- Research
- Open access
- Published:
Chromosome-level genome assembly and single-cell analysis unveil molecular mechanisms of arm regeneration in the ophiuroid Ophiura sarsii vadicola
Genome Biology volume 26, Article number: 82 (2025)
Abstract
Background
Ophiuroids, belonging to Ophiuroidea in Echinodermata, possess remarkable regenerative capacities in their arms, relying on cellular recruitment and de-differentiation. However, limited high-quality genomic resources have hindered the investigation of the underlying molecular mechanisms of ophiuroid regeneration.
Results
Here, we report a chromosome-level genome of Ophiura sarsii vadicola, 259.28 Mbp in length with a scaffold N50 length of 66.91 Mbp. We then perform bulk and single-cell RNA sequencing analysis to investigate gene expression and cellular dynamics during arm regeneration. We identify five distinct cellular clusters involved in the arm regeneration and infer the dynamic transformations from sensory stimulation to injury response, wound healing, and tissue regeneration. We find that progenitor cells derived from connective tissue cells differentiate into muscle, cartilage, endothelial, and epithelial cells. Pseudotime analysis indicates that muscle differentiation occurs early in the regeneration process.
Conclusions
Our genomic resource and single-cell atlas shed light on the mechanisms of organ regeneration in ophiuroids.
Background
Regeneration is a common and essential process across all organisms, from unicellular to multicellular forms, and occurs in both invertebrates and vertebrates, although regenerative capacities vary among different taxa [1, 2]. Echinodermata, a phylum of deuterostomes sharing a common ancestor with Chordata, possesses robust and rapid regenerative capacity [3]. Organ regeneration is a prevalent characteristic across all five classes of the phylum Echinodermata in response to trauma and occurs in various body parts, including arms [4,5,6], internal organs [7, 8], and even across the entire body [9]. Stellate echinoderms, including crinoids, asteroids, and ophiuroids, display unique combinations of regenerative abilities through re-epithelialization, inflammatory responses, and remodeling of post-injury tissues, with arm regeneration being one of the most extensively studied processes [10, 11].
Brittle stars (Ophiuroidea) are the most diverse group of echinoderms, comprising over 2100 species worldwide [12]. Ophiuroids have emerged as an ideal model for studying organ regeneration in both natural [13] and experimental settings [14, 15], due to their exceptional regenerative abilities and ease of laboratory maintenance. Their rapid regeneration rates [16], distinct morphogenesis [6], diverse morphologies [17], and flexible locomotion with different arm vertebrae shapes [18] provide valuable insights into regeneration mechanisms in deuterostomes and the evolution of regenerative capabilities across taxa.
Studies have been conducted to understand several aspects of arm regeneration in brittle stars [6, 19]. Arm regeneration comprises four main stages: wound healing/repair, early regeneration, intermediate regeneration, and advanced regeneration, each marked by specific cellular and molecular events [14]. Interactions between wound healing and early regeneration stages, coupled with the transmission of cytoplasmic molecular regeneration signals, determine whether ophiuroids successfully progress through all four stages [14]. In recent decades, several studies have sought to unveil the underlying gene regulatory networks involved in different regeneration phases, employing various molecular and cellular techniques, including in situ hybridization, immunohistochemistry [20], proteomics [15], and transcriptomics [21,22,23]. Notably, two prominent signaling pathways were found to play key roles during arm regeneration: the fibroblast growth factor (FGF) pathway [24], which activates fibroblasts and facilitates cellular proliferation and tissue repair, and the Wnt [24, 25] pathway, which stabilizes stem cells and promotes their differentiation. However, uncertainties remain regarding arm regeneration, including the comprehensive understanding of cellular dynamics, cell type interactions during the regenerative process, and the signaling pathways and gene regulatory networks involved in each cell type.
Over the past decade, the emergence of single-cell RNA sequencing (scRNA-seq) has revolutionized the study of regeneration mechanisms [26]. This powerful technique has provided new insights into cellular composition [27], gene expression dynamics, and cell differentiation trajectories during regeneration [28]. Moreover, scRNA-seq has revealed previously unknown cell types and identified unique gene expression signatures associated with specific regeneration stages [3]. This approach has been successfully applied to investigate regeneration in diverse model organisms, including earthworm segments [29, 30], mouse muscles [31], African clawed frog tails [32], and axolotl limbs [33]. These investigations highlight the potential of single-cell analysis in elucidating arm regeneration mechanisms in brittle stars. However, the limited availability of high-quality reference genomes for the class Ophiuroidea, with only two published genomes in 2024 [34, 35], has constrained the application of scRNA-seq in understanding the molecular mechanisms of regeneration in ophiuroids [16].
The brittle star Ophiura sarsii vadicola (Ophiurida: Ophiuridae) is a dominant epibenthos in the Yellow Sea [36, 37] that exhibits remarkable natural regeneration capacity. In this study, we generated a high-quality chromosome-level reference genome for O. sarsii vadicola to facilitate comparative genomic and transcriptomic analyses of its regeneration capacity. To gain insights into the molecular mechanisms underlying wound repair and early regeneration stages, we employed scRNA-seq technology to generate a dynamic multicellular transcriptomic profile spanning regeneration stages. By analyzing amputation tissues collected from different time points, we elucidated the composition and gene expression dynamics of cellular constituents involved in arm repair and regeneration. Leveraging the findings from single-cell analysis, we proposed a hypothesis regarding the transformation of cell types during different stages of arm regeneration, providing a foundation for future investigations into echinoderm regeneration mechanisms.
Genome assembly and characterization
Specimens of O. sarsii vadicola were collected from the Yellow Sea for laboratory culture and genome sequencing. A total of 143.7 Gbp (114.47×) of PacBio SMRT long reads and 218.32 Gbp (173.92×) of Illumina short reads were generated for genome assembly (Additional file 2: Table S1). K-mer analysis estimated the size of O. sarsii vadicola genome to be 1.21 Gbp with a heterozygosity rate of 2.21% (K-mer = 17, Additional file 1: Fig. S1). Using a hybrid assembly approach with 362.02 Gbp of sequence data, we assembled a 1.27 Gbp genome, consisting of 1988 contigs with an N50 of 2.44 Mbp and GC content of 37.2% (Additional file 2: Table S2). After HI-C scaffolding, 99.62% of the assembled contigs were anchored into 19 chromosomes, yielding a final genome size of 1.27 Gbp across 216 scaffolds with an N50 of 66.91 Mbp (Fig. 1A, Additional file 1: Fig. S2 and Additional file 2: Tables S3–S4). Repeat sequences constituted 54.8% of the assembly (Additional file 2: Table S5), predominantly comprising long terminal repeats (LTRs, 48.69%).
Genomic features of Ophiura sarsii vadicola and phylogenetic relationships among echinoderms with available genome data. A Circos plot of Ophiura sarsii vadicola genomic features with an image of an O. sarsii vadicola specimen (5 cm in size). Circles show (1) the length of each chromosome, (2) the density of genes, (3) the density of repetitive sequences, and (4) GC content. B Phylogenetic relationships and divergence times of ten echinoderms with genome resources. A yellow asterisk marks the node where time calibration was applied for divergence time analysis. The pie diagram (orange and blue) shows the percentage of expanded and contracted gene families for each tip species. C Collinearity relationships between O. sarsii vadicola (Osav) and O. sarsii (Osar), and between O. sarsii vadicola and Amphiura filiformis (Afil)
We validated the genome assembly quality using multiple methods. The high quality of the genome assembly was demonstrated by the 96.62% mapping rate of Illumina reads covering 99.59% of the genome with an average sequencing depth of 143.30× (Additional file 2: Table S6). The quality value (QV) calculated with the Merqury package was 36.88, indicating 99.9% accuracy. Genome completeness, assessed using the Core Eukaryotic Genes Mapping Approach (CEGMA), showed 93.95% of the 248 core eukaryotic genes (Additional file 2: Table S7). The genome encodes 26,226 protein-coding genes, with an estimated 94.0% Benchmarking Universal Single-Copy Orthologs (BUSCO) completeness score; 92.3% of these were annotated (Additional file 1: Fig. S3 and Additional file 2: Table S7). The average transcript length was 22,018 bp (exons + introns), with mean lengths for coding sequences (CDS, 1583.07 bp), exons (218.82 bp), and introns (3277.68 bp) comparable to those in other echinoderms (Additional file 1: Fig. S4 and Additional file 2: Tables S8–S9).
A total of 10 echinoderm genome assemblies were used to conduct comparative analysis (Fig. 1B). We identified 1537 single-copy orthogroups among a total of 27,305 orthogroups. O. sarsii vadicola exhibited 48 significantly expanded gene families (Additional file 2: Table S10), including the coagulation factor 5/8 C-terminal domain and the discoidin domain (FA58C), both known to be involved in post-amputation coagulation. Phylogenetic analysis confirmed crinoids as the basal echinoderm lineage, with ophiuroids most closely related to asteroids, and echinoids grouped with holothuroids. The ophiuroid-asteroid and echinoid-holothuroid clades diverged around 378–553 Ma, with ophiuroid diversification occurring earlier than other clades (170–350 Ma).
The three brittle star species shared 11,588 orthogroups, with O. sarsii vadicola, O. sarsii, and Amphiura filiformis possessing 269, 381, and 1444 species-specific orthogroups respectively (Additional file 1: Fig. S5). Enrichment analysis of species-specific orthogroups revealed distinct functional adaptations. O. sarsii vadicola showed enrichment in sensory-related functions, including visual perception, G-protein coupled receptor (GPCR) signaling pathway, and sensory perception of pain (Additional file 2: Table S11). All three ophiuroids shared enrichment in visual and sensory (such as visual perception and GPCR, Additional file 2: Tables S12–S13). O. sarsii displayed unique enrichment in sensory perception of temperature stimulus and pain, regulation of growth, response to oxidative stress, stress-activated MAPK cascade, and wound healing (Additional file 2: Table S12). A. filiformis showed distinct enrichment in Wnt signaling pathway, wound healing, sensory perception of pain and sound, and response to oxidative stress and bacterium (Additional file 2: Table S13). Chromosomal structure analysis revealed high conservation between O. sarsii vadicola and O. sarsii compared to A. filiformis (Fig. 1C). We identified nine conserved chromosomes (chr) between the two superorders Euryophiurida (O. sarsii vadicola and O. sarsii) and Ophintegrida (A. filiformis) in the order Ophiurida. Several rearrangements of chromosomes were observed, including the fusion of A. filiformis chr5, chr11, and part of chr2 into chr1 in the Ophiura species, and the split of A. filiformis chr1 into chr7 and chr19 in the Ophiura species.
Arm regeneration experiment, morphological observations, and bulk RNA analysis
To identify crucial stages of arm regeneration in O. sarsii vadicola, we investigated both morphological changes and gene expression profiles in the distal tissues at four post-amputation time points: spanning wound repair (days 1 and 3) and early regeneration (days 7 and 13). Complete wound healing, observed within 3 days post-amputation (Additional file 1: Fig. S6), marked the transition to early regeneration phase, consistent with previous reports of brittle star regeneration [29]. The wound surface recovered within 1 to 3 days, and an obvious distal blastema was observed by day 13 (Fig. 2A).
Arm regeneration and differential gene expression in O. sarsii vadicola at four post-amputation time points. A Histological observations of the arm tip of O. sarsii vadicola on day 13 post-amputation. N: nerve tissue, M: muscle tissue, V: vertebral structure. B Zoomed-in view of the blastema from A. N: nerve tissue, b: blastema, RWC: radial water canal, ACC: aboral coelomic cavity, ct: connective tissue. C Principal component analysis (PCA) of gene expression profiles at four time points during regeneration. D Up- and downregulated gene expression at day 13 compared to earlier time points. E Annotation and enrichment analysis of differentially expressed genes at day 13 post-amputation compared to earlier time points
Analysis of bulk RNA sequencing data revealed distinct transcriptional patterns. Principal component analysis (PCA) revealed a clear distinction in gene expression profiles between day 13 (D13) samples and earlier time points (Fig. 2C and Additional file 1: Fig. S7). Pairwise comparisons (D13 vs D1, D13 vs D3, D13 vs D7, D13 vs three other groups) were conducted using DESeq2 to identify differentially expressed genes (DEGs) across regeneration stages (Additional file 2: Table S14). Comparisons between day 13 and the earlier three time points (days 1, 3, 7) revealed 2675 significant DEGs, comprising 1149 significantly upregulated and 1526 significantly downregulated genes (Fig. 2D, D13 vs D [1, 3, 7]). The D13 DEGs were predominantly enriched in ribosomal genes, indicating a transition to the tissue growth phase of regeneration. Among these DEGs, we identified 26 genes with known regenerative functions (Additional file 2: Table S14).
Comparative analysis between the stable growth phase (D13) and day 1 revealed 788 significant DEGs, including 423 upregulated and 365 downregulated genes (Fig. 2D). During this critical first day post-amputation, we identified eight regeneration-related DEGs (Fig. 2D, D13 v D1, Additional file 2: Table S14). These included three significantly upregulated genes involved in immunity and wound healing (e.g., MUC2) and five upregulated genes enriched in key pathways: endocytosis (ko04144), leukocyte transendothelial migration (ko04670), and T cell receptor signaling pathway (ko04660). This expression pattern indicates robust activation of immune response pathways immediately following amputation. By day 3 (D3), we identified 1823 significant DEGs (Fig. 2D, D13 vs D3), including nine regeneration-related DEGs. Within the 726 significantly upregulated genes, we identified four genes, including MAP-1B and CSRP2, associated with nervous system development and smooth muscle differentiation. Pathway enrichment analysis revealed disease controls (apelin signaling pathway, ko04371) and cell differentiation/proliferation (Hippo signaling pathway), suggesting concurrent regulation of immune functions and preparation for tissue regeneration on day 3. Day 7 exhibited substantial changes in DEG functionality compared to day 13, with nine regeneration-related genes among 1539 significant DEGs (Fig. 2D). Significantly upregulated genes included SIK3 and FAM13A, which regulate growth and regeneration signals through GPCR and mTOR pathways. Additionally, we observed significant enrichment of the mitogen-activated protein kinases (MAPK) signaling pathway (ko04010), which mediates various cellular functions including proliferation, differentiation, and migration.
Single-cell transcriptome analysis during regeneration
To elucidate the cellular composition and dynamics during wound healing and early regeneration, scRNA-seq analysis was conducted for the regenerative arm tissues collected at days 1, 3, 7, and 13 post-amputation, using day 0 wound tissue as the control group. Individual sample cell counts ranged from 3024 (D13) to 5399 cells (D7, Additional file 2: Table S15). After quality filtering and normalization, we retained 21,001 cells for downstream analysis (Additional file 1: Fig. S8 and Additional file 2: Table S16). Using t-distributed stochastic neighbor embedding (t-SNE), we identified and annotated six major cell clusters based on marker gene expression profiles (Fig. 3B, Additional file 1: Fig. S9, and Additional file 2: Table S17): two connective tissue clusters (CT I and CT II, marked by Lrp6 and ACTB), muscle cells (marked by Mhc and Mp20 for body movement), nerve cells (marked by ADCY3 and KCNK13 for nerve signaling), immune cells (marked by psap and SRCR for phagocytosis and nonspecific immunity), and endothelial cells (marked by FCGBP and MFGE8 related to water vascular systems).
The CT clusters constituted the largest cell population (68.6% of total cells, Fig. 3A, Additional file 2: Tables S16, S18) and showed significant upregulation of regeneration markers, including profilin (Fig. 3C, marker genes of CT I), a known stem cells marker in sea urchins [31]. Detailed analysis revealed two distinct CT subpopulations. CT I was characterized by significant expression of cofilin and ACTB, along with other actin-regulatory genes [38, 39]. CT II showed active expression during regeneration, with upregulation of developmental/regenerative genes including BP10 and Lrp6 [40, 41]. Functional annotation (Additional file 2: Table S17) revealed that CT I was involved in rapid passive post-amputation responses, while CT II activated healing and regeneration processes through the expression of connective tissue-related genes (e.g., Ppn and TIMP1, Fig. 3C) and regeneration-related genes like VEGF and Hyalin (Fig. 3C).
Cell type annotations of O. sarsii vadicola arm tips at various time points based on scRNA-seq data. A Bar plot showing cell number proportions. B t-distributed stochastic neighbor embedding (t-SNE) diagram for the single-cell RNA of O. sarsii vadicola. Each point represents a cell with circles highlighting CT I and CT II cells. C Expression levels of cell type marker genes within the six clusters. Circle sizes indicate the proportion of cells in each cluster expressing a given gene, while the color reflects the mean expression across those cells. D Expression levels of marker genes across the six cell clusters at various time points (X-axis), with the Y-axis showing expression levels
We observed distinct temporal patterns in gene expression across regeneration stages. Immune system-related genes, such as DMBT1, showed initial suppression followed by increased expression (Fig. 3D). Profilin and the actin regulator ARHGDIA maintained significant expression during all regeneration phases. Lrp6 and VEGF exhibited gradual increase during the early stage of regeneration (Fig. 3D), suggesting CT II cells’ direct involvement in the regenerative process. Marker genes of neural cell populations, such as Phm and PCSK2, showed marked upregulation by day 13, suggesting that neural regeneration initiated between days 7 and 13. Additionally, the expression of immune-related genes was affected to varying degrees after amputation. DMBT1 exhibited transient suppression followed by gradual recovery (Fig. 3D), while the pasp gene, which mediates macrophage phagocytic function, showed minimal changes.
To further explore the roles of CT I and CT II groups during arm regeneration, we separated these two groups and re-analyzed their expression profiles using t-SNE clustering (Fig. 4A–B). We identified four cell subgroups in CT I cells with relevant marker genes (Additional file 2: Table S19): mesenchymal stem cells (MSCs), glial fibroblasts (marked by various collagens), glial effector cells (marked by actin-related genes), and state sensing cells (Fig. 4C, marked by HSP70). In addition, we identified five cell subgroups in CT II cells based on marker genes (Fig. 4D, Additional file 2: Table S20): cartilage cells, myogenic progenitor cells, functional endothelial-like cells, epithelial cells, and juxtaligamental cells. The coordinated activity of diverse CT cells appeared to drive the continuous healing and regeneration in O. sarsii vadicola (Fig. 4E). Following receipt of injury signals (NF-κB and TNF-α) by state sensing cells, epithelial cells undergo conversion to mesenchymal stem cells (MSCs). These MSCs regulate cell differentiation and proliferation while migrating along the coelomic and water vascular system endothelium toward the amputation site to establish the blastema. During this migration process, MSCs continuously differentiate into diverse cell types (muscle cells, neuron cells, cartilage-like cells, juxtaligamental-like cells, Fig. 4E, solid arrows) and propagate regenerative signals (Fig. 4E, dashed arrows) through the Wnt/β-catenin signaling pathway by state sensing cells, effector cells, and neuron cells (Fig. 4E).
Gene-expression dynamics of early-stage regeneration at a single-cell resolution
Several key gene expression events occurred after arm amputation. State sensing cells (marked by SCOP2 and HSP70, Additional file 2: Table S19) initially detected physiological changes, simultaneously activating glial effector cells and glial fibroblasts. This stress response facilitated rapid cellular state transitions. Compared with the control group (day 0), endothelial cell abundance rapidly increased within the first 3 days post-amputation (Fig. 3A). Wound healing was initiated through the expression of fibrinolysis genes, including Klkb1, (Additional file 2: Table S20), followed by activation of regenerative signaling pathways, including NOTCH and Wnt/β-catenin. MSCs, characterized by expression of NOTCH 1, fibrosurfin, and other genes (Fig. 4C, Additional file 2: Table S19), integrated signals from the NOTCH, Wnt/β-catenin, and GPCR pathways [19, 41,42,43], bridging emergency responses and regeneration while regulating cellular differentiation and proliferation [20]. Notably, fibrosurfin, crucial for inter-fibrous assembly in connective tissue regeneration [44], showed specific upregulation. Whole-mount in situ hybridization (WMISH) results revealed that fibrosurfin expression was primarily in intervertebral regions and the body cavity (Additional file 1: Fig. S10), extending across the regeneration region by day 13.
Regulated by MSCs, four CT II subgroups performed distinct functions (Additional file 2: Table S20). Epithelial cells expressed mucous membrane markers ADAM9 and MFGE8 [45, 46], mediating endothelial tissue regeneration through BP10 expression. Cartilage-like cells, characterized by COL9A1 and COL2A1 expression, facilitated ligament and bone regeneration through COL11A1. Myogenic progenitor cells, marked by high AGRN and NCNA expression, may drive muscle and tendon regeneration via COL13A1, while functional endothelial-like cells and juxtaligamental cells performed similar neurosecretion functions (PCSK2, Gyc76C), with endothelial-like cells specifically supporting nerve regeneration through NRSN and Shtn1 expression. To verify key genes expression in CT II subgroups during tissue regeneration, we conducted WMISH analysis targeting the Lrp6 gene, a myogenic progenitor cell marker. This gene, which encodes a Wnt co-receptor and activates the Wnt signaling pathway through LDL-receptor activity, showed localized expression in muscle tissue at the regenerating arm tip (Additional file 1: Fig. S10).
To elucidate the sequential progression of tissue regeneration, the Monocle2 program was used to analyze the pseudotemporal ordering of CT II cells (Fig. 5). Integration of pseudotime trajectory (Fig. 5A) and differentiation status plot (Fig. 5B) revealed that the right trajectory branch marks the initiation of differentiation, subsequently diverging into branches 2 and 3 at the differentiation node. Cell population distribution along the pseudotime axis (Fig. 5C) indicates a sequential differentiation pattern: muscle progenitor cells (marked by AGRN and Naca, Fig. 5D) initiated the process, followed by chondrocyte differentiation, ultimately branching into neural and epithelial tissue regeneration pathways. The first branch involved the expression of cartilage genes (i.e., COL9A1 and COL2A1) and neural markers (i.e., Gyc76C and PCSK2), while the second branch showed enrichment in mucosal endothelial genes (MFGE8 and Fcer2). Additionally, the mesenchymal stem cell marker Lrp6 and VEGF showed sustained activity during early regeneration (until day 13), coinciding with muscle cell regeneration (Fig. 5E). Validation using Slingshot analysis of CT II cells (Additional file 1: Fig. S11) corroborated the Monocle2 results, confirming muscle progenitor cells as the initiating population, followed by cartilage cell differentiation, before diverging into distinct endothelial cells and juxtaligamental-like cells.
Pseudotiming analysis of CT I and CT II cells during regeneration. A Pseudotime ordering of cells. B Differentiation status of each branch. C Distribution of each cell group along the pseudotime axis. D Marker gene expression in cell subgroups over pseudotime. E Expression of Lrp6 and VEGF over pseudotime, with the X-axis showing pseudotime and the Y-axis showing expression levels
Discussion
Chromosome-level genome features in the phylum Echinodermata
The limited genome resources of brittle stars have impeded effective genomic analyses for this group, particularly for cellular and molecular studies related to regeneration. The draft genome of Ophioderma brevispinum, reported in 2022, was the first sequenced ophiuroid genome [16]. Recently, two additional ophiuroid genomes were sequenced [34, 35]. Our genome showed higher completeness (94.0%) and comparable assembly quality, with a scaffold N50 of 66.91 Mbp, relative to O. sarsii (93.5%, 78.3 Mbp) and A. filiformis (92.7%, 68.8 Mbp) (Additional file 2: Table S4). Comparative genomic analysis (Additional file 2: Table S10) revealed that some rapidly evolving gene families, such as GPCR, play essential roles in skeletal [47] and neuronal regeneration [48]. The high-quality O. sarsii vadicola genome documented here provides a valuable new resource for studies, particularly in regeneration research.
The three sequenced species represent two superorders: Euryophiurida (O. sarsii vadicola and O. sarsii) and Ophintegrida (A. filiformis). These three genome resources from two superorders provide a valuable foundation for diverse downstream analyses. In this study, analysis of their genomic data revealed similarities and divergences, highlighting adaptations in sensory perception and wound healing post-amputation (Additional file 2: Tables S11–S13). This functional differentiation between O. sarsii and O. sarsii vadicola revealed key adaptations in temperature response and oxidative stress pathways, likely reflecting response to historical geographical events in their habitats [49]. Population genetics and other molecular techniques will be employed to further reveal the historical dynamics of their divergence and environmental adaptation. We identified several conserved chromosomes and rearrangements across the two superorders in Ophiuroidea. Macrosynteny analysis serves as a powerful tool for tracking orthologous chromosomes across highly divergent species and is increasingly valuable for studying genome rearrangements [50]. Future research should determine the precise timeline of chromosomal rearrangements and investigate the contributions of repeat expansion and chromatin architecture [35] in ophiuroids.
Organ regeneration at the cellular level in ophiuroids and other echinoderms
Regeneration in ophiuroids has been investigated across species from the orders Amphilepidida, Euryalida, and Ophiurida, with regeneration times ranging from weeks to months [13]. Most studies have centered on A. filiformis (superorder Ophintegrida), examining morphological features [14], cellular differentiation [51], and gene expression [52]. A recent genomic and comparative transcriptomic study [35] systematically analyzed gene expression during adult arm regeneration. However, regeneration in other ophiuroid taxonomic groups remains poorly understood, especially given their high morphological and skeletal disparities. Regeneration biology and histology have been documented for the order Euryalida (basket star) in the superorder Euryophiurida [6], highlighting distinct morphogenesis capabilities relative to Ophintegrida. Studies of other groups in Euryophiurida remain limited. Within Ophiurida, although regeneration rates have been reported for some species [13], detailed follow-up studies are lacking. Only one study has examined regeneration in Ophiurida, demonstrating slow regeneration in Ophionotus victoriae (family Ophiuridae) through transcriptomic analysis [53] and identifying relevant gene families and pathways, including Hox, Sox, Wnt/β-catenin, TGF-β, and Notch families.
This study represents the first comprehensive molecular investigation of regeneration in Ophiurida, examining early arm regeneration in O. sarsii vadicola through multiple techniques. Distinct changes in both cell types and gene expression profiles were observed during the transition from wound repair (days 1 and 3) to early regeneration (days 7 and 13). The results suggest that regeneration occurs through epimorphosis, involving de-differentiation, differentiation, and proliferation of epithelial and other cell types in the wound and blastema areas (Fig. 5). After distal arm amputation, recovery begins with immune and stress responses from functional endothelial-like cells (HSP70 and SCOP2). Similar to other echinoderms [54, 55], the amputated surfaces of O. sarsii vadicola coordinate passive mechanical changes. Effector cells stimulated neural control and muscle activity, increasing collagen fibers produced by fibroblasts to support wound repair, primarily through innate immune responses. While MSCs likely regulate immune function and tissue regeneration [56, 57], no specific O. sarsii vadicola MSC genes were identified in this study. During early regeneration, MSCs may regulate immune function through NF-κB and MAPK pathways by inhibiting genes such as DMBT1 (Additional file 2: Table S19), thereby reducing epithelial differentiation.
The cellular composition during regeneration exhibits considerable complexity. During epimorphosis, both blastema and arm tissues undergo various cellular processes, including differentiation, proliferation, de-differentiation, and re-differentiation [11]. During arm regeneration in ophiuroids, re-epithelialization occurs around the wound site, where endothelial cells transform into MSCs through NF-κB and TNF-α pathways [58, 59]. Morphological studies of ophiuroid and crinoid regeneration suggest that MSCs in the blastema possess pluripotent capabilities and originate from migrating coelomic epithelium [60, 61]. Early calcified spicules initially form within MSCs and are regenerate through specific cell signaling. Concurrently, tissue differentiation is initiated via the Notch and Wnt/β-catenin signaling pathways [62]. Coelomic tissue cells, including muscle cells, also contribute to regeneration by producing cells that differentiate into various tissue types [63, 64]. Muscle cells de-differentiate from myoepithelial cells before re-differentiating [14], whereas in starfish, cavity cells originate from the cavity epithelium [65]. Studies have shown that muscle tissue de-differentiation and regeneration commonly occur in mature echinoderm muscles [10]. De-differentiated muscle cells of the sea cucumber (Holothuria tubulosa) contribute to endothelial regeneration in the water vascular system [66], while the presence or absence of longitudinal muscle critically influences planarian regeneration [67]. In our study, muscle regeneration was observed during the initial stage of arm regeneration. However, further investigation is needed to fully understand the potential roles and mechanisms of muscle regeneration in echinoderms.
Connective tissue provides a crucial migratory matrix during blastema and arm tissue regeneration [68]. CT cells emerged as the predominant functional cell type in O. sarsii vadicola arm regeneration (Fig. 3). Based on scRNA-seq analyses, we further annotated and identified cell types within this group (Fig. 4) and tracked their dynamics (Fig. 5) to reveal cellular mechanisms. Within CT groups, myogenic progenitor cells served as the primary source of muscle regeneration, responding to Wnt/β-catenin signals from MSCs. Through pseudotime analysis of dynamically expressed marker genes, we found that these myogenic progenitor cells may also initiate tissue differentiation. Moreover, we found that RACK1 inhibited Wnt/β-catenin signaling (Additional file 2: Table S20), suggesting that the direction of differentiation for each tissue type may depend on the timing of signals to cartilage cells.
Single-cell sequencing in regeneration studies of model organisms
Our study highlights the essential role of CT cells in ophiuroid arm regeneration, although the original cellular source remains uncertain. Connective tissue, comprised extracellular matrices and abundant interconnecting cells, can revert mature cells to a homogeneous progenitor state during regeneration, as demonstrated in forelimb regeneration in the salamander Ambystoma mexicanum [33]. Specific connective tissue cell types can regenerate into their corresponding tissues, as observed in periosteal cartilage regeneration at human bone defect sites [69] and mouse muscle tissue regeneration by muscle stem cells and fibroblasts [70]. Similar connective tissue cell types were also identified in O. sarsii vadicola, including MSCs, myogenic progenitor cells, and cartilage-like cells. The presence of connective tissue fibers in the regeneration region, as shown in the clustering results (Fig. 3A), further supports the essential role of connective tissues in O. sarsii vadicola regeneration.
CT cells appear to serve similar functions in A. mexicanum and O. sarsii vadicola (Additional file 1: Fig. S12). Our analysis revealed a high similarity between CT cells in this study and those reported in axolotl [71]. Our study produced 21,001 cells for five stages (an average of 4200 cells), close to the output of axolotl limb regeneration (~5000 cells) studies on average [48, 71]. We identified 13 core marker genes, annotated as ribosomal proteins (e.g., S and L genes, RPA and RPL), with elevated expression in both species, particularly in O. sarsii vadicola (Additional file 2: Table S21). These core markers are conserved across diverse biological processes, including eukaryotic embryo development [72], post-injury neuroregeneration [73], and exhibiting significant heterogeneity across different tissues [74]. Notably, RPL44 (evm.TU.CTG.1969.9) is specifically expressed in mammalian skeletal muscle stem cells [75], highlighting the importance of RP genes in regeneration across both invertebrates and vertebrates. Despite the distant evolutionary relationship between axolotls and ophiuroids, we identified these conserved RP genes as potential key markers for CT cells during regeneration across species, though future experiments are needed to verify their functions.
Conclusions
This study establishes a chromosome-level genome assembly of Ophiura sarsii vadicola and elucidates the cellular and molecular dynamics of arm regeneration in ophiuroids through combined bulk and single-cell transcriptomic profiling. We demonstrate a hierarchical regeneration mechanism characterized by five distinct cell clusters, highlighting the multipotent differentiation of connective tissue-derived progenitor cells into four other lineages. Notably, muscle differentiation emerges as an early-phase event in regeneration, suggesting temporal prioritization of functional tissue reconstruction. These results provide fundamental genomic and cellular insights into echinoderm regeneration and establish an evolutionary framework for understanding marine invertebrate tissue restoration. The high-quality genomic resource and single-cell atlas herein presented offer a benchmark for comparative studies in regenerative biology across metazoans.
Methods
Sample collection
Ophiura sarsii vadicola specimens used in this study were collected in April 2019 from a cold-water mass (35 m water depth, 35.00 °N and 124.00 °E) in the South Yellow Sea using a benthic trawl. The specimens were maintained in a circulating water system for cultivation. For genome sequencing, arm tissues were immediately preserved in liquid nitrogen and stored at −80 °C. For transcriptome sequencing, specimens were preserved in RNA preservation solution (Tiangen, China).
Genome sequencing and assembly
High-quality DNA was extracted from an O. sarsii vadicola specimen using the cetyl trimethylammonium bromide (CTAB) method. DNA quality and quantity were examined using 1% agarose gel electrophoresis, Qubit 4 instrument (Thermo Fisher Scientific, USA), 2100 Bioanalyzer instrument (Agilent, USA), and qPCR. Multiple libraries were then generated for genome sequencing (Additional file 2: Table S1).
For PacBio sequencing, a SMRTbell library was prepared following the manufacturer’s recommended protocol (Pacific Biosciences, Palo Alto, USA). DNA was randomly fragmented into 20 kbp segments using a Covaris ultrasonicator, followed by enrichment and purification of large DNA fragments with magnetic beads. The fragmented DNA underwent damage and end repair, followed by ligation of stem-loop adapters to both ends. Failed ligations were removed by exonuclease treatment. The library was sequenced on the PacBio Sequel II platform at the Novogene sequencing center (Tianjin, China). For Illumina sequencing, 350 bp DNA libraries were constructed and validated for 150 bp paired-end (PE) sequencing on the HiSeq2000 platform at Novogene. The Illumina reads were trimmed with Trimmomatic v0.33 [76]. Draft genome assembly from short fragments was performed using Jellyfish v2.0 [77] and SOAPdenovo2 [78] with a K-mer size of 17.
PacBio sequencing generated 143.7 Gbp of data with 114.47× coverage and an N50 length of 20,943 bp. Single-pass long reads were preassembled before genome assembly. The longest reads were then selected as seeds and all reads were aligned using DALIGNER [79, 80]. Consensus sequences were generated using the LASort and LAMerge functions of the DALIGNER and pbdacgon programs (https://github.com/pb-cdunn/pbdagcon). The pre-assembled long-read data were assembled into contigs with FALCON v0.3 (https://github.com/PacificBiosciences/falcon3) with the Overlap-Layout-Consensus algorithm. Contigs were polished with Quiver and aligned to 218.32 Gbp of Illumina data (173.92×) using the BWA v0.6 [81]. The final genome assembly was refined using Pilon v.1.24 [82].
For RNA sequencing and genome annotation, total RNA was extracted from skin, arm muscle, podium, and gonad tissues using an animal tissue total RNA extraction kit (Tiangen, China). RNA quality was verified using an Agilent qPCR system. Qualified RNA samples underwent polyA tails enrichment using magnetic beads, followed by fragmentation with NEB Fragmentation Buffer. Fragmented mRNA was converted to Illumina RNA-seq libraries using the NEBNext® Ultra™ RNA Library Prep Kit according to the manufacturer’s instructions. Library quantification was performed using Qubit and fragment length assessment was conducted using a 2100 Bioanalyzer (Agilent, USA). Libraries were sequenced on the Illumina HiSeq2000 platform using the paired-end PE150 strategy at Novogene.
HI-C sequencing, chromosome-level assembly, and quality control
For HI-C analysis, genomic DNA was extracted from an O. sarsii vadicola arm following previously described methods [83]. The HI-C libraries were sequenced on the Illumina HiSeq platform at Novogene, generating 309 Gbp of high-quality data. The HI-C reads were then aligned to the assembled genome using BWA and repetitive and unpaired reads were filtered using Samtools [84]. Reads adjacent to enzyme cut sites were selected to assist genome assembly. The order and orientation of chromosomes were determined (Additional file 2: Table S3) based on contig interaction patterns and locations using Lachesis v180922 [85]. The potential genome assembly contamination was identified using FCS-GX [86] and eliminated.
Genome assembly quality was evaluated through multiple approaches. Illumina reads were aligned to the assembly using BWA. Genome completeness was assessed using two methods: the Core Eukaryotic Genes Mapping Approach (CEGMA) [87], which analyzes 248 conserved genes from six model eukaryotic organisms, and the Benchmarking Universal Single-Copy Orthologs (BUSCO) method [88]. Assembly quality value (QV) was determined using Merqury [89].
Repeat gene annotation, gene prediction, and functional annotation
Repeats in the assembly were identified using both homology alignment and a de novo approach. Tandem repeats were extracted with the Tandem Repeat Finder (TRF) program via ab initio prediction. Homolog prediction was conducted using RepeatMasker v2.10.0 (https://www.repeatmasker.org/) and associated scripts (e.g., RepeatProteinMask) to extract repeat regions using default parameters. Ab initio prediction generated a de novo repetitive element database using LTR_FINDER v1.0.7 [90], RepeatScout [91], and RepeatModeler (https://www.repeatmasker.org/RepeatModeler/) with default parameters. All repeat sequences with lengths > 100 bp and gaps of “N”s making up less than 5% were retained in the raw transposable element (TE) library. DNA-level repeat identification used RepeatMasker with both Repbase (https://www.girinst.org/repbase) and our de novo TE library.
Protein-coding genes were predicted (Additional file 2: Table S6) using an integrated pipeline incorporating Augustus v3.2.3 [92], Geneid v1.4 (https://genome.crg.cat/software/geneid/), Genescan v1.0 (https://hollywood.mit.edu/GENSCAN.html), GlimmerHMM v3.04 (https://ccb.jhu.edu/software/glimmerhmm/), and SNAP v2013-11–29 [93]. Homologous proteins from Ensembl (https://uswest.ensembl.org/index.html) and NCBI (https://www.ncbi.nlm.nih.gov/protein) databases were aligned to the genome using tblastn v2.2.26 (E value ≤ 1e−5), followed by generation of spliced alignments using GeneWise v2.4.1 (https://www.ebi.ac.uk/Tools/psa/genewise/). RNA-seq reads were aligned using Hisat2 v2.0.4 [94] and assembled into genome-based transcripts using Cufflinks v2 [95], both with default parameters.
Gene function annotation involved multiple approaches. We performed BLASTp search against the Swiss-Prot database (https://www.uniprot.org/) with an E value of ≤1e−5. InterProScan v5.31 program (https://github.com/ebi-pf-team/interproscan) was used to annotate motifs and domains through comparison with multiple databases: ProDom (https://prodom.prabi.fr/prodom/current/html/home.php), PRINTS (https://130.88.97.239/PRINTS/index.php), Pfam (https://pfam.xfam.org/), SMART (https://smart.embl-heidelberg.de/), PANTHER (https://www.pantherdb.org/), and PROSITE (https://prosite.expasy.org/). Gene Ontology (GO) IDs were assigned from InterPro entries (https://www.ebi.ac.uk/interpro/). Additional protein functions were annotated based on the top BLAST hits in the nr database (E values of <10−5) and the Kyoto Encyclopedia of Genes and Genomes (KEGG) database. tRNAs were identified with tRNAscan-SE v2.0 [96]. rRNA identification used BLAST searches against sequences from related species, given their high conservation. Other non-functional RNAs, including miRNAs and snRNAs, were identified using the Rfam database and INFERNAL v1.1.5 [97].
Regeneration culture experiments and tissue sampling over time
Complete and healthy O. sarsii vadicola individuals were acclimated in laboratory conditions for 1 month before experimentation. Ten individuals were housed in each of five 20-L tanks maintained at 12 °C and salinity of 31 psu. Following acclimation, two arm tips per individual were amputated at 13, 7, 3, and 1 days before tissue collection. After amputation, ophiuroids were returned to their respective tanks. Regeneration tissue samples (~1 cm) were collected from the wound edge toward the central disk using sterilized scissors and processed for multiple analyses: 1) for scRNA-seq, tissues from three individuals per time point were enzymatically digested; 2) for bulk RNA sequencing, regenerative distal tissues for three individuals per tank were flash-frozen in liquid nitrogen and preserved at −80 °C; 3) for histology and in situ hybridization, the remaining samples were fixed overnight at 4 °C in 4% PFA prepared in 1× PBT reagent (phosphate-buffered saline with 0.1% Tween-20), washed in 1× PBT, and stored in 100% methanol at −20 °C.
Single-cell RNA sequencing and bulk RNA sequencing
To obtain sufficient live cells for scRNA-seq analysis, regenerative tissues from four individuals were pooled per time point, sampling at 13, 7, 3, 1, and 0 days post-amputation.
Single-cell capture was performed using a Chromium Controller Instrument (10× Genomics; Pleasanton, CA). A 1-cm section of arm tissue was excised and immediately digested in 0.1 mg/mL collagenase 1 (Invitrogen, USA) for 30 min at 37 °C. Digestion was stopped using DMEM/F12 with 15% FBS. Dissociated cells were collected by centrifugation (1300 rpm, 4 °C, 5 min). The cell suspension was filtered through a 40-μm filter (Greiner, Germany) and resuspended in PBS with 1% bovine serum albumin (BSA). Cells were then collected by centrifugation at 4 °C. After centrifugation, cell count and concentration were determined using a Countess II Automated Cell Counter and adjusted to 1000 cells/μL before library preparation.
According to the manufacturer’s instructions, cell suspensions were processed using a Chromium Controller Instrument (10× Genomics, Pleasanton, CA, USA). Single-cell gel bead emulsions (GEMs) were generated using the Single Cell 3’ library and Gel Bead Kit v2 (10× Genomics, 120237). Full-length barcode cDNAs were then PCR-amplified. The final Single Cell 3’ library comprised P5 and P7 primers for PCR amplification. Barcoded sequencing libraries were quantified using standard curve qPCR and Agilent Bioanalyzer 2100. Finally, sequencing was performed on a NovaSeq 6000 platform. The library preparation and sequencing were conducted at the Gene Denovo sequencing center (Guangzhou, China). Bulk RNA sequencing of regenerated tissues followed the same RNA extraction, library preparation, and sequencing protocols described in the genome assembly section above and was conducted at Novogene.
Divergence time analysis
We analyzed genomic data from nine echinoderm species (Additional file 2: Table S9), including two ophiuroids [34, 35], to investigate the phylogenetic relationship of O. sarsii vadicola. After filtering genomic repeats using TBtools [98], we extracted representative CDS and peptide sequences from the filtered annotations. Using the OrthoFinder v2.3.14 [99], we clustered the peptide sequences into ortholog groups. A total of 1537 single-copy genes were selected for phylogenetic relationship reconstruction. We identified the optimal substitution model, LG+F+R7, using ModelFinder [100]. We then performed maximum likelihood (ML) analysis using IQ-TREE v1.6.12 [101] with 1000 bootstrap replicates. For molecular dating, we used a fossil calibration point between Apostichopus japonicus and Holothuria leucospilota (CI: 308.8–157.8 Mya, median: 233 Mya, adjusted time: 250 Mya) obtained from TIMETREE5 [102,103,104]. Using the ML tree topology and fossil calibrations, we estimated divergence times with the MCMCtree program embedded in PAML [105]. The analysis employed an independent rates clock model with the root set at the deuterostome-protostome split (RootAge < 700 Mya). We used LG.dat as the amino acid substitution rate file and specified the following MCMC parameters: burnin = 2500, sampfreq = 100, and nsample = 10,000.
Comparative genomic analyses
From the OrthoFinder results, we selected 27,305 gene families that contained genes present in at least 50% of the analyzed species. Using CAFÉ v4 [106], we analyzed these orthogroups to estimate gene family expansions and contractions based on our divergence time in the phylogenetic tree. We then annotated rapidly evolved gene families (p < 0.05) using eggNOG-mapper2 and assessed enrichment using ClusterProfiler v4 [107]. To identify core and ophiuroid-specific gene families, we analyzed orthogroups from O. sarsii vadicola, O. sarsii, and A. filiformis using OrthoVenn2 [108]. These orthogroups were functionally annotated using eggNOG-mapper2 [109] and GO enrichment analysis was performed using clusterProfiler v4 [107]. For synteny analysis between O. sarsii vadicola and the two closely related ophiuroids, we performed reciprocal BLAST searches (E value < 1e−10) using complete protein sequences to identify putative orthologs and visualize collinearity among the three species.
Bulk RNA sequence analysis
Raw reads for the bulk RNA sequence analysis were pre-processed and filtered using Trimmomatic v0.36 [76], yielding the clean read dataset. Clean reads were then aligned to the O. sarsii vadicola reference genome using Hisat2 v2.0.5 [94]. The alignment results were then compressed and sorted using Samtools (Additional file 2: Table S14), and expression quantification was performed using the FeatureCounts program to obtain read count data. The fragments per kilobase of exon model per million mapped fragments (FPKM) value was then calculated for each gene.
Differential expression analysis was conducted using DESeq2 v1.34.0 [110] based on the read count matrix to determine differentially expressed genes between time points. Thresholds including a p value ≤ 0.01 and |log2 (fold change)| ≥ 1 were used to identify genes that were significantly differentially expressed. Inter-sample correlation analysis (correlation) was performed using the “Cor” function in R (v4.0.0) and correlations were plotted using the pheatmap package (https://CRAN.R-project.org/package=pheatmap), hcluster for sample clustering, and PCAtools (https://github.com/kevinblighe/PCAtools) for principal component analysis. The expression of key functional genes in the early stages of wound healing and regeneration was analyzed alongside annotations of significantly differentially expressed genes to assess their roles and expression trends in the process. These results were visualized using the EnhancedVolcano v1.12.0 package (https://github.com/kevinblighe/EnhancedVolcano). KEGG enrichment analysis of gene function was performed using significantly differentially expressed genes (p value ≤ 0.01 and |log2FC| ≥ 1) across different time points with ClusterProfiler v4.0.2.
scRNA-seq data analysis
We used Cell Ranger v3.1.0 (https://support.10xgenomics.com/single-cell-gene-expression/software/pipelines/latest/installation) for initial data processing, including FASTQ file generation (mkfastq function), read quantification (count function), and data aggregation (aggr function) to generate gene expression matrices for individual cells. Using the Seurat v4.0.1 [111], we performed cell filtering based on the O. sarsii vadicola genome, followed by subsequent data standardization.
For the five samples, we filtered cells using the following criteria: gene count per cell (>3000), UMI count per cell (>10,000), and mitochondrial gene percentage (>20%). We standardized the data using SCTransform and performed dimensionality reduction through principal component analysis (PCA). Cell clustering was performed using the first 30 PCA components with the shared-nearest neighbor (SNN) method at an optimal resolution parameter of 0.4. We visualized dimensionality reduction results using Clustree [112] for initial exploratory analysis. To determine the optimal SNN resolution (0.4), we conducted merged contour analysis for each time point, balancing expected cluster numbers based on known marker expression with the largest average contour width. We initiated clustering at a resolution of 0.03 and employed t-SNE for visualization. For differential expression analysis, we used the “FindAllMarkers” function with a likelihood ratio test. We identified differentially expressed genes using three criteria (1) p value ≤ 0.01; (2) log2 (fold change [FC]) ≥ 0.25; and (3) gene detection in >25% of cells within a specific cluster. We then performed GO [113, 114] and KEGG pathway [115] enrichment analyses to characterize the primary functions of each cluster. To further elucidate the functions of significantly differentially expressed genes and assist in cell type identification, we consulted multiple databases: Universal Protein (UniProt, https://www.uniprot.org/), Single Cell Expression Atlas (https://www.ebi.ac.uk/), The Human Protein Atlas (https://www.proteinatlas.org/), and Echinobase Home (https://www.echinobase.org/echinobase/).
We analyzed single-cell trajectories and gene expression patterns of CT II using the Monocle v2.8.0 [116]. This analysis reduces dimensional space to one or two dimensions for cell sorting, revealing tree-like structures with distinct branches and tips. We identified divergently expressed genes using a false discovery rate (FDR) threshold of <1e−5. For pseudotime analysis, we used Slingshot v2.2.0 [117] with default parameters. Slingshot constructed a minimum spanning tree (MST) based on cellular subpopulations to estimate global lineage structure. Differentiation trajectories were fitted using simultaneous principal curves and orthogonal projection methods, generating smooth differentiation trajectories with corresponding pseudotime values.
For cross-species comparisons between axolotls and ophiuroids regeneration capabilities, we analyzed the axolotl limb bud tissue dataset [71]. We first filtered the data using Seurat v4.3.0, retaining cells with UMI counts between 1000 and 25,000, resulting in 4113 normalized cells. We identified highly variable genes for PCA using 50 principal components (number of principal components, npcs). Using the PCA results, we performed cell neighbor identification and clustering at a resolution of 0.6. Cell type annotation of CT was based on marker gene expression in these clusters, followed by repeated dimensionality reduction and clustering. The core marker genes were identified using two criteria: adjusted p values < 0.05 and log fold change > 0.25. For dataset integration between the axolotl and O. sarsii vadicola, we first conducted homologous protein sequences alignment using BLASTp v.2.2.28+. We retained 11,608 one-to-one homologous gene pairs present in the single-cell data, identifying 8984 gene pairs. Using these pairs, we conducted canonical correlation analysis (CCA) to integrate axolotl CT cells with ophiuroid CT I and CT II cells to remove batch effects. Finally, we visualized gene expression patterns between species using Uniform Manifold Approximation and Projection (UMAP).
Connective tissue and immunohistology observations
We analyzed paraffin-embedded connective tissue samples from each regeneration stage using Masson’s trichrome staining and immunohistology (IHC). We sectioned the embedded tissues to 10-μm thickness. For Masson’s trichrome staining, we used a commercial kit (Solarbio, Beijing, China) following the manufacturer’s protocols to differentiate connective tissue fibers (blue) from muscle fibers (red). IHC was performed with a standard protocol (ZSGB-BIO, OriGene Technologies, Inc., China). After deparaffinization and antigen retrieval, we blocked endogenous peroxidase activity using 3% hydrogen peroxide. For antibody blocking, we used normal rabbit serum for goat-derived antibodies and BSA for antibodies from other sources. For antibody treatment, we incubated sections with the primary antibody overnight at 4 °C. We then washed the sections in PBS (pH = 7.4) with agitation. We incubated the sections with HRP-labeled secondary antibody specific to Ophiura sarsii vadicola at room temperature for 50 min.
For visualization, we applied DAB color-developing solution and monitored color development microscopically, with brownish-yellow indicating positive signals.
We stopped the reaction with tap water and proceeded with counterstaining. The sections were counterstained with hematoxylin for 3 min, followed by treatment with hematoxylin differentiation solution and then the bluing solution, with running water washes between steps.
The final dehydration process involved sequential treatment with increasing concentrations of ethanol (75%, 85%, 100%), followed by anhydrous ethanol, n-butanol, and xylene for 5 min each. After brief air-drying, we mounted the sections with neutral gum. We visualized and photographed the stained sections with a Nikon Eclipse Ci microscope. Successfully stained sections showed blue nuclei from hematoxylin staining and brownish-yellow signals indicating a positive DAB reaction.
In situ hybridization
We performed in situ hybridization on arm regeneration sections using DIG-labeled antisense probes for Lrp6 and fibrosurfin (Additional file 2: Table S22), following a published protocol [118]. We first rehydrated sections through graded ethanol (70%, 50%, and 30%) and washed them three times in 1× MABT buffer (0.1 M maleic acid [pH = 7.5], 0.5 M NaCl, 0.1% Tween-20). After proteinase K digestion for 5 min, we stopped the reaction with 4% paraformaldehyde in 1× PBS for 20 min, followed by 5-min washes in 1× PBT (1× PBS and 0.1% Tween-20). The sections were pre-treated with HB buffer (50% deionized formamide, 10% PEG, 0.05 M NaCl, 0.1% Tween-20, 0.005 M EDTA, 0.02 M yeast Tris pH 7.5, 0.1 mg/ml yeast tRNA, 1× Denhardt’s solution, and DEPC-treated water) at 50 °C for 1 h. Sections were then incubated with HB containing 0.2 ng/μL antisense probe for 8 days at 50 °C, followed by further incubation in fresh HB without probe for 3 h at 50 °C, and then washed once in MABT at 50 °C and once at room temperature. We then performed three washes in 0.1× MABT and one in 1× MABT before overnight incubation in anti-DIG AP antibody (1:1000) at 4 °C. After five MABT washes and two alkaline phosphatase buffer washes, we performed chromogenic detection for 3–4 h. The reaction was stopped with 1× MABT containing 0.05 M EDTA, followed by three MABT washes. Specimens were stored in 50% glycerol at 4 °C and imaged using an Olympus SZX16 microscope (Olympus Corporation, Japan).
Data availability
The data of genome sequencing and the genome assembly data of Ophiura sarsii vadicola in this study have been deposited at NCBI under the BioProject PRJNA732836 [119]. The bulk RNA sequencing data and single-cell RNA sequencing data have been deposited at NCBI under BioProject PRJNA942107 [120]. Other echinoderm genomes applied in comparative analyses are listed in Additional File 2: Table S4. The cell expression information, bulk RNA expression data, histology photos relevant to arm regeneration and the script for across-species analysis used in this study, along with two additional files, are available in FigShare [121].
References
Zullo L, Bozzo M, Daya A, Di Clemente A, Mancini FP, Megighian A, Nesher N, Röttinger E, Shomrat T, Tiozzo S, Zullo A, Candiani S. The diversity of muscles and their regenerative potential across animals. Cells. 2020;9:1925.
Bideau L, Kerner P, Hui J, Vervoort M, Gazave E. Animal regeneration in the era of transcriptomics. Cell Mol Life Sci. 2021;78:3941–56.
Byrne M. The link between autotomy and CNS regeneration: echinoderms as non-model species for regenerative biology. BioEssays. 2020;42:e1900219.
Sköld M, Rosenberg R. Arm regeneration frequency in eight species of Ophiuroidea (Echinodermata) from European sea areas. J Sea Res. 1996;35:353–62.
Byrne M, Mazzone F, Elphick MR, Thorndyke MC, Cisternas P. Expression of the neuropeptide SALMFamide-1 during regeneration of the seastar radial nerve cord following arm autotomy. Proc Royal Society B: Biological Sciences. 2019;286:20182701.
Okanishi M, Kohtsuka H, Miura T. Morphogenesis and histogenesis during the arm regeneration in a basket star Astrocladus dofleini (Euryalida, Ophiuroidea, Echinodermata). J Morphol. 2021;282:205–16.
Medina-Feliciano JG, Pirro S, García-Arrarás JE, Mashanov V, Ryan JF. Draft genome of the sea cucumber Holothuria glaberrima, a model for the study of regeneration. Front Mar Sci. 2021;8:603410.
Murray G, García-Arrarás JE. Myogenesis during holothurian intestinal regeneration. Cell Tissue Res. 2004;318:515–24.
Dolmatov IY. Variability of regeneration mechanisms in echinoderms. Russ J Mar Biol. 2020;46:391–404.
García-Arrarás JE, Dolmatov IY. Echinoderms: potential model systems for studies on muscle regeneration. Curr Pharm Des. 2010;16:942–55.
Ben Khadra Y, Sugni M, Ferrario C, Bonasoro F, Oliveri P, Martinez P, Candia Carnevali MD. Regeneration in stellate echinoderms: Crinoidea, Asteroidea and Ophiuroidea. Marine Organ Model Syst Biol Med. 2018;65:285–320.
Stöhr S, O’Hara TD, Thuy B. Global diversity of brittle stars (Echinodermata: Ophiuroidea). PLoS One. 2012;7:e31940.
Clark MS, Souster T. Slow arm regeneration in the Antarctic brittle star Ophiura crassa (Echinodermata, Ophiuroidea). Aquat Biol. 2012;16:105–13.
Biressi ACM, Zou T, Dupont S, Dahlberg C, Di Benedetto C, Bonasoro F, Thorndyke M, Carnevali MDC. Wound healing and arm regeneration in Ophioderma longicaudum and Amphiura filiformis (Ophiuroidea, Echinodermata): comparative morphogenesis and histogenesis. Zoomorphology. 2010;129:1–19.
Purushothaman S, Saxena S, Meghah V, Swamy CVB, Ortega-Martinez O, Dupont S, Idris M. Transcriptomic and proteomic analyses of Amphiura filiformis arm tissue-undergoing regeneration. J Proteomics. 2015;112:113–24.
Mashanov V, Machado DJ, Reid R, Brouwer C, Kofsky J, Janies DA. Twinkle twinkle brittle star: the draft genome of Ophioderma brevispinum (Echinodermata: Ophiuroidea) as a resource for regeneration research. BMC Genomics. 2022;23:1–17.
Martynov A. Reassessment of the classification of the Ophiuroidea (Echinodermata), based on morphological characters. I. General character evaluation and delineation of the families Ophiomyxidae and Ophiacanthidae. Zootaxa. 2010;2697:1–154.
Goharimanesh M, Ghassemzadeh F, De Kegel B, Van Hoorebeke L, Stöhr S, Mirshamsi O, Adriaens D. The evolutionary relationship between arm vertebrae shape and ecological lifestyle in brittle stars (Echinodermata: Ophiuroidea). J Anat. 2021;240:1034–47.
Mashanov V, Akiona J, Khoury M, Ferrier J, Reid R, Machado DJ, Zueva O, Janies D. Active Notch signaling is required for arm regeneration in a brittle star. PLoS One. 2020;15:e0232981.
Czarkwiani A, Ferrario C, Dylus DV, Sugni M, Oliveri P. Skeletal regeneration in the brittle star Amphiura filiformis. Front Zool. 2016;13:18.
Burns G. Gene expression and regeneration in ophiuroids: a study of transcriptional activity and regeneration in the temperate Amphiura filiformis and Antarctic Ophionotus victoriae. PhD thesis, University of Gothenburg, Department of Biological and Environmental Sciences. 2012.
Burns G, Thorndyke MC, Peck LS, Clark MS. Transcriptome pyrosequencing of the Antarctic brittle star Ophionotus victoriae. Mar Genomics. 2013;9:9–15.
Dylus DV, Czarkwiani A, Blowes LM, Elphick MR, Oliveri P. Developmental transcriptomics of the brittle star Amphiura filiformis reveals gene regulatory network rewiring in echinoderm larval skeleton evolution. Genome Biol. 2018;19:1–17.
Czarkwiani A, Dylus DV, Carballo L, Oliveri P. FGF signalling plays similar roles in development and regeneration of the skeleton in the brittle star Amphiura filiformis. Development. 2021;148:dev180760.
Pond KW, Doubrovinski K, Thorne CA. Wnt/β-catenin signaling in tissue self-organization. Genes. 2020;11:939.
Lu W, Fuchou T. Recent progress in single-cell RNA-seq analysis. Yi Chuan. 2014;36:1069–76.
Mayr U, Serra D, Liberali P. Exploring single cells in space and time during tissue development, homeostasis and regeneration. Development. 2019;146:dev176727.
Greaney AM, Adams TS, Brickman Raredon MS, Gubbins E, Schupp JC, Engler AJ, Ghaedi M, Yuan Y, Kaminski N, Niklason LE. Platform effects on regeneration by pulmonary basal cells as evaluated by single-cell RNA sequencing. Cell Reports. 2020;30:4250–4265.e4256.
Shao Y, Wang X-B, Zhang J-J, Li M-L, Wu S-S, Ma X-Y, Wang X, Zhao H-F, Li Y, Zhu HH, Irwin DM, Wang D-P, Zhang G-J, Ruan J, Wu D-D. Genome and single-cell RNA-sequencing of the earthworm Eisenia andrei identifies cellular mechanisms underlying regeneration. Nat Commun. 2020;11:2656.
Bodó K, Kellermayer Z, László Z, Boros Á, Kokhanyuk B, Németh P, Engelmann P. Injury-induced innate immune response during segment regeneration of the earthworm, Eisenia andrei. Int J Mol Sci. 2021;22:2363.
De Micheli AJ, Laurilliard EJ, Heinke CL, Ravichandran H, Fraczek P, Soueid-Baumgarten S, De Vlaminck I, Elemento O, Cosgrove BD. Single-cell analysis of the muscle stem cell hierarchy identifies heterotypic communication signals involved in skeletal muscle regeneration. Cell Rep. 2020;30:3583–95.
Aztekin C, Hiscock TW, Marioni JC, Gurdon JB, Simons BD, Jullien J. Identification of a regeneration-organizing cell in the Xenopus tail. Science. 2019;364:653–8.
Gerber T, Murawala P, Knapp D, Masselink W, Schuez M, Hermann S, Gac-Santel M, Nowoshilow S, Kageyama J, Khattak S, Currie Joshua D, Camp JG, Tanaka Elly M, Treutlein B. Single-cell analysis uncovers convergence of cell identities during axolotl limb regeneration. Science. 2018;362:eaaq0681.
Han C, Zhang Q, Li Y, Sun Y, Dong Y, Ge M, Li Z, Hu X, Liu B, Zhang X. Chromosome-level genome assembly and annotation of the cold-water species Ophiura sarsii. Scientific Data. 2024;11:560.
Parey E, Ortega-Martinez O, Delroisse J, Piovani L, Czarkwiani A, Dylus D, Arya S, Dupont S, Thorndyke M, Larsson T. The brittle star genome illuminates the genetic basis of animal appendage regeneration. Nature Ecology & Evolution. 2024;8:1505–21.
Li Y, Dong Y, Xu Q, Fan S, Lin H, Wang M, Zhang X. Genetic differentiation and evolutionary history of the circumpolar species Ophiura sarsii and subspecies Ophiura sarsii vadicola (Ophiurida: Ophiuridae). Cont Shelf Res. 2020;197:104085.
Xu Y, Ma L, Sui J, Li X, Wang H, Zhang B. Potential effects of climate change on the habitat suitability of macrobenthos in the Yellow Sea and East China Sea. Mar Pollut Bull. 2022;174:113238.
Bai SW, Herrera-Abreu MT, Rohn JL, Racine V, Tajadura V, Suryavanshi N, Bechtel S, Wiemann S, Baum B, Ridley AJ. Identification and characterization of a set of conserved and new regulators of cytoskeletal organization, cell morphology and migration. BMC Biol. 2011;9:54.
Drazic A, Aksnes H, Marie M, Boczkowska M, Varland S, Timmerman E, Foyn H, Glomnes N, Rebowski G, Impens F, Gevaert K, Dominguez R, Arnesen T. NAA80 is actin’s N-terminal acetyltransferase and regulates cytoskeleton assembly and cell motility. Proc Natl Acad Sci. 2018;115:4399.
Pascual-Carreras E, Sureda-Gómez M, Barrull-Mascaró R, Jordà N, Gelabert M, Coronel-Córdoba P, Saló E, Adell T. WNT-FRIZZLED-LRP5/6 signaling mediates posterior fate and proliferation during planarian regeneration. Genes. 2021;12:101.
Chen M, Philipp M, Wang J, Premont RT, Garrison TR, Caron MG, Lefkowitz RJ, Chen W. G protein-coupled receptor kinases phosphorylate LRP6 in the Wnt pathway. J Biol Chem. 2009;284:35040–8.
Ross J, Busch J, Mintz E, Ng D, Stanley A, Brafman D, Sutton VR, Van den Veyver I, Willert K. A rare human syndrome provides genetic evidence that WNT signaling is required for reprogramming of fibroblasts to induced pluripotent stem cells. Cell Rep. 2014;9:1770–80.
Reinardy HC, Emerson CE, Manley JM, Bodnar AG. Tissue regeneration and biomineralization in sea urchins: role of Notch signaling and presence of stem cell markers. PLoS One. 2015;10:e0133860.
Cluzel C, Lethias C, Humbert F, Garrone R, Exposito JY. Characterization of fibrosurfin, an interfibrillar component of sea urchin catch connective tissues. J Biol Chem. 2001;276:18108–14.
Bu HF, Zuo XL, Wang X, Ensslin M, Koti V, Hsueh W, Raymond A, Shur B, Di X. Milk fat globule-EGF factor 8/lactadherin plays a crucial role in maintenance and repair of murine intestinal epithelium. The Journal of Clinical Investigation. 2008;117:3673–83.
Guaiquil V, Swendeman S, Yoshida T, Chavala S, Campochiaro Peter A, Blobel Carl P. ADAM9 is involved in pathological retinal neovascularization. Mol Cell Biol. 2009;29:2694–703.
Berdeaux R, Stewart R. cAMP signaling in skeletal muscle adaptation: hypertrophy, metabolism, and regeneration. American Journal of Physiology-Endocrinology and Metabolism. 2012;303:E1–13.
Li S, Yang C, Zhang L, Gao X, Wang X, Liu W, Wang Y, Jiang S, Wong YH, Zhang Y. Promoting axon regeneration in the adult CNS by modulation of the melanopsin/GPCR signaling. Proc Natl Acad Sci. 2016;113:1937–42.
Itaki T. Transitional changes in microfossil assemblages in the Japan Sea from the Late Pliocene to Early Pleistocene related to global climatic and local tectonic events. Prog Earth Planet Sci. 2016;3:1–21.
Lewin TD, Liao IJY, Luo YJ. Annelid comparative genomics and the evolution of massive lineage-specific genome rearrangement in bilaterians. Mol Biol Evol. 2024;41:msae172.
Piovani L, Czarkwiani A, Ferrario C, Sugni M, Oliveri P. Ultrastructural and molecular analysis of the origin and differentiation of cells mediating brittle star skeletal regeneration. BMC Biol. 2021;19:9.
Ferrario C, Czarkwiani A, Dylus DV, Piovani L, Candia Carnevali MD, Sugni M, Oliveri P. Extracellular matrix gene expression during arm regeneration in Amphiura filiformis. Cell Tissue Res. 2020;381:411–26.
Clark MS, Dupont S, Rosetti H, Burns G, Thorndyke MC, Peck LS. Delayed arm regeneration in the Antarctic brittle star Ophionotus victoriae. Aquat Biol. 2007;1:45–53.
Wilkie I. Mutable collagenous tissue: overview and biotechnological perspective. Echinodermata. 2005;39:221–50.
Wilkie IC. Mutable collagenous tissue: overview and biotechnological perspective. In: Matranga V, editor. Echinodermata. Berlin, Heidelberg: Springer Berlin Heidelberg; 2005. p. 221-250.
Shi Y, Su J, Roberts AI, Shou P, Rabson AB, Ren G. How mesenchymal stem cells interact with tissue immune responses. Trends Immunol. 2012;33:136–43.
Li P, Ou Q, Shi S, Shao C. Immunomodulatory properties of mesenchymal stem cells/dental stem cells and their therapeutic applications. Cell Mol Immunol. 2023;20:558–69.
Chen S-E, Jin B, Li Y-P. TNF-α regulates myogenesis and muscle regeneration by activating p38 MAPK. Am J Physiol Cell Physiol. 2007;292:C1660–71.
Haenold R, Weih F, Herrmann K-H, Schmidt K-F, Krempler K, Engelmann C, Nave K-A, Reichenbach JR, Löwel S, Witte OW. NF-κB controls axonal regeneration and degeneration through cell-specific balance of RelA and p50 in the adult CNS. J Cell Sci. 2014;127:3052–65.
Candia Carnevali MD, Bonasoro F. Microscopic overview of crinoid regeneration. Microscopy Research and Technique. 2001;55:403–26.
Carnevali MDC, Lucca E, Bonasoro F. Mechanisms of arm regeneration in the feather star Antedon mediterranea: healing of wound and early stages of development. J Exp Zool. 1993;267:299–317.
Ozhan G, Weidinger G. Wnt/β-catenin signaling in heart regeneration. Cell Regeneration. 2015;4:4–3.
Giorgio GD, Rubilar T, Brogger MI. Histological analysis after arm tip amputation in the brittle star Ophioplocus januarii (Echinodermata: Ophiuroidea). Rev Biol Trop. 2015;63:297–308.
Zueva O, Khoury M, Heinzeller T, Mashanova D, Mashanov V. The complex simplicity of the brittle star nervous system. Front Zool. 2018;15:1–26.
Ben Khadra Y, Ferrario C, Benedetto CD, Said K, Bonasoro F, Candia Carnevali MD, Sugni M. Re-growth, morphogenesis, and differentiation during starfish arm regeneration. Wound Repair Regen. 2015;23:623–34.
Dolmatov IY. Molecular aspects of regeneration mechanisms in holothurians. Genes. 2021;12: 250.
Scimone ML, Cote LE, Reddien PW. Orthogonal muscle fibres have different instructive roles in planarian regeneration. Nature. 2017;551:623–8.
Li H, Wei X, Zhou L, Zhang W, Wang C, Guo Y, Li D, Chen J, Liu T, Zhang Y. Dynamic cell transition and immune response landscapes of axolotl limb regeneration revealed by single-cell analysis. Protein Cell. 2021;12:57–66.
Esfahanian V, Golestaneh H, Moghaddas O, Ghafari MR. Efficacy of connective tissue with and without periosteum in regeneration of intrabony defects. Journal of Dental Research, Dental Clinics, Dental Prospects. 2014;8:189.
Murphy MM, Lawson JA, Mathew SJ, Hutcheson DA, Kardon G. Satellite cells, connective tissue fibroblasts and their interactions are crucial for muscle regeneration. Development. 2011;138:3625–37.
Lin TY, Gerber T, Taniguchi-Sugiura Y, Murawala P, Hermann S, Grosser L, Shibata E, Treutlein B, Tanaka EM. Fibroblast dedifferentiation as a determinant of successful regeneration. Dev Cell. 2021;56:1541–1551.e1546.
Chang Z, Wang X, Pan X, Yan W, Wu W, Zhuang Y, Li Z, Wang D, Yuan S, Xu C. The ribosomal protein P0A is required for embryo development in rice. BMC Plant Biol. 2023;23:465.
Nawabi H, Belin S. Translational machinery and translation regulation in axon regeneration. Neural Regen Res. 2025;20:1392–4.
Guimaraes JC, Zavolan M. Patterns of ribosomal protein expression specify normal and malignant human cells. Genome Biol. 2016;17:1–13.
Gabut M, Bourdelais F, Durand S. Ribosome and translational control in stem cells. Cells. 2020;9:497.
Bolger AM, Lohse M, Usadel B. Trimmomatic: a flexible trimmer for Illumina sequence data. Bioinformatics. 2014;30:2114–20.
Marçais G, Kingsford C. A fast, lock-free approach for efficient parallel counting of occurrences of k-mers. Bioinformatics. 2011;27:764–70.
Luo R, Liu B, Xie Y, Li Z, Huang W, Yuan J, He G, Chen Y, Pan Q, Liu Y. SOAPdenovo2: an empirically improved memory-efficient short-read de novo assembler. Gigascience. 2012;1:2047-2217X-2041–2018.
Will S, Stadler PF. Algorithms in bioinformatics: 14th international workshop, WABI 2014, Wroclaw, Poland, September 8-10, 2014. Proceedings. Berlin, Heidelberg, Germany: Springer Berlin Heidelberg; 2014:135–47.
Myers G. Algorithms in bioinformatics. In: 14th international workshop, WABI 2014, Wroclaw, Poland, September 8-10, 2014. Proceedings 14. Springer; 2014. p. 52-67.
H. Li, Aligning sequence reads, clone sequences and assembly contigs with BWA-MEM. ArXiv Preprint ArXiv, (2013).
Walker BJ, Abeel T, Shea T, Priest M, Abouelliel A, Sakthikumar S, Cuomo CA, Zeng Q, Wortman J, Young SK. Pilon: an integrated tool for comprehensive microbial variant detection and genome assembly improvement. PLoS One. 2014;9:e112963.
Belton J-M, McCord RP, Gibcus JH, Naumova N, Zhan Y, Dekker J. Hi–C: a comprehensive technique to capture the conformation of genomes. Methods. 2012;58:268–76.
Danecek P, Bonfield JK, Liddle J, Marshall J, Ohan V, Pollard MO, Whitwham A, Keane T, McCarthy SA, Davies RM. Twelve years of SAMtools and BCFtools. Gigascience. 2021;10:giab008.
Burton JN, Adey A, Patwardhan RP, Qiu R, Kitzman JO, Shendure J. Chromosome-scale scaffolding of de novo genome assemblies based on chromatin interactions. Nat Biotechnol. 2013;31:1119–25.
Astashyn A, Tvedte ES, Sweeney D, Sapojnikov V, Bouk N, Joukov V, Mozes E, Strope PK, Sylla PM, Wagner L. Rapid and sensitive detection of genome contamination at scale with FCS-GX. Genome Biol. 2024;25:60.
Parra G, Bradnam K, Korf I. CEGMA: a pipeline to accurately annotate core genes in eukaryotic genomes. Bioinformatics. 2007;23:1061–7.
Simão FA, Waterhouse RM, Ioannidis P, Kriventseva EV, Zdobnov EM. BUSCO: assessing genome assembly and annotation completeness with single-copy orthologs. Bioinformatics. 2015;31:3210–2.
Rhie A, Walenz BP, Koren S, Phillippy AM. Merqury: reference-free quality, completeness, and phasing assessment for genome assemblies. Genome Biol. 2020;21:1–27.
Xu Z, Wang H. LTR_FINDER: an efficient tool for the prediction of full-length LTR retrotransposons. Nucleic Acids Res. 2007;35:W265–8.
Price AL, Jones NC, Pevzner PA. De novo identification of repeat families in large genomes. Bioinformatics. 2005;21:i351–8.
Stanke M, Diekhans M, Baertsch R, Haussler D. Using native and syntenically mapped cDNA alignments to improve de novo gene finding. Bioinformatics. 2008;24:637–44.
Korf I. Gene finding in novel genomes. BMC Bioinformatics. 2004;5:1–9.
Kim D, Paggi JM, Park C, Bennett C, Salzberg SL. Graph-based genome alignment and genotyping with HISAT2 and HISAT-genotype. Nat Biotechnol. 2019;37:907–15.
Trapnell C, Williams BA, Pertea G, Mortazavi A, Kwan G, Van Baren MJ, Salzberg SL, Wold BJ, Pachter L. Transcript assembly and quantification by RNA-Seq reveals unannotated transcripts and isoform switching during cell differentiation. Nat Biotechnol. 2010;28:511–5.
Chan PP, Lin BY, Mak AJ, Lowe TM. tRNAscan-SE 2.0: improved detection and functional classification of transfer RNA genes. Nucleic Acids Res. 2021;49:9077–96.
Nawrocki EP, Eddy SR. Infernal 1.1: 100-fold faster RNA homology searches. Bioinformatics. 2013;29:2933–5.
Chen C, Chen H, Zhang Y, Thomas HR, Frank MH, He Y, Xia R. TBtools: an integrative toolkit developed for interactive analyses of big biological data. Mol Plant. 2020;13:1194–202.
Emms DM, Kelly S. OrthoFinder: solving fundamental biases in whole genome comparisons dramatically improves orthogroup inference accuracy. Genome Biol. 2015;16:1–14.
Kalyaanamoorthy S, Minh BQ, Wong TK, Von Haeseler A, Jermiin LS. ModelFinder: fast model selection for accurate phylogenetic estimates. Nat Methods. 2017;14:587–9.
Nguyen L-T, Schmidt HA, Von Haeseler A, Minh BQ. IQ-TREE: a fast and effective stochastic algorithm for estimating maximum-likelihood phylogenies. Mol Biol Evol. 2015;32:268–74.
Kumar S, Stecher G, Suleski M, Hedges SB. TimeTree: a resource for timelines, timetrees, and divergence times. Mol Biol Evol. 2017;34:1812–9.
Miller AK, Kerr AM, Paulay G, Reich M, Wilson NG, Carvajal JI, Rouse GW. Molecular phylogeny of extant Holothuroidea (Echinodermata). Mol Phylogenet Evol. 2017;111:110–31.
Delsuc F, Philippe H, Tsagkogeorga G, Simion P, Tilak M-K, Turon X, López-Legentil S, Piette J, Lemaire P, Douzery EJ. A phylogenomic framework and timescale for comparative studies of tunicates. BMC Biol. 2018;16:1–14.
Yang Z. PAML: a program package for phylogenetic analysis by maximum likelihood. Comput Appl Biosci. 1997;13:555–6.
Han MV, Thomas GW, Lugo-Martinez J, Hahn MW. Estimating gene gain and loss rates in the presence of error in genome assembly and annotation using CAFE 3. Mol Biol Evol. 2013;30:1987–97.
Wu T, Hu E, Xu S, Chen M, Guo P, Dai Z, Feng T, Zhou L, Tang W, Zhan L, Fu X. clusterProfiler 4.0: a universal enrichment tool for interpreting omics data. Innovation. 2021;2(3):100141.
Xu L, Dong Z, Fang L, Luo Y, Wei Z, Guo H, Zhang G, Gu YQ, Coleman-Derr D, Xia Q. OrthoVenn2: a web server for whole-genome comparison and annotation of orthologous clusters across multiple species. Nucleic Acids Res. 2019;47:W52–8.
Cantalapiedra CP, Hernández-Plaza A, Letunic I, Bork P, Huerta-Cepas J. eggNOG-mapper v2: functional annotation, orthology assignments, and domain prediction at the metagenomic scale. Mol Biol Evol. 2021;38:5825–9.
Love MI, Huber W, Anders S. Moderated estimation of fold change and dispersion for RNA-seq data with DESeq2. Genome Biol. 2014;15:1–21.
Hao Y, Hao S, Andersen-Nissen E, Mauck WM, Zheng S, Butler A, Lee MJ, Wilk AJ, Darby C, Zager M. Integrated analysis of multimodal single-cell data. Cell. 2021;184:3573–3587. e3529.
Zappia L, Oshlack A. Clustering trees: a visualization for evaluating clusterings at multiple resolutions. Gigascience. 2018;7(7):giy083.
Ashburner M, Ball CA, Blake JA, Botstein D, Butler H, Cherry JM, Davis AP, Dolinski K, Dwight SS, Eppig JT, Harris MA, Hill DP, Issel-Tarver L, Kasarskis A, Lewis S, Matese JC, Richardson JE, Ringwald M, Rubin GM, Sherlock G. Gene Ontology: tool for the unification of biology. Nat Genet. 2000;25:25–9.
T. G. O. Consortium, S. A. Aleksander, J. Balhoff, S. Carbon, J. M. Cherry, H. J. Drabkin, D. Ebert, M. Feuermann, P. Gaudet, N. L. Harris, The Gene Ontology knowledgebase in 2023. Genetics 224, (2023).
Kanehisa M, Goto S. KEGG: Kyoto Encyclopedia of Genes and Genomes. Nucleic Acids Res. 2000;28:27–30.
Trapnell C, Cacchiarelli D, Grimsby J, Pokharel P, Li S, Morse M, Lennon NJ, Livak KJ, Mikkelsen TS, Rinn JL. The dynamics and regulators of cell fate decisions are revealed by pseudotemporal ordering of single cells. Nat Biotechnol. 2014;32:381–6.
Street K, Risso D, Fletcher RB, Das D, Ngai J, Yosef N, Purdom E, Dudoit S. Slingshot: cell lineage and pseudotime inference for single-cell transcriptomics. BMC Genomics. 2018;19:1–16.
Czarkwiani A, Dylus DV, Oliveri P. Expression of skeletogenic genes during arm regeneration in the brittle star Amphiura filiformis. Gene Expr Patterns. 2013;13:464–72.
First Institute of Oceanography, Ministry of Natural Resources, Ophiura sarsii vadicola isolate: osv1 genome sequencing. Datasets. GenBank. https://www.ncbi.nlm.nih.gov/bioproject/?term=PRJNA732836 (2021).
The First Institute of Oceanography, Ophiura sarsii vadicola raw sequence reads. Datasets. GenBank. https://www.ncbi.nlm.nih.gov/bioproject/?term=PRJNA942107 (2023).
Li Y-X, Shi W, Xu Q. Supplementary files: chromosome-level genome assembly and single-cell analysis unveil molecular mechanisms of arm regeneration in the ophiuroid Ophiura sarsii vadicola. Datasets, FigShare. 2025. https://doiorg.publicaciones.saludcastillayleon.es/10.6084/m9.figshare.28468406.
First Institute of Oceanography, Ophiura sarsii genome sequencing and assembly. Datasets. GenBank. 2024. https://www.ncbi.nlm.nih.gov/bioproject/?term=PRJNA1057532.
University College London. Amphiura filiformis isolate: Afil_Kberg_Andreas genome sequencing. Datasets. GenBank. 2023. https://www.ncbi.nlm.nih.gov/bioproject/?term=PRJNA1029566.
Whitney Laboratory for Marine Bioscience, Low coverage sequencing of the brittle star Ophionereis fasciata. Datasets. GenBank. https://www.ncbi.nlm.nih.gov/bioproject/?term=PRJEB10339. (2015).
Sea Urchin Genome Sequencing and Analysis Consortium, Ophiothrix spiculata isolate: Osp01 genome sequencing and assembly. Datasets. GenBank. https://www.ncbi.nlm.nih.gov/bioproject/PRJNA182997 (2015).
Okinawa Institute of Science and Technology, Genome sequence of Acanthaster planci. Datasets. GenBank. https://www.ncbi.nlm.nih.gov/bioproject/275725. (2014).
University of California Conservation Genomics Consortium. Pisaster ochraceus isolate: M0D055189C genome sequencing and assembly. Datasets. GenBank. 2020. https://www.ncbi.nlm.nih.gov/bioproject/?term=PRJNA532896.
Wellcome Sanger Institute, Asterias rubens (common starfish) genome assembly, eAstRub1. Datasets. GenBank. https://www.ncbi.nlm.nih.gov/bioproject/560024 (2019).
Baylor College of Medicine, Strongylocentrotus purpuratus isolate: Spur 01 genome sequencing and assembly. Datasets. GenBank. https://www.ncbi.nlm.nih.gov/bioproject/?term=PRJNA10736 (2005).
H Guo, Genome project of Lytechinus variegatus. Datasets. CNGB. https://db.cngb.org/search/project/CNP0000959/. (2020).
Chinese Academy of Sciences. Holothuria leucospilota isolate: nanhai2018 genome sequencing and assembly. Dataets, GenBank. 2023. https://www.ncbi.nlm.nih.gov/bioproject/PRJNA747844/.
Institute of Oceanology, Chinese Academy of Sciences, Apostichopus japonicus isolate: Shaxun genome sequencing and assembly. Datasets. GenBank. https://www.ncbi.nlm.nih.gov/bioproject/?term=PRJNA354676. (2017).
Center for Ecological and Environmental Sciences. Anneissia japonica RefSeq genome sequencing and assembly. Datasets. GenBank. 2020. https://www.ncbi.nlm.nih.gov/bioproject/?term=PRJNA615663.
Acknowledgements
Thanks to Dr. Igor Yu. Dolmatov (Far Eastern Branch, Russian Academy of Sciences) for helpful comments on this manuscript. Thanks to Glbizzia Biosciences Co., Ltd for their assistance on the across-species analysis.
Peer review information
Wenjing She was the primary editor of this article and managed its editorial process and peer review in collaboration with the rest of the editorial team. The peer review history is available in the online version of this article.
Funding
This work was financially supported by the National Natural Science Foundation of China (42176135) and the National Key R&D Program of China (2022YFF0802202).
Author information
Authors and Affiliations
Contributions
Conceptualization: Q.-Z.X. Supervision: Q.-Z.X. and X.-L.Z. Methodology: Q.-Z.X., Y.-X.L., W.-G.S., Y.D. Z.L. M.P.G., C.H., Q.Z., Y.-Y.S., L.-L.Z., and K.-M.S. Visualization: Y.-X.L. and W.-G.S. Writing - original draft: Q.-Z.X., Y.-X.L. and W.-G.S. Writing - review and editing: Q.-Z.X., Y.-X.L., W.-G.S., J.C.-H.I., M.P.G., C.H., Q.Z., Y.-Y.S., L.-L.Z., K.-M.S., Z.-L.W., J.-W.Q. and X.-L.Z. All authors read, edited, and approved the final manuscript.
Corresponding author
Ethics declarations
Ethics approval and consent to participate
Not applicable. This study involves specimens not belonging to endangered species.
Competing interests
The authors declare no competing interests.
Additional information
Publisher's Note
Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Rights and permissions
Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/.
About this article
Cite this article
Xu, QZ., Li, YX., Shi, WG. et al. Chromosome-level genome assembly and single-cell analysis unveil molecular mechanisms of arm regeneration in the ophiuroid Ophiura sarsii vadicola. Genome Biol 26, 82 (2025). https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s13059-025-03542-5
Received:
Accepted:
Published:
DOI: https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s13059-025-03542-5