Updated project metadata. T-cells are a critical component of the adaptive immune system and play a key role in immunological surveillance. Upon engagement of T-cell receptor (TCR), CD4+ and CD8+ T-cells acquire effector functions through a complex interplay between mRNA and proteins yet to be fully understood. In this study we explored the temporal transcriptomic and proteomic changes mediated by TCR engagement in both CD4+ and CD8+ T-cells. T-cells isolated from peripheral blood mononuclear cells of three healthy volunteers over 90% purity as assessed by fluorescence-labeled flow cytometry (FACS) and monoclonal antibodies were in vitro activated using anti-CD3/CD28 Dynabeads. Samples obtained before the activation, and 6h, 12h, 24h, 3 days (d), and 7d following activation were analyzed using label-free data-dependent acquisition mass spectrometry-based proteomics (DDA-proteomics), to identify the temporal dynamics in CD4+ and CD8+ T-cell proteomes during activation. A parallel analysis was performed to explore the transcriptomic dynamics during T-cell activation. Our data revealed a time-dependent dissociation between the T-cell transcriptome and proteome: the onset of activation was driven by rapid changes of the mRNA content with sluggish increase in protein synthesis, ultimately leading to rewired transcriptome and proteome. We surprisingly found that CD4+ and CD8+ T-cells became transcriptionally more divergent while their proteome became more similar over the time course of activation. Several changes in the content of mRNAs and proteins associated with metabolic pathways were detected through KEGG pathway analysis, revealing a transient disconnection between the aerobic glycolysis and glutaminolysis pathways in activated T-cells. This dataset provides a comprehensive framework for understanding the main temporal changes that regulate metabolic pathways governing the acquisition of effector functions by CD4+ and CD8+ T-cells.