Oocyte-to-Embryo Transition and Pre-implantation Development
Journal: Molecular Therapy
Publication date: June 2022
Impact factor: 12.91
Translational regulation is a central layer of gene expression control during early development.
In mammalian oocytes and early embryos, global transcription remains largely silenced from oocyte maturation until zygotic genome activation (ZGA).
During this transcriptionally inactive window, protein synthesis relies almost entirely on post-transcriptional control.
As a result, translation regulation becomes the dominant mechanism governing cell fate decisions during the oocyte-to-embryo transition (OET).
Despite its importance, studying translation during OET has remained technically challenging.
Extremely limited sample input has historically restricted high-resolution analysis of ribosome dynamics in mammalian systems.
This study addresses these limitations by applying ultrasensitive Ribo-seq, enabling systematic investigation of translational control during oocyte maturation and early embryogenesis.
Mouse oocytes and embryos at multiple developmental stages were collected for integrated multi-omics profiling.
Experimental groups
Control group
Each group was analysed with biological replicates.
The authors performed parallel sequencing analyses, including:
This integrated approach enabled systematic evaluation of relationships between transcription, translation, and poly(A) tail regulation during OET.
Figure 2. Translational regulation during oocyte maturation and early embryonic development.
In oocytes and embryos prior to zygotic genome activation, translation showed low correlation with transcript abundance.
After genome activation, concordance between the translatome and transcriptome increased substantially.
This shift highlights a developmental transition from translation-dominated regulation to transcription-driven control.
Genes transcriptionally upregulated during OET exhibited suppressed translation in fully grown oocytes.
These transcripts were characterised by relatively short poly(A) tails.
Their 3' untranslated regions frequently contained multiple polyadenylation signal sites and proximal cytoplasmic polyadenylation elements (papCPEs).
Upon meiotic resumption, poly(A) tail length increased, triggering translational activation.
In contrast, genes transcriptionally downregulated during OET showed high translation efficiency in fully grown oocytes.
These transcripts possessed long poly(A) tails during the oocyte stage.
Following meiotic resumption, poly(A) tails were shortened, leading to translational repression.
This repression was mediated by deadenylation factors, which selectively controlled poly(A) tail dynamics.
Importantly, transcripts with shortened poly(A) tails in MII oocytes displayed reduced mRNA signals.
In contrast, RNAs retaining longer poly(A) tails remained relatively stable.
Together, these findings reveal a finely tuned regulatory system in which:
Ultrasensitive Ribo-seq, combined with RNA-seq and PAIso-seq, provides a powerful framework for dissecting gene regulation during early mammalian development.
Multifaceted Deregulation of Gene Expression and Protein Synthesis with Age
Title
Multifaceted Deregulation of Gene Expression and Protein Synthesis with Age
Journal: Proceedings of the National Academy of Sciences of the United States of America (PNAS)
Publication date: July 2020
Impact factor: 12.78
Protein synthesis represents one of the most energy-intensive and tightly regulated cellular processes.
Despite its central role in maintaining cellular homeostasis, how protein production changes with ageing—and how these changes impact cell function—remains incompletely understood.
To address this gap, the authors systematically characterised age-associated alterations in both the transcriptome and the translatome across the mouse lifespan.
By integrating transcriptional and translational profiling, the study aimed to uncover how gene expression control is progressively remodelled during ageing.
Mouse liver and kidney tissues were collected at multiple ages spanning early adulthood to advanced ageing.
Each age group included biological replicates to ensure robust statistical analysis.
The study combined two complementary sequencing approaches:
This integrated design enabled direct comparison between transcriptional regulation and protein synthesis efficiency as a function of age.
Figure 3. Age-associated changes in ribosome-protected fragment (RPF) profiles in mouse liver and kidney.
Analysis of RPF expression in liver and kidney tissues revealed that transcriptional changes strongly constrain age-related translational outputs.
Genes involved in inflammation, extracellular matrix organisation, and lipid metabolism showed pronounced age-dependent shifts at both RNA and translation levels.
At the translational level, the authors identified a specific class of transcripts that progressively declined with ageing.
These transcripts encoded proteins involved in ribosome biogenesis and protein synthesis.
Notably, many belonged to the 5' terminal oligopyrimidine (5'-TOP) motif-containing mRNAs, which are known to be regulated by the mTOR signalling pathway.
With increasing age, these transcripts exhibited reduced ribosome occupancy, indicating suppressed translation.
Ageing was also associated with changes in ribosome positioning along mRNAs.
Ribosome density decreased near start codons, while coverage increased around stop codons.
This redistribution suggests age-related impairment in translation initiation and elongation efficiency, further contributing to reduced protein synthesis capacity.
Together, these findings demonstrate that ageing is accompanied by multifaceted deregulation of protein synthesis, rather than a single uniform decline.
The study highlights how transcriptional reprogramming, mTOR-dependent translational control, and altered ribosome dynamics collectively reshape protein production with age.
These results underscore the complexity of translational regulation during ageing and position Ribo-seq as a critical tool for dissecting age-associated molecular decline.
The Cardiac Translational Landscape Reveals Micropeptides as New Regulators of Cardiomyocyte Hypertrophy
Journal: Molecular Therapy
Publication date: July 2021
Impact factor: 12.91
Cardiomyocyte hypertrophy is a major compensatory response of the heart to physiological or pathological stress.
This process is characterised by enhanced protein synthesis driven primarily at the level of mRNA translation.
Despite its importance, the molecular basis of translational activation during cardiac hypertrophy remains incompletely understood.
It is unclear whether increased protein output results from higher ribosome abundance or from accelerated translation rates.
Previous studies have also reported limited concordance between transcriptomic and proteomic data.
Changes in mRNA abundance alone often fail to explain protein-level remodelling observed during cardiac hypertrophy.
These discrepancies highlight the need for deep sequencing of actively translated transcripts.
By directly profiling ribosome-associated RNAs, translational analysis can provide a more accurate and comprehensive view of gene expression changes during cardiac remodelling.
In this study, cardiomyocyte hypertrophy was induced using phenylephrine (PE), a well-established hypertrophic stimulus.
Hypertrophic cardiomyocytes were subjected to integrated multi-omics profiling.
The authors performed:
Pathway enrichment analyses, including KEGG and Gene Ontology (GO), were used to identify biological processes underlying enhanced protein synthesis during hypertrophy.
In parallel, Ribo-seq data were mined to systematically identify translated small open reading frames (sORFs).
The authors detected more than 100 short open reading frames within selected long non-coding RNAs (lncRNAs).
Subsequent validation narrowed these candidates to three lncRNA-encoded micropeptides with potential roles in cardiomyocyte hypertrophy.
Two cytoplasm-localised micropeptides, RNO-sORF6 and RNO-sORF8, were found to promote cardiomyocyte hypertrophy.
RNO-sORF8 induced hypertrophic growth by modulating mitochondrial function.
RNO-sORF6 enhanced hypertrophy by upregulating MAPK signalling-related genes while suppressing TNF signalling and cell cycle-associated pathways.
In contrast, RNO-sORF7, which localised to mitochondria, exerted an inhibitory effect on cardiomyocyte hypertrophy.
This micropeptide suppressed hypertrophic growth by downregulating calcium signalling and MAPK pathway hub genes involved in cardiac remodelling.
By combining RNA-seq and Ribo-seq, this study systematically mapped the cardiac translational landscape under hypertrophic stress.
These findings expand the functional repertoire of the translatome and identify micropeptides as previously unrecognised regulators of cardiac remodelling.
This work highlights the power of Ribo-seq to uncover hidden layers of gene regulation that are invisible to transcriptome analysis alone.
By revealing micropeptide-driven control of cardiomyocyte hypertrophy, the study provides new mechanistic insights and potential therapeutic entry points for cardiovascular disease research.
One common application of integrated translatome and transcriptome analysis is to elucidate how key molecular factors regulate gene expression at the level of translation.
This research strategy typically starts with the identification of genes showing translationally driven expression differences. These differences often emerge when phenotypic changes cannot be adequately explained by transcriptional data alone.
Functional perturbation experiments are then used to assess the biological relevance of these candidates. Approaches such as translation interference, gene overexpression, or gene knockdown help determine whether the observed phenotypes are causally linked to translational regulation.
Following functional validation, integrated Ribo-seq and RNA-seq analysis is performed to distinguish transcriptional effects from translational control. By comparing transcript abundance with ribosome occupancy, researchers can pinpoint genes whose protein output is regulated primarily at the translation level.
Candidate genes identified through this process are further prioritised based on changes in translation efficiency and supported by molecular validation assays. This stepwise approach enables systematic discovery of key translational regulators and clarifies their role in shaping biological phenotypes.
Figure 4. Study design for identifying genes regulated at the translational level using Ribo-seq and RNA-seq.
Phenotypic differences guide the selection of candidate genes, which are further analysed through integrated translatome and transcriptome profiling and validated by functional assays.
A frequent challenge in gene expression studies is the inconsistency observed between transcriptomic and proteomic analyses. In many cases, key genes show comparable mRNA levels across conditions, while protein abundance differs substantially, complicating biological interpretation.
This research strategy typically begins with prior observations showing discordance between transcriptome data and protein-level measurements for genes of interest. To determine whether these differences arise from post-translational regulation, protein degradation pathways are first evaluated.
Pharmacological treatments such as proteasome inhibitors (e.g. MG132) or translation elongation inhibitors (e.g. cycloheximide, CHX) are commonly applied to assess whether altered protein abundance is driven by differential protein stability or degradation. If protein degradation is identified as the primary regulatory mechanism, downstream analysis focuses on post-translational control.
When protein degradation is excluded as the dominant factor, integrated Ribo-seq and RNA-seq analysis is performed to investigate translational regulation. This approach enables direct assessment of ribosome occupancy and translation efficiency, distinguishing translational control from transcriptional effects.
Through this analysis, researchers can quantify translation efficiency, translation elongation dynamics, and associated regulatory factors for key genes. Candidate genes showing translationally driven regulation are then prioritised for further investigation.
Finally, functional assays are conducted to validate how specific molecular regulators influence translation levels and, in turn, modulate disease-related or biological phenotypes. This step establishes a mechanistic link between translational regulation and functional outcomes, providing a clear explanation for transcript–protein discordance.
Figure 5. Workflow for distinguishing translational regulation from protein degradation in gene expression studies.
The diagram outlines a stepwise approach to determine whether transcript–protein discordance arises from post-translational degradation or translational control, followed by functional validation of key regulatory genes.
Another important application of integrated translatome and transcriptome analysis is the systematic identification of key genes and signalling pathways associated with disease states.
This approach typically involves comparative analysis between normal and diseased tissues or cells. By profiling both conditions in parallel, researchers can capture gene expression changes that occur specifically during disease progression.
Integrated Ribo-seq and RNA-seq analysis is then performed on the same samples. RNA-seq reveals transcriptional alterations, while Ribo-seq provides direct insight into ribosome occupancy and translational activity. Together, these datasets enable identification of genes whose functional output is shaped by translational regulation rather than transcription alone.
Based on the integrated results, candidate genes and pathways are prioritised according to consistent changes in translation efficiency and biological relevance. Functional enrichment analyses further highlight signalling networks and biological processes that may drive disease-related phenotypes.
Finally, selected genes and pathways are subjected to validation using cell-based or animal models. These functional assays confirm the contribution of translationally regulated targets to disease mechanisms, strengthening the causal link between molecular regulation and phenotypic outcomes.
Through this strategy, integrated translatome and transcriptome profiling provides a powerful framework for uncovering disease-relevant genes and pathways that may remain hidden when analysing transcriptional data alone.
Figure 6. Workflow for discovering disease-relevant genes and pathways through integrated Ribo-seq and RNA-seq analysis.
Comparative profiling of normal and diseased samples reveals translationally regulated genes and pathways that contribute to pathological phenotypes.
Integrated translatome analysis also provides a powerful strategy to reassess the coding potential of transcripts previously annotated as non-coding RNAs. Increasing evidence suggests that a subset of these RNAs may encode functional micropeptides that are overlooked by conventional transcriptome analysis.
This research approach begins by evaluating whether certain transcripts classified as non-coding may in fact associate with ribosomes. Combined RNC-seq and Ribo-seq analysis enables detection of ribosome-bound ncRNAs, highlighting candidates with potential translational activity.
Candidate ncRNAs identified through ribosome association are then examined for the presence of actively translated small open reading frames (sORFs). These analyses allow researchers to distinguish true translational events from background ribosome binding.
To confirm protein-coding capacity, candidate sORFs are validated at the protein level using experimental approaches such as Western blotting or mass spectrometry. This step provides direct evidence that the ncRNA-derived transcript produces a detectable peptide.
Once protein expression is confirmed, functional assays are performed to determine whether the newly identified proteins possess biological activity. Phenotypic analyses at the cellular or organismal level further establish the functional relevance of these micropeptides.
Through this strategy, integrated translatome profiling enables systematic discovery and validation of previously unrecognised protein-coding genes, expanding the functional landscape of the genome beyond conventional annotations.
Figure 7. Strategy for identifying protein-coding potential within non-coding RNAs using translatome profiling.
RNC-seq and Ribo-seq are used to detect ribosome-associated ncRNAs and identify translated sORFs. Protein-level validation and functional assays confirm the biological activity of newly discovered micropeptides.
Most translatome studies do not rely on Ribo-seq alone.
Instead, researchers routinely perform Ribo-seq and RNA-seq on the same samples, followed by integrated analysis.
Through combined translatome–transcriptome profiling, researchers can:
Importantly, this strategy also enables:
Together, integrated Ribo-seq and RNA-seq analysis provides a robust framework for studying gene regulation beyond transcription and for uncovering novel layers of biological complexity.
Integrated translatome and transcriptome analysis is particularly informative in research scenarios where transcriptional data alone fails to explain biological outcomes.
This approach is well suited for studies where:
In these contexts, integrating Ribo-seq with RNA-seq can provide a more complete and mechanistically grounded view of gene regulation beyond transcription.
References: