Abstract Background The Pacific white shrimp (Litopenaeus vannamei) is the most widely farmed shrimp species globally, yet the epigenetic regulation underlying its embryonic development remains largely unexplored. Histone modifications are known to orchestrate gene expression during early development in model organisms, but their role in crustaceans is poorly understood. Results In this study, we present the first comprehensive histone modification landscape during L. vannamei embryogenesis using CUT&Tag (Cleavage Under Targets and Tagmentation). We profiled high-resolution landscapes of four histone marks (H3K4me1, H3K4me3, H3K27ac, H3K27me3) across seven developmental stages from blastula to nauplius, revealing dynamic chromatin state transitions associated with developmental progression. Integration with transcriptomic data uncovered a strong temporal correlation between chromatin states and gene expression, particularly during zygotic genome activation (ZGA). Furthermore, our analysis uncovered key developmental genes associated with critical biological processes such as molting, body segmentation, and neurogenesis, providing novel insights into the epigenetic regulation of these events. Functional annotation of cis-regulatory elements based on histone marks identified candidate enhancers and regulatory loci linked to these key genes. Conclusions Our study provides the first epigenomic framework of shrimp embryogenesis, uncovering chromatin-based regulatory mechanisms during early development. The identification of stage-specific enhancers and active chromatin regions offers valuable resources for functional genomics in crustaceans and sheds light on conserved and divergent aspects of ZGA regulation beyond model systems. Supplementary Information The online version contains supplementary material available at 10.1186/s13072-025-00615-4. Keywords: Litopenaeus vannamei, CUT&Tag, Histone modifications, Epigenetic regulation, Zygotic genome activation Background The Pacific white shrimp (Litopenaeus vannamei) is one of the most economically significant farmed shrimp species globally, owing to its rapid growth, high yield, and superior meat quality. According to the 2022 statistics from the Food and Agriculture Organization (FAO), the aquaculture production of L. vannamei accounted for 51.7% of the world’s total crustacean production, reaching 5.8 million tons, ranking it first worldwide [[36]1]. As a member of the Decapoda within Crustacea, L. vannamei undergoes a unique early developmental pattern, which includes several distinct stages: embryo, nauplius, zoea, mysis, and postlarvae. During the embryonic phase, development progresses from a zygote to a multicellular structure, and through key stages such as blastula, gastrula, limb bud embryo, and larva in membrane. Following hatching, the shrimp progresses through successive nauplius, zoea, mysis, and post larval stages, marked by distinct morphological changes [[37]2, [38]3]. These early developmental stages are marked by significant morphological and physiological changes, which play a crucial role in the overall life cycle. The process of metamorphosis represents a critical transition in L. vannamei’s development and serves as an example of life cycle modularity [[39]3, [40]4]. However, these pronounced physiological changes during early development can lead to high variability and difficulty in larval rearing, posing a significant challenge in shrimp aquaculture [[41]3, [42]5]. The early embryonic development of animals is primarily regulated by maternal gene products. As development progresses, control gradually shifts to the zygotic genome during the maternal-to-zygotic transition (MZT), a process characterized by precise and coordinated changes in gene expression. Studies have shown that the timing and intensity of ZGA vary across species, reflecting the diversity of regulatory mechanisms involved [[43]6]. Despite its critical role in development, ZGA in L. vannamei remains unexplored, leaving a significant gap in understanding how gene expression is regulated during this pivotal phase. Notably, L. vannamei exhibits unique developmental features, such as extensive physiological changes in tissue formation, dietary transitions, muscle development, and exoskeleton remodeling. These processes, while essential for its life cycle, are still not fully understood. Current studies on L. vannamei have primarily focused on its morphological, biochemical, and transcriptomic characteristics [[44]3], with limited attention to the epigenetic mechanisms, particularly histone modifications, that underlie gene expression regulation during early embryogenesis. Epigenetic regulation, including DNA methylation, histone modifications, and chromatin remodeling, modulates gene expression without altering the DNA sequence [[45]7]. Histones, as fundamental components of chromatin, undergo various post-translational modifications (PTM), such as methylation, acetylation, and phosphorylation, which impact chromatin dynamics and facilitate the binding of transcriptional regulators [[46]8, [47]9]. Post-translational modifications of histones have been widely demonstrated to play critical roles in regulating gene expression during animal embryonic development [[48]8, [49]10]. Moreover, specific types of histone modifications can serve as predictors of gene expression activity [[50]11, [51]12]. Among these, four well-characterized histone modifications include H3K4me1 (monomethylation of histone 3 lysine 4), H3K4me3 (trimethylation of histone 3 lysine 4), H3K27me3 (trimethylation of histone 3 lysine 27), and H3K27ac (acetylation of histone 3 lysine 27). H3K4me1 is associated with active enhancers, H3K4me3 marks promoters of actively transcribed genes, H3K27me3 is linked to transcriptional repression, and H3K27ac indicates regions of open chromatin. These histone modifications play pivotal roles in regulating gene expression and chromatin structure during developmental processes [[52]11, [53]12]. However, their roles in the early embryonic development of L. vannamei remain largely unexplored. Cleavage Under Targets & Tagmentation (CUT&Tag) is an advanced genome-wide chromatin profiling technique that has emerged as a powerful tool for studying histone modifications [[54]13]. Compared to traditional chromatin immunoprecipitation followed by sequencing (ChIP-seq), CUT&Tag offers several advantages, including a higher signal-to-noise ratio, greater sensitivity, reduced experimental time, and lower cell requirements [[55]13]. This method has proven effective for analyzing chromatin modifications and transcription factor binding sites [[56]14]. This innovative approach provides a powerful tool to investigate the epigenetic landscape regulating early embryonic development in L. vannamei. In this study, we employed CUT&Tag and RNA-seq technologies to generate high-resolution maps of the histone modifications H3K4me1, H3K4me3, H3K27me3, and H3K27ac during critical stages of early embryonic development in L. vannamei. Our primary goal is to investigate the role of these histone modifications in the MZT and to capture the dynamic epigenetic changes that regulate gene expression during this period. This study advances our understanding of epigenetic regulation during L. vannamei embryogenesis, offering insights to improve shrimp aquaculture. Furthermore, by annotating the genome-wide cis-regulatory elements of L. vannamei using multiple histone modifications, this research lays the groundwork for further functional genomics studies in this economically important species. Methods Experimental animal The samples of L. vannamei used in this study were collected on-site at Hairen Aquatic Seed Industry Technology Co., Ltd (Hebei, China). High-quality male and female broodstock were selected by experienced technicians for artificial mating. The reproductive behavior of the shrimp was closely monitored, and once spawning was observed, fertilized eggs were immediately collected. The eggs were washed with clean seawater and incubated under strictly controlled conditions (temperature: 30 ± 0.5 °C; salinity: 33‰). Embryonic development was monitored in real time under a microscope. At each target developmental stage, embryos were promptly collected using a nylon mesh to ensure stage-specific sampling. Samples for CUT&Tag were processed immediately after collection. For RNA-seq, embryos at the same developmental stages were washed three times with sterile seawater, flash-frozen in liquid nitrogen, and transported on dry ice to our laboratory, where they were stored at − 80 °C until further processing. This study included seven developmental stages: blastula (blas), gastrula (gast), limb bud embryo (lbe), larva in membrane (lim), nauplius I (N1), nauplius III (N3) and nauplius VI (N6) stages. CUT&Tag experiment and data analysis The CUT&Tag libraries were prepared using the Illumina Hyperactive Universal CUT&Tag Assay Kit (Vazyme, China, #TD903), with nuclei extracted from the embryos following a previously described protocol [[57]14]. Approximately 200 embryos were collected at each developmental stage for each biological replicate using a nylon mesh. The freshly collected embryos were then washed 2–3 times with Wash Buffer to remove residual seawater and impurities. To isolate nuclei, 300 µL of pre-chilled NE Buffer was added to the embryos, which were then homogenized on ice using sterile surgical scissors. An additional 500 µL of pre-chilled NE Buffer was added, and the homogenate was pipetted thoroughly to generate a single-cell suspension. The suspension was incubated on ice for 5 min and filtered through a 40 μm strainer to remove debris. The filtrate was centrifuged at 1300 × g for 4 min, and the resulting pellet was washed 2–3 times with 500 µL of Wash Buffer. Finally, the nuclei were resuspended in 100 µL of Wash Buffer, mixed gently, and prepared for downstream applications. All CUT&Tag procedures were performed in accordance with the manufacturer’s instructions and the published protocol [[58]14]. Two biological replicates were included for each developmental stage, and approximately 50,000 nuclei were used per sample. Antibodies used in this study are listed in Table [59]S1. The CUT&Tag libraries were sequenced on the Illumina NovaSeq 6000 platform (Novogene, Tianjin, China). Raw sequencing data were subjected to quality control using the NGS QC Toolkit v2.3 to generate clean reads [[60]15]. The clean reads were aligned to the L. vannamei reference genome (NCBI accession: ASM378908v1) using the BWA-MEM v0.7.17 algorithm [[61]16], and mapping statistics are summarized in Table [62]S2. Using awk, reads with abnormal pairing relationships (e.g., no distance or distances greater than 1000 bp) were removed, retaining only fragments with lengths between 100 and 1000 bp. These filtered fragments were subsequently used for peak calling with MACS2 v2.2.7.1 [[63]17], applying the following parameters: macs2 callpeak -t A.bam -c IgG.bam -f BAMPE -g 1.66e9 --keep-dup auto -q 0.05. Annotation and visualization of the peaks were conducted using R packages GenomicFeatures [[64]18], ChIPseeker [[65]19], and ggplot2 [[66]20]. Additional analyses included fragment size distribution, reproducibility assessment of replicate samples, signal heatmaps of H3K4me3 at transcription start sites (TSS), peak signal enrichment heatmaps, and peak distribution across genomic features. The analyses followed R scripts from a previously published pipeline [[67]21]. Chromatin state analysis Chromatin state analysis was performed using the ChromHMM v1.26 model, which identifies distinct chromatin states and annotates their genome-wide distribution based on combinatorial patterns of histone modifications at each locus [[68]22]. The model was trained using dynamic profiles of four key histone modifications (H3K4me1, H3K4me3, H3K27me3, H3K27ac) obtained from the above mentioned seven embryonic developmental stages. To determine the optimal number of chromatin states, we performed parameter tuning and model evaluation. The results indicated that models with more than 10 states exhibited substantial functional redundancy among states, while models with fewer than 10 states failed to effectively distinguish certain biologically meaningful features. Therefore, the final model employed the LearnModel function to define 10 chromatin states, balancing the need to reduce redundancy while preserving the resolution required to capture critical biological signatures. The genome-wide distribution patterns of chromatin states across distinct embryonic developmental stages are presented in Supplementary File [69]1. The chromatin state maps were visualized using Integrative Genomics Viewer (IGV) v2.15.2 to facilitate interpretation [[70]23]. RNA-seq experiment and data analysis RNA-seq samples were collected in parallel with those used for the CUT&Tag experiment, from the same batch of embryos, with three biological replicates for each developmental stage. Total RNA was extracted using the RNAprep Pure Tissue Kit DP431 (TIANGEN, Beijing, China), followed by DNase I (Takara Bio, Inc.) treatment to remove DNA contamination. RNA quality and purity were assessed using a Nanovue Plus spectrophotometer (GE Healthcare, New Jersey, USA) and agarose gel electrophoresis. Transcriptome libraries were prepared using the NEBNext^® Ultra™ RNA library prep kit for Illumina^® (NEB, Ipswich, MA, USA) and sequenced on the Illumina NovaSeq PE150 platform (Illumina, San Diego, CA, USA), generating approximately 6 GB of raw data per sample. The raw sequencing data underwent quality control using the NGS QC Toolkit v2.3 [[71]15] to produce clean reads (quality control results are summarized in Table [72]S3). The clean reads were aligned to the L. vannamei reference genome using STAR v2.7.11b [[73]24]. Gene quantification was performed using featureCounts [[74]25], and expression levels were normalized to transcripts per million mapped reads (TPM). For data exploration, principal component analysis (PCA) was conducted after z-score normalization. Differential gene expression analysis was carried out using DESeq2 [[75]26] with thresholds of|fold change| >2 and p < 0.05. KEGG Pathway enrichment analysis of differentially expressed genes (DEGs) was conducted using the R package clusterProfiler. The expression levels of key genes during embryonic development were quantified in TPM and visualized using GraphPad Prism v8.0.2. Temporal analysis of gene expression dynamics Gene expression dynamics across developmental stages were analyzed using Short Time-series Expression Miner (STEM) v1.3.13. The average TPM values from three biological replicates per stage were log2-normalized, and nine model profiles were pre-defined. Significant trends (p < 0.05) were identified, with a minimum fold-change threshold of 2 for gene selection. Genes from each profile were further analyzed for functional enrichment using KEGG pathway analysis via the clusterProfiler R package. Integrated analysis of cut & tag and RNA-seq data To minimize the impact of low-confidence and weak signals that might confound downstream functional analyses and to enhance the robustness of our conclusions, we employed a stringent peak filtering strategy. Specifically, Bedtools v2.31.1 [[76]27] was used to identify intersecting peaks between the two biological replicates, and only high-confidence peaks that were consistently detected across both samples were retained. These validated peaks were then used for subsequent analyses, including histone modification peak quantification, gene-level annotation, and integration with transcriptomic datasets. Histone modifications were analyzed based on their well-characterized regulatory roles: H3K4me3 is enriched at the promoters of actively transcribed genes, H3K27me3 is associated with transcriptional repression mediated by the Polycomb Repressive Complex 2 (PRC2), H3K4me1 serves as a marker of enhancers, and H3K27ac identifies accessible chromatin regions [[77]11, [78]12]. Based on these definitions, genomic regions concurrently marked by H3K4me3 and H3K27ac, but devoid of H3K27me3, were designated as “active open sites”. Genes associated with these regions and exhibiting an expression level of TPM > 0.5 were defined as actively expressed genes. KEGG pathway enrichment analysis was performed on this gene set to identify significantly overrepresented pathways (p < 0.05). To assess dynamic expression trends, we employed STEM [[79]28] on log₂-normalized expression data. Trends were considered significant if p < 0.05, with a minimum fold change ≥ 2 used as a selection criterion. In parallel, genomic regions showing an overlap of at least 1 bp between H3K4me1 and H3K27ac peaks, while lacking H3K27me3, were defined as active enhancers at specific developmental stages. Visualization of selected gene loci was performed using IGV v2.15.2 [[80]23], and statistical analyses of gene expression were conducted using GraphPad Prism v8.0.2. Gene expression heatmaps were standardized using z-score normalization to facilitate cross-sample comparison. Results High resolution mapping of histone modifications in L. vannamei embryos using CUT&Tag To validate the application of CUT&Tag in L. vannamei embryos, we first assessed its efficiency. Alignment rates of sequencing reads to the L. vannamei reference genome consistently exceeded 90% across all samples, confirming high-quality data generation (Table [81]S2). Taking the N3 stage as an example, the fragment size distribution displayed the typical nucleosomal ladder pattern, characteristic of successful CUT&Tag experiments in the histone modification samples. In contrast, control samples exhibited a random size distribution, further validating the specificity of the technique (Fig. [82]1A) [[83]29]. The reproducibility of the experiment was assessed by correlation analysis, which showed high consistency among biological replicates (Fig. [84]1B & S1A). To further validate the specificity of the CUT&Tag results and the accuracy of peak calling, we conducted signal enrichment analysis at peak regions using heatmaps. The results showed significant signal enrichment at peak centers for histone modification groups, whereas the control group (Immunoglobulin G antibody, IgG antibody) exhibited no enrichment due to the lack of specific binding at these regions (Fig. [85]1C). In addition, H3K4me3 showed strong enrichment around transcription start sites (TSS) (Fig. [86]1C), consistent with its known localization pattern [[87]30], confirming the specificity and reliability of the data. Similar patterns were observed for the other six developmental stages (Figure [88]S1A). Genome-wide analysis of histone mark distribution at the seven developmental stages revealed that H3K4me1, H3K4me3, H3K27ac, and H3K27me3 were predominantly located in promoter regions and distal intergenic regions, rather than within gene bodies, consistent with their regulatory roles in gene expression [[89]11, [90]12]. Specifically, approximately 20%-30% of H3K4me1, 50%-70% of H3K4me3, 10% of H3K27me3, and 30%-40% of H3K27ac were located in promoter regions. In distal intergenic regions, around 50%-60% of H3K4me1, 30% of H3K4me3, 60%-70% of H3K27me3, and 40%-50% of H3K27ac were distributed (Fig. [91]1D and E, S1B; Table [92]S4). These results demonstrate that CUT&Tag provides robust detection of histone modifications in the embryonic L. vannamei genome, offering reliable insights into histone mark distribution and their potential regulatory roles during early development. Fig. 1. [93]Fig. 1 [94]Open in a new tab Quality assessment of CUT&Tag at the nauplius III stage. (A) Fragment size distribution. Plots showing the proportion (left) and raw number (right) of DNA fragments by size. Histone modification samples show strong enrichment of mononucleosome-sized fragments, whereas control samples exhibit a random size distribution. (B) Replicate reproducibility. Correlation analysis of genome-wide read distribution using 500 bp bins demonstrates high reproducibility between biological replicates of histone modification samples. (C) Signal enrichment at specific loci. The left two panels show the enrichment of CUT&Tag signal for H3K4me3 and IgG control at transcription start sites (TSS) of annotated genes. The right two panels depict signal enrichment within ± 1 kb of the peak summit center for H3K27me3 and IgG. Heatmaps (bottom) and line plots (top) confirm significant enrichment for histone marks compared to controls. (D) Peak distribution across genomic features. (E) Enrichment of peaks relative to the TSS Chromatin state annotation based on histone modification marks during early embryonic development in L. vannamei The distribution patterns of histone modifications and their influence on chromatin accessibility enable genome-wide chromatin state annotation [[95]22]. Such annotations could serve as a valuable resource for identifying regulatory features associated with key developmental processes in the L. vannamei genome. To provide the L. vannamei research community with a comprehensive, dynamic, and accessible genomic annotation, we utilized the dynamic profiles of four informative histone modifications obtained across multiple early developmental stages in this study. Using ChromHMM, we performed a detailed analysis of chromatin states in the L. vannamei genome during early embryonic development. As this is the first chromatin state analysis conducted for this species, our study provides a novel perspective. Ten distinct chromatin states were identified and named following ChromHMM conventions [[96]22]. The states are as follows: Bivalent enhancer 1 (EnhBiv1), Weak transcription (TxWk), Active TSS (TssA), Flanking TSS downstream (TssFlnkD), Weak flanking bivalent TSS/Enhancer (BivFlnkWk), Bivalent enhancer 2 (EnhBiv2), Quiescent/low (Quies), Flanking bivalent TSS/Enhancer (BivFlnk), Weak repressed polycomb (ReprPCWk), and Repressed polycomb (ReprPC). These chromatin states encompass a range of functional patterns, including active transcription, enhancers, bivalent regions, repressive marks, and quiescent regions (Figs. [97]2A, B). Fig. 2. [98]Fig. 2 [99]Open in a new tab ChromHMM-based chromatin state analysis in L. vannamei. (A) Heatmap (left) showing emission parameters for the Roadmap Epigenomics 10-state expanded model, based on H3K4me1, H3K4me3, H3K27me3, and H3K27ac signals. Darker colors indicate higher signal enrichment, while lighter colors represent weaker signals. The adjacent heatmap (right) displays fold enrichment for each chromatin state in 200-bp bins within 2 kb of transcription start sites (TSSs). State descriptions and abbreviations are provided to the right of the heatmaps. (B) Example browser view of ChromHMM-defined chromatin states for the nauplius III embryo genome. The top track represents the annotated reference genome, followed by an integrated chromatin state track and individual tracks for each chromatin state. (C) ChromHMM genome annotations for seven embryonic developmental stages, illustrated using the [100]XM_027355294.1 (e75) gene region as an example. Each row corresponds to a specific stage, with consistent color coding for the chromatin states in A, B and C. (D) Bar chart displaying the temporal expression levels of the e75 gene during L. vannamei embryogenesis, quantified in TPM. (E) IGV tracks showing e75 expression and histone modification patterns from the gastrula to nauplius VI stages. The signal dynamics of active chromatin marks (H3K4me3 and H3K27ac) correlate well with the expression level of e75 To assess the validity of chromatin state annotations, we manually examined the relationship between gene expression levels and chromatin state categories. Overall, gene expression levels corresponded well with the annotated chromatin states. As a representative example, we investigated the e75 gene ([101]XM_027355294.1), known to be associated with molting. From the gastrula to N6 stages, the promoter region of e75 was annotated as an Active TSS (TssA) chromatin state, characterized by strong enrichment of H3K4me1, H3K4me3, and H3K27ac. Visualization using IGV and gene expression analysis revealed that e75 expression begins during the gastrula stage, consistent with its chromatin state and histone modification patterns (Fig. [102]2C-E). These findings support the reliability and biological relevance of the chromatin state predictions. The chromatin state analysis results, including the stage-specific tracks of each individual chromatin state as well as composite tracks integrating all identified states, as illustrated in Fig. [103]2B and C, are detailed in Supplementary file [104]1. This is the first chromatin state annotation study for L. vannamei and it offers a foundational resource for future studies investigating the molecular mechanisms of L. vannamei embryonic development. Dynamic changes in histone modifications and transcriptome during ZGA The precise and orderly activation of the zygotic genome is crucial for normal embryonic development, and histone modifications have been shown to play a key role in this process [[105]6]. To uncover the dynamic changes in histone modification and transcriptome during ZGA in L. vannamei, we analyzed the four histone modifications (H3K4me1, H3K4me3, H3K27me3, and H3K27ac) alongside transcriptomic profiles across seven key developmental stages. Principal component analysis (PCA) demonstrated strong similarity between the transcriptomes of the replicate samples across developmental stages, confirming the reliability of the transcriptomic data (Figure [106]S2). The number of significantly enriched histone modification peaks (Fig. [107]3A) and the heatmap of signals surrounding the peaks (Fig. [108]3C) revealed that H3K4me1 and H3K4me3 were already highly enriched starting from the blastula stage, while H3K27me3 and H3K27ac exhibited significant enrichment beginning at the gastrula stage. Notably, H3K4me1, H3K27me3, and H3K27ac reached their highest levels of enrichment at the N1 stage, whereas H3K4me3 peaked during the lim stage. The number of genes corresponding to these histone modification peaks also followed the same trend (Fig. [109]3B), indicating that the signal changes of these histone modifications were relatively evenly distributed across these different developmental stages rather than being concentrated in some specific regions. Fig. 3. [110]Fig. 3 [111]Open in a new tab Dynamics changes of histone modifications and transcriptome during ZGA. (A) The number of histone mark-associated peaks across seven developmental stages. (B) The number of histone mark-associated genes. (C) Heatmaps of signal enrichment. Enrichment patterns of H3K4me1, H3K4me3, H3K27me3, and H3K27ac, with signals centered on peak summits and extended to ± 1,000 bp regions across the seven developmental stages. (D) The proportion of expressed genes (with an average TPM value > 0.5) relative to the total gene count in the L. vannamei genome at each developmental stage. (E) A schematic model depicting the temporal dynamics of histone marks (red curves) and transcription levels (blue curves) during early embryonic development. Developmental stage-specific embryonic schematics in this figure were adapted from a previously published study [[112]3] During the MZT, maternal mRNA undergoes degradation before the activation of zygotic gene expression. Consequently, the number of expressed genes in the early embryonic transcriptome decreases initially, followed by a gradual increase upon ZGA [[113]6]. In L. vannamei, our study showed that expressed genes during the blastula stage accounted for 23.27% of the total genes, with a steady increase observed across developmental stages until expression plateaued during the lim stage (Fig. [114]3D). This suggests that ZGA in L. vannamei likely initiates before the blastula stage. This interpretation is supported by the enrichment of H3K4me1 and H3K4me3 signals, along with detectable levels of H3K27me3 and H3K27ac, as early as the blastula stage. This is consistent with observations in other species, where ZGA is typically accompanied by the re-establishment of histone modifications [[115]6]. Overall, our data demonstrate that histone modifications and transcriptomic changes are tightly coordinated during early embryonic development in L. vannamei, displaying a strong temporal dependency and correlation (Fig. [116]3E). Differential gene expression analysis across developmental stages of L. vannamei embryos To investigate gene expression dynamics across developmental stages of L. vannamei embryos, differential gene expression (DEG) analysis was performed between adjacent stages, followed by KEGG pathway enrichment analysis. The number of upregulated and downregulated DEGs at each stage is shown in Fig. [117]4A. Fig. 4. [118]Fig. 4 [119]Open in a new tab Analysis of differentially expressed genes (DEGs) by RNA-seq. (A) Bar chart of DEGs between neighboring developmental stages. (B) Bubble chart of KEGG pathway enrichment analysis of DEGs across developmental stages. Bubble size represents the number of DEGs associated with each pathway, and bubble color intensity reflects the significance level of enrichment Notably, a comparison between the gastrula and blastula stages revealed a predominance of upregulated DEGs, likely reflecting the large-scale activation of the zygotic genome during this phase. As development progressed, both upregulated and downregulated genes increased in number, correlating with the heightened levels of repressive H3K27me3 and active H3K27ac histone modifications observed at later stages. This pattern aligns with the increasing complexity of embryonic tissues and organs, necessitating precise gene expression regulation for proper cell differentiation [[120]3]. KEGG pathway enrichment analysis identified stage-specific biological processes and signaling pathways. In the gastrula stage, DEGs were significantly enriched in pathways related to cell status and signal transduction, including tight junctions, apoptosis, and the cGMP-PKG pathway. During the lbe stage, DEGs were enriched in pathways involved in transcription, translation, and signal transduction, such as ribosome biogenesis, mRNA surveillance, and the Hippo signaling pathway. For the lim stage, DEGs were associated with pathways related to the nervous system, immune system, digestive system, endocrine system, and sensory systems, including Wnt signaling, Hippo signaling, neuroactive ligand-receptor interaction, platelet activation, fat digestion and absorption, thyroid hormone signaling, and phototransduction. At the nauplius stages (N1, N3, N6), DEGs were enriched in pathways related to metabolism (amino acids, lipids, carbohydrate, glycan biosynthesis) and hormone biosynthesis, as well as pathways linked to the digestive and nervous systems (Fig. [121]4B). These results demonstrate that DEGs at different developmental stages are involved in distinct biological processes and signaling pathways, reflecting the dynamic changes in cell status and function during embryonic development in L. vannamei. Functional characterization of actively expressing genes at each developmental stage of L. vannamei embryo While transcriptomics reflects cumulative transcript levels, histone modifications provide real-time insights into the chromatin’s accessibility and regulatory status. These modifications help identify cis-regulatory elements, such as promoters and enhancers, and clarify their roles in gene expression and regulation [[122]11, [123]12]. During embryonic stages that inherit large amounts of maternal mRNA, histone modification data help mitigate the confounding effects of maternal mRNA fluctuations, enabling the identification of key zygotically expressed genes essential for developmental progression [[124]31]. To define such regulatory elements, we identified “active open sites” as genomic regions marked by H3K4me3 and H3K27ac, but lacking H3K27me3. Genes associated with these regions and exhibiting nonzero expression were classified as actively expressing genes, which are likely to play crucial regulatory roles during specific developmental stages. To investigate this, we integrated CUT&Tag and RNA-seq data to identify actively expressing genes at each developmental stage of L. vannamei embryogenesis. The results indicated that most genes associated with active open sites exhibited expression at certain stages (Fig. [125]5A), suggesting that chromatin modification states can reliably predict gene expression. The absence of expression in some cases could be due to the fact that chromatin opening precedes transcriptional activation [[126]32]. To validate the functional significance of these genes, we performed a KEGG pathway enrichment analysis, revealing stage-specific functional patterns. The results indicated that the majority of significantly enriched pathways were associated with signal transduction, and the key pathways at different developmental stages exhibited a high degree of consistency with their biological characteristics. During the gastrula and lbe stages, the significant enrichment of pathways related to cell growth and differentiation (e.g., the Hippo signaling pathway) closely aligned with the rapid proliferation of embryonic cells and the events of germ layer differentiation [[127]3, [128]33]. At the lim stage, the significant enrichment of immune system pathways (e.g., the T cell receptor signaling pathway) coincided with the critical period when larvae lose the protection of the egg membrane, reflecting the biological need for the rapid establishment of immune defense mechanisms during this stage [[129]34]. In the nauplius stages (N1-N6), the significant enrichment of transport and catabolism pathways (e.g., fatty acid metabolism) was closely related to the energy remodeling required for enhanced locomotion and dietary shifts in the larvae (Fig. [130]5B) [[131]3, [132]33]. Fig. 5. [133]Fig. 5 [134]Open in a new tab Functional enrichment of actively expressed genes at each developmental stage. (A) Venn diagram illustrating the overlap between genes identified by CUT&Tag and RNA-seq. The pink circle represents genes expressed at a given stage (TPM > 0.5), while the blue circle represents genes associated with active open sites (marked by H3K4me3 at promoter regions and overlapping with H3K27ac peaks, but not H3K27me3). The intersection of these two sets highlights genes that are actively expressed at that stage. (B) KEGG pathway enrichment analysis of the actively expressed genes, with significantly enriched pathways (p < 0.05) visualized in a bubble plot Further, to understand the temporal expression patterns of these genes and their relationship to developmental stages, we conducted gene expression temporal analysis. This categorized the genes into nine distinct profiles. These profiles exhibit expression characteristics correlated with key developmental events and significantly enriched biological pathways (Fig. [135]6). Profile 1 shows the strongest expression during the blastula stage, followed by a gradual downregulation. It is significantly enriched in pathways such as the Hippo signaling pathway and apoptosis-related pathways, suggesting its role in regulating early embryonic proliferation and morphogenesis. During the blastula stage, cells maintain proliferative homeostasis through the Hippo pathway, while the apoptosis pathway eliminates abnormal cells [[136]35, [137]36]. The downregulation of proliferation genes after the gastrula stage marks a shift in developmental focus toward tissue differentiation [[138]3]. Profile 3 exhibits peak expression during the lim to N3 stages and is enriched in pathways such as T-cell receptor signaling, neurotrophin signaling, and the phosphatidylinositol signaling system. These pathways may drive immune cell differentiation, synaptic formation, and neural crest migration, indicating that the lim stage is a critical node for the development of the nervous and immune systems [[139]34, [140]37]. Profile 6 exhibits a fluctuating expression pattern, with its dynamic expression significantly associated with transcriptional regulation, translation mechanisms, and the TGF-β signaling pathway. This dynamic regulatory mechanism may precisely coordinate the spatiotemporal specificity of embryonic development by phase-specific activation or inhibition of key pathways [[141]38]. In contrast, Profile 8 maintains high expression levels from the gastrula stage to the lim stage and from the N3 to N6 stage, and is significantly enriched in signaling transduction pathways such as mTOR and HIF-1. It likely provides sustained support for global differentiation and organ maturation during embryonic development by regulating metabolic adaptability [[142]39, [143]40]. Profile 9 exhibits a continuous upregulation trend and is enriched in pathways such as oxidative phosphorylation, amino acid metabolism, and lipid metabolism. These pathways provide energy for the later stages of embryonic volume expansion and post-hatching material transformation, while maintaining metabolic and tissue homeostasis [[144]3, [145]33]. The detailed KEGG enrichment results for each expression profile are provided in Table [146]S5. Fig. 6. [147]Fig. 6 [148]Open in a new tab Temporal profiles of actively expressed genes and KEGG pathway enrichment for each profile. The left panel shows the 9 temporal expression profiles identified from actively expressed genes at each developmental stage, with the number of genes in each profile indicated above the corresponding line graph, which illustrates the trends in gene expression over time. The right panel displays bubble charts of KEGG pathway enrichment analysis for each gene expression profile Overall, this study integrates CUT&Tag and RNA-seq data to reveal the dynamic changes in histone modifications and gene expression during the embryonic development of L. vannamei, highlighting significant enrichment of signaling pathways at various stages. It provides insights into the regulatory patterns and molecular mechanisms driving embryonic development in L. vannamei by linking histone modifications to gene expression changes. Identification and functional characterization of key enhancer-associated genes during L. vannamei embryonic development Enhancers are key cis-regulatory elements that regulate gene expression levels. H3K4me1 is a characteristic histone modification associated with enhancers [[149]12]. A genomic region marked by both H3K4me1 and the accessible chromatin mark H3K27ac, but lacking the repressive histone modification H3K27me3, is indicative of an active enhancer at a given developmental stage [[150]11, [151]12, [152]41]. Genes associated with such enhancers are likely to be in an enhanced expression state. Using CUT&Tag data, we identified active enhancers and their corresponding genes, hypothesizing that genes harboring active enhancers and exhibiting concurrent transcriptional activity play pivotal roles in embryonic development. To refine our selection of developmentally essential genes, we intersected enhancer-associated genes with actively expressed genes identified in the previous section, revealing key regulatory genes at each developmental stage (Fig. [153]7A). Fig. 7. [154]Fig. 7 [155]Open in a new tab Identification and KEGG functional enrichment of actively expressing genes with active enhancer marks. (A) Venn diagrams illustrating the overlap between actively expressing genes and active enhancer-marked genes. The blue circle represents the actively expressing genes, defined as genes with H3K4me3 peaks in their promoter regions, overlapping with H3K27ac peaks (≥ 1 bp overlap) but showing no overlap with H3K27me3 peaks. The pink circle represents the active enhancer-marked genes, defined as genes exhibiting H3K4me1 and H3K27ac peak overlap (≥ 1 bp) while lacking H3K27me3 overlap. The intersection of these two groups represents genes that are both actively expressed and marked by active enhancers. (B) KEGG pathway enrichment analysis of actively expressing genes with active enhancer marks. The significantly enriched pathways (p < 0.05) were visualized using a bubble plot, where bubble size represents the number of intersecting genes associated with each pathway, and color intensity reflects the statistical significance (P-value) of enrichment To elucidate the biological pathways and functional processes enriched in genes marked by active enhancers, we performed KEGG enrichment analysis. The results demonstrated a significant enrichment in signaling pathways, with stage-specific variations (Fig. [156]7B). During the gastrula and lbe stages, pathways related to cell growth and differentiation were significantly enriched, while at the lim stage, pathways associated with differentiation and the immune system were prominently enriched. From the N1 to N6 stages, pathways related to transport and catabolism were significantly enriched, which was consistent with the KEGG results of actively expressing genes (Fig. [157]7B). The characteristic biological events at each stage are closely associated with the pathway enrichment patterns [[158]3, [159]33, [160]34] Notably, during the gastrula and lbe stages, PRC-related pathways involved in H3K27me3 establishment were significantly enriched, consistent with our earlier observation of rapid H3K27me3 deposition during these stages (Fig. [161]3A, B). Additionally, gastrula-stage enrichment analysis identified the ATP-dependent chromatin remodeling pathway, suggesting that chromatin reprogramming during early embryonic development is not solely dictated by maternal factors but also requires zygotic gene activation. These findings highlight a complex interplay between histone modifications, chromatin remodeling, and zygotic genome activation. Our approach successfully identified several key regulatory genes associated with stage-specific developmental events in L. vannamei, encompassing core biological processes such as cell proliferation and differentiation, segmental pattern formation, tissue morphogenesis, molting regulation, systemic development, metabolic reprogramming, muscle differentiation, and cuticle formation (Fig. [162]8A-F, Table [163]S6). For instance, the Hox family gene [164]XM_027356713.1 (exd-like) regulates the development of body segments and the nervous system, and participates in establishing the proximal, medial, and distal regionalization patterns of appendages [[165]37, [166]42]. Its promoter maintains an active open state from the lbe to N3 stages and is regulated by enhancers (Fig. [167]8A and D). The [168]XM_027355389.1 (60a-like) gene controls imaginal disc development and embryonic morphogenesis, and its mutation leads to phenotypes in Drosophila such as small eyes, increased thoracic bristles, abnormal appendages, and female sterility [[169]43]. Its promoter remains in an active open state from the lim to N3 stages and is regulated by enhancers (Fig. [170]8B and E). The [171]XM_027365316.1 (pro-resilin-like) gene is involved in cuticle formation, enhancing flexibility and providing elastic support for joints, cuticles, and appendages [[172]44]. Its promoter maintains an active open state from the lim to N6 stages and is regulated by enhancers (Fig. [173]8C and F). Further analysis reveals a high correlation between the histone modification patterns of these key genes and their dynamic expression profiles across different developmental stages. These findings not only validate the effectiveness of our active expression gene and enhancer screening strategy but also demonstrate that histone modification signatures can serve as important molecular markers for identifying key developmental regulatory genes. Fig. 8. [174]Fig. 8 [175]Open in a new tab Representative actively expressing genes with active enhancer marks at certain stages. (A-C) Bar chart depicting the expression levels of key genes during the embryonic development of L. vannamei, quantified in transcripts per million (TPM). (D-F) Integrated genome viewer (IGV) tracks displaying RNA-seq and CUT&Tag peak profiles for RNA expression, H3K4me1, H3K4me3, H3K27ac, and H3K27me3 signals at these gene loci, illustrating their expression dynamics and histone modification landscapes throughout embryonic development. Control tracks are included for comparative analysis, providing a reference for background signal levels Through an integrative analysis of histone modifications and gene expression dynamics, this study reveals how histone modifications cooperate with transcriptional regulation to drive embryonic development. This systematic approach not only enables the precise identification of developmentally essential genes but also offers novel insights into the molecular mechanisms underlying early embryogenesis in L. vannamei. Discussion Embryonic development is initially governed by maternal gene products, and control is gradually transferred to the zygotic genome during zygotic genome activation (ZGA). This transition is tightly regulated through dynamic changes in histone modifications, which have been extensively studied in model organisms but remain largely unexplored in crustaceans [[176]6]. In this study, we provide the first high-resolution, genome-wide maps of four histone modifications (H3K4me1, H3K4me3, H3K27ac, and H3K27me3) during embryonic development in a crustacean species. By integrating CUT&Tag data with transcriptomic profiles, we systematically identified cis-regulatory elements and their stage-specific activity, thus offering novel insights into the epigenetic regulation of zygotic genome activation (ZGA), cell lineage specification, and morphogenesis in L. vannamei embryos. Histone modifications and ZGA in L. vannamei Although the timing and patterns of histone modification reprogramming and ZGA vary among species, these processes are evolutionarily conserved in sexually reproducing animals [[177]31, [178]45, [179]46]. Since germ cells are highly specialized, they must regain cellular totipotency after fertilization to enable proper embryonic development and tissue differentiation. Our findings suggest that ZGA in L. vannamei likely occurs before or around the blastula stage. This conclusion is supported by the significant increase in expressed genes from the blastula to gastrula stages (Fig. [180]3D), suggesting large-scale zygotic genome activation. Additionally, the presence of active histone modifications in the blastula stage and their substantial upregulation in the gastrula stage further corroborate this (Fig. [181]3B). Due to the reliance of L. vannamei on external fertilization mechanisms (where males transfer spermatophores to the female’s thelycum during mating, and sperm-egg binding occurs externally upon ovulation) [[182]47], coupled with the rapid transition from multicellularization to the blastula stage within minutes post-fertilization, the developmental process is highly sensitive to environmental fluctuations such as temperature and salinity variations [[183]44, [184]47, [185]48]. This significantly exacerbates asynchrony in embryonic development prior to the blastula stage. Consequently, obtaining earlier-stage samples to precisely determine the timing of the maternal-to-zygotic transition (MZT) remains challenging. Previous studies in model species and humans already showed tremendous difference in the timing of MZT and dynamics of histone modifications during embryogenesis across a broad range of species [[186]6, [187]49]. Comparative analysis of L. vannamei with other marine invertebrates also highlights interspecies differences in MZT timing and histone modification dynamics. In Mulinia lateralis, MZT occurs between the morula and gastrula stages, with weak histone modification signals before the morula stage. H3K27me3 is established first, while H3K4me1 and H3K4me3 are significantly upregulated only until the gastrula stage (our unpublished data). In contrast, Crassostrea gigas exhibits high levels of all four histone modifications as early as the zygote stage [[188]50]. These findings suggest that, despite evolutionary proximity, species-specific differences exist in histone modification reprogramming and MZT timing. Future studies incorporating more species and higher-resolution techniques such as single-cell CUT&Tag could provide deeper insights into chromatin accessibility and cell-type-specific regulatory mechanisms during early development. Regulation of key developmental processes Our study identified significant enrichment of H3K4me1, H3K27me3, and H3K27ac at the N1 stage, while H3K4me3 levels peaked at the lim stage. This suggests that histone modification dynamics are closely linked to the developmental needs of each stage. The increased H3K4me3 during the lim stage may reflect extensive tissue differentiation as the embryo prepares to hatch, while the concurrent enrichment of activating (H3K4me1, H3K27ac) and repressive (H3K27me3) marks at the N1 stage suggests a complex interplay between activation and repression during metamorphosis [[189]3, [190]33]. The time-series analysis of histone marks and the transcriptome revealed a high degree of temporal correlation between the two, indicating that histone modifications and transcriptional activities collaboratively drive gene regulatory processes during early embryonic development. This underscores the significance of epigenetic mechanisms in orchestrating developmental transitions. L. vannamei exhibits unique stages during early embryonic development, undergoing significant physiological changes, particularly in tissue formation, dietary transitions, muscle development, and exoskeleton remodeling. However, current research on early embryonic development of L. vannamei mainly focuses on morphological, biochemical, and transcriptomic characteristics, with limited studies on epigenetic aspects, particularly histone modifications [[191]3, [192]33, [193]34]. In this study, we utilized CUT&Tag and RNA-seq analysis for the first time to explore potential molecular mechanisms regulating physiological changes during early embryonic development from the perspective of histone modification-mediated gene expression. Identification of key developmental genes and their cis-regulatory elements Through a comprehensive analysis of histone modifications and transcriptomic data, we identified genes that are actively expressing at each stage and further screened out those with active enhancer marks. This approach not only circumvents the interference of maternal mRNA fluctuations in transcriptomic analysis but also enables precise identification of developmental regulatory genes and their networks through epigenetic features, providing new insights into the molecular mechanisms underlying embryonic development in invertebrates. By defining “active open regions” (genomic regions marked by both H3K4me3 and H3K27ac activation marks but lacking the repressive H3K27me3 modification) and integrating RNA-seq expression data (TPM > 0.5), we successfully identified epigenetically regulated actively expressed genes across various stages of embryonic development in L. vannamei (Fig. [194]5A). To further elucidate the regulatory mechanisms of key genes, we screened for genes marked by active enhancers by combining the co-localization signals of the enhancer marker H3K4me1 and the open chromatin marker H3K27ac (Fig. [195]7A). These genes not only exhibited significant transcriptional activity but also showed a strong correlation between the dynamics of histone modifications in their enhancer regions and the stage-specific functional requirements. For instance, the promoter regions of the segmental and nervous system development regulator gene exd-like [[196]37, [197]42] and the cuticle formation gene pro-resilin-like [[198]44] maintained an active chromatin state from the lim to the nauplius stages, with their enhancer modification levels significantly upregulated during development, showing high synchronization with gene expression dynamics (Fig. [199]8). This finding indicates that epigenetic regulation and transcriptional activation collaboratively and precisely drive the execution of developmental programs. This study represents the first successful localization of key developmental genes and their cis-regulatory elements in L. vannamei through dynamic histone modification profiling. This strategy not only validates the feasibility of histone modifications as predictive markers of gene expression but also provides valuable candidate genes and regulatory element resources for future functional genomics research [[200]11, [201]12]. In addition, we conducted an in-depth analysis of the chromatin states of L. vannamei using the open-source software package ChromHMM [[202]22]. By annotating chromatin states based on histone modifications, our study provides a valuable resource for future basic biological research on shrimp embryonic development. However, it is worth noting that although our integrated CUT&Tag and RNA-seq analyses revealed a strong correlation between predicted regulatory elements and gene expression patterns, cellular heterogeneity inherent in whole-embryo samples may introduce confounding signals into chromatin state annotations. In future studies, the application of single-cell CUT&Tag or spatially resolved epigenomic techniques will be essential for generating more precise, stage- and tissue-specific chromatin state maps. Conclusions This study provides new epigenetic resources for the economically significant crustacean species L. vannamei. Notably, this is the first successful application of CUT&Tag in L. vannamei. By analyzing histone marks (H3K4me1, H3K4me3, H3K27me3, and H3K27ac) and corresponding transcriptomes at seven critical stages of early embryonic development, we revealed that dynamic changes in histone modifications coincide closely with transcriptional activation during the ZGA of L. vannamei. This correlation suggests that histone modifications and transcriptional activity jointly drive gene regulation during early embryonic development, offering new insights into the ZGA process in shrimp. Additionally, through differential expression analysis, and integrated analysis of histone modification and transcriptome data, we explored potential molecular mechanisms underlying physiological changes during early embryonic development mediated by histone modifications. This study identified a series of potential key genes involved in development, providing new insights from the perspective of histone modification-mediated gene expression. This represents the first characterization of histone modifications during early development in shrimp. The findings of this study will serve as a significant reference for research in shrimp developmental biology and aquaculture. Supplementary Information Below is the link to the electronic supplementary material. [203]Supplementary Material 1^ (9.4KB, xlsx) [204]Supplementary Material 2^ (11.2KB, xlsx) [205]Supplementary Material 3^ (11.1KB, xlsx) [206]Supplementary Material 4^ (13KB, xlsx) [207]Supplementary Material 5^ (29.4KB, xlsx) [208]Supplementary Material 6^ (14.1KB, xlsx) [209]Supplementary Material 7^ (2MB, docx) Acknowledgements