Synopsis
The ability to regenerate can greatly vary between animal groups and cell types. Some of the outstanding questions in the field of regeneration include: (1) How has regeneration evolved? and (2) What features underlie differences in regeneration potential between animals? Whether regeneration evolved once and diversified or if it evolved multiple times independently by co-opting similar pathways remains unknown. Current research seeks to identify conserved cellular and molecular features that allow for regeneration. However, comparisons between distantly related regenerating animals have revealed a large amount of diversity. In this perspective, I review discussions on the mechanisms, cell types, and genes underlying regeneration. I propose using Cnidaria as a group in which to investigate the evolution of regeneration. As the sister group to Bilateria with notable regenerative capacity, studies in Cnidaria offer insights into the evolutionary history and conservation of regenerative mechanisms. I then highlight how genome-wide studies, single-cell genomics, multi-omics, and gene editing can be used to identify cell types and unknown features of regeneration. Applying these approaches across organisms will give insight into the cell and molecular features that allow for regeneration competency and may be used to alter an organism’s regeneration potential.
Introduction
Regeneration is the ability to regrow missing body parts. In animals, the extent of regeneration can be variable for different cell types, tissues, and especially between species. As regeneration can require growth and patterning similar to development, many studies focus on a candidate gene approach to identify conserved developmental genes and pathways involved in regeneration. Early fundamental studies using in situ hybridization, chemical inhibitions, and even gene knockdowns identified genes essential for regeneration (Endo et al. 1997; Hobmayer et al. 2000). Yet, whether there is a defined set of competency factors—mechanisms, cell types, genes, and genetic pathways—shared across species remains unknown. Of special interest is identifying the cellular and molecular underpinnings of regeneration and characterizing conserved features of regenerating animals. Still unresolved and important to this topic is determining whether regeneration has evolved once and diversified or whether it has evolved more than once and undergone convergence (Brockes and Kumar 2008; Bely and Nyberg 2010; Tiozzo and Copley 2015; Maden 2018; Elchaninov et al. 2021; Srivastava 2021). Understanding the features of regeneration competency and its evolution will provide insight into mechanisms that may also advance the field of regenerative medicine (Alvarado and Tsonis 2006).
In determining whether the ability to regenerate is an ancestral trait, we would assume homology in the features of regeneration, such as the mechanisms, cell types, and genetic pathways involved. A traditional separation of regenerating animals is the division of vertebrates, some of which are capable of limb regeneration, and invertebrates, some of which are capable of whole-body regeneration. This traditional separation is a remnant from early studies on Hydra and planarians to a pivot toward using vertebrate model organisms to understand the principles of regeneration (see review Elchaninov et al. 2021). This separation may also be due to whole-body regeneration being possible in some invertebrate animals but absent in all extant vertebrate species. The division of invertebrate and vertebrate regenerating organisms supported a hypothesis that regeneration competency decreases as organisms become more complex in structure and have more derived traits (Zhao et al. 2016). Hypotheses as to the differences of regeneration competency include differences in the complexity of tissues, immune systems, life cycles, and development (Aztekin and Storer 2022). For example, invertebrate models for regeneration have stem cells, and vertebrate animals capable of regeneration have cells that can undergo dedifferentiation and transdifferentiation; these mechanisms are limited in animals that are not able to regenerate (Tanaka and Reddien 2011). Likewise, animals with more complex immune systems seem to have more difficulty undergoing regeneration (Zhao et al. 2016; Elchaninov et al. 2021; Aztekin and Storer 2022). Complex immune systems are those that include scarring and inflammation and possess specialized immune cells with signaling molecules that coordinate the immune response (Zhao et al. 2016). While some comparative studies have identified signaling pathways that are similar between model organisms for regeneration (Somorjai et al. 2012; Darnet et al. 2019), studies in regenerating organisms across taxa have revealed a large amount of diversity (Fig. 1) (Endo et al. 1997; Chera et al. 2011; Lehoczky et al. 2011; Galliot 2012; Sanders and Kent 2014; Srivastava et al. 2014; Oulhen et al. 2016; Dolan et al. 2018; Aztekin et al. 2019; Gehrke et al. 2019; Ivankovic et al. 2019; Bölük et al. 2022; Zheng et al. 2022). It should be noted that this comparison is only a subset of regenerating animals, model organisms that are typically used for comparative cellular and genetic studies. A comprehensive comparison may yield additional diversity. Even comparisons of related organisms reveal additional levels of variation. As an example, Hydra belong to the phylum Cnidaria, which are diploblastic radial animals, sister to Bilateria. Hydra are capable of whole-body regeneration (Trembley 1744). Yet, cnidarians vary in life history strategies, and not all have the same regenerative ability (discussed below). Similarly, while some planarians are capable of full body regeneration, others may have lost the ability to regenerate (Ivankovic et al. 2019). In echinoderms, the tissues that can regenerate, the timing, cells, and the mechanisms used to regenerate vary between different groups (Mashanov et al. 2008; Hernroth et al. 2010; Reinardy et al. 2015; Oulhen et al. 2016; Khadra et al. 2018).
Fig. 1.
Features of regenerating animals. Summary of tissues, mechanisms, cell types, and genetic pathways of regeneration for some organisms typically used in regeneration studies: Hydra, planaria, Hofstenia, echinoderms, axolotl, Xenopus, zebrafish, and mice. Looking across organisms reveals that there may be diversity in the factors that underlie the ability to regenerate. The use of different genetic pathways for regeneration may suggest multiple independent origins (convergent evolution) of the trait.
Regeneration has traditionally been divided into two mechanisms: morphallaxis and epimorphic. Morphallaxis is a process that does not require cell proliferation but rather rearrangement of existing cells. Epimorphic regeneration, on the other hand, requires the formation of a group of undifferentiated cells at the injury site called a blastema. Model organisms for regeneration are traditionally associated with these mechanisms, such as Hydra with morphallaxis and amphibians with epimorphosis. Cells in the Hydra polyp are constantly undergoing mitosis and being shed at the extremities. WhenHydra are bisected, epithelial cells close the wound within 6 h, and a new head with tentacles is fully formed within 72 h (Bode 2003). Early studies found no difference in the mitotic rate of intact Hydra and a Hydra undergoing head regeneration, so it was assumed that head regrowth occurred through existing cell movement (morphallaxis) (Park et al. 1970). However, it was later found that Hydra bisected in the mid-gastric region have an area of cell proliferation that is similar to a blastema (Chera et al. 2009). Blastema formation, commonly associated with amphibians, is a feature that defines epimorphic regeneration. After the loss of a limb or tail, amphibians exhibit wound closure by epidermal cells, these apical-epithelial-cap cells signal to a group of undifferentiated cells (termed the blastema). Progenitor cells in the blastema undergo proliferation and differentiation (Brockes 1997; Aztekin and Storer 2022). Similar to Hydra, blastema-like structures have been identified in some planarians, echinoderms, and lancelets (Somorjai et al. 2012; Khadra et al. 2018; Ivankovic et al. 2019). Since both mechanisms can be found in the same animal group and even in the same organisms, it was proposed that these concepts be redefined (Agata et al. 2007). It has also been suggested that the epimorphic process evolved from morphallaxis (Bely and Nyberg 2010; Elchaninov et al. 2021). Authors hypothesize this evolutionary history of the processes due to their underlying similarities, but epimorphic being a “bilaterian innovation” (Bely and Nyberg 2010; Elchaninov et al. 2021). However, as mentioned above, blastema-like structure, typical of epimorphic regeneration, has been discovered in Hydra, a non-bilaterian. This highlights the importance of correctly defining the mechanisms of regeneration and identifying their homology to unravel whether these mechanisms evolved once or multiple times.
Another important feature for regenerating animals is the types of cells that can be used to regrow missing body parts. Some animals, such as Hydra and planarians, can use pluripotent stem cells, while others, such as vertebrates, make use of a mix of multipotent and/or unipotent cells. These multipotent or unipotent progenitor cells can arise from dedifferentiation, transdifferentiation, or migration. The comparative cellular underpinnings of regenerating organisms have been reviewed in the literature and highlight many unknowns (Tanaka and Reddien 2011; Storer and Miller 2020; Srivastava 2021). As an example, the contribution of amoebocytes and coelomocytes as pluripotent cells in different echinoderms is still unresolved (Khadra et al. 2018). Similarly, heterogenous multipotent and unipotent cells are still being identified in regenerating complex structures such as limbs in amphibians and digit tips in mice (Storer and Miller 2020; Aztekin and Storer 2022). Moreover, the organizing cells for regenerating tissues and their signaling components have also yet to be characterized. Characterization of organizing cells and cells used for regeneration will give further insight into the mechanisms of regeneration and potential cell homologies (Arendt et al. 2016; Shafer 2019; Srivastava 2021).
A third component of interest in determining the evolution of regeneration is the genetic and signaling pathways underlying this complex process. The field of evolutionary development (EvoDevo) highlighted similarities in the molecular underpinnings of development across organisms (Carroll 2008). EvoDevo theory posits that complex trait evolution has a deep homology and novel traits arise by making use of established genes and gene networks (Carroll 2008; Monteiro and Podlaha 2009; Shubin et al. 2009). As regeneration requires tissue patterning, similar to development, many researchers have focused on conserved developmental pathways to investigate their role in regeneration. Such studies have found a role in regeneration for pathways including Wnt, MAPK/JNK, FGF, TGF, and BMP signaling (Hobmayer et al. 2000; Srivastava et al. 2014; Scimone et al. 2016; Aztekin et al. 2019; Darnet et al. 2019). However, while some of the genes in these pathways may result in a loss of regeneration when inhibited or knocked down, the complete gene regulatory networks and their conservation across organisms remain unknown (Srivastava 2021). An alternative model to regeneration being homologous is that regeneration arose convergently and makes use of similar but non-orthologous genes through co-option of a different member of the gene family (paralog switching), co-option of different genes, or use of different gene regulatory networks. To determine the evolutionary history of regeneration, we need to incorporate a phylogenetic framework in investigating the homology of cellular and molecular mechanisms underlying this complex process (see Srivastava 2021). The availability of genomic tools in non-model organisms and advances in transgenics can now help us decipher some of the missing links or additional diversity in the molecular underpinnings of regeneration.
Here, I highlight how comparative approaches and new technologies can help resolve some of the unknown factors of regeneration. Firstly, I propose an expansion of comparative studies into non-model organisms and suggest a broadening of cnidarian species used to further investigate the features of regeneration. Next, I discuss insights gained by genome-wide and multi-omics approaches and the limitations of current studies using high-throughput sequencing. Lastly, I emphasize how new tools and approaches can be used to identify unknown features of regeneration. One such advancement in the field is the rise of single-cell sequencing that can now be used to identify cell types and trajectories associated with regeneration. Another advancement is the application of CRISPR to generate transgenics in non-model organisms, which can be used to functionally validate candidate genes and pathways. We can now make broad comparisons between closely related species and across taxa to identify components of regeneration that are conserved or co-opted, thus giving insight into the evolution of this complex trait.
Cnidaria as a model to investigate regeneration
Cnidaria, the phylum that Hydra belong to, includes corals, sea anemones, and jellyfish. As a sister group to Bilateria (the group that encompasses all bilateral animals), studies in cnidarians can give insight into the evolution of genomes, gene networks, and gene functions. In addition to their phylogenetic placement, cnidarians are of interest in the scientific community due to their venomous nature (Klompen et al. 2021), eye complexity (Picciani et al. 2018), and regenerating capabilities (Galliot and Schmid 2002). A rise in popularity of cnidarian research has resulted in new resources for comparative and genetic studies, such as genome assemblies (Putnam et al. 2007; Chapman et al. 2010; Gold et al. 2019; Leclère et al. 2019; Ohdera et al. 2019; Cazet et al. 2023; Zimmermann et al. 2023; Schnitzler et al. 2024) and single-cell atlases (Sebé-Pedrós et al. 2018; Siebert et al. 2019; Chari et al. 2021) (see Table 1). These resources along with the unique biology of cnidarians present an opportunity to investigate the features and evolutionary history of regeneration. While some species such as Nematostella, Hydra, Hydractinia, Clytia, and Turritopsis have quite a few tools to build from, many species outside of Hydrozoan remain underrepresented and should be developed for comparative approaches.
Table 1.
Publicly available cnidarian resources.
| Group | Species | Genome (NCBI RefSeq) | Number of scaffolds | Single-cell atlas | Transgenic methods |
|---|---|---|---|---|---|
| Anthozoa | Nematostella vectensis | Chromosome-level: GCA_033964005.1 | 29 | Yes | https://doi.org/10.1242/dev.204387 |
| Acropora digitifera | Genome assembly: GCF_000222465.1 | 2420 | No | N/A | |
| Exaiptasia diaphana | Genome assembly: GCF_001417965.1 | 4312 | No | N/A | |
| Anthopleura elegantissima | Genome assembly: GCA_042767785.1 | 4216 | No | N/A | |
| Edwardsiella lineata | none | - | No | N/A | |
| Hydrozoa | Hydra vulgaris | Chromosome level (AEP): GCF_038396675.1 | 15 | Yes | https://doi.org/10.1007/978-1-0716-2172-1_34 |
| Hydra oligactis | Genome assembly: GCA_024195425.1 | 16,310 | No | N/A | |
| Hydractinia symbiolongicarpus | Chromosome level: GCF_029227915.1 | 78 | Yes | https://doi.org/10.1186/s12864-018-5032-z | |
| Clytia hemisphaerica | Genome assembly: GCF_902728285.1 | 1396 | Yes | https://doi.org/10.1038/s41598-018-30188-0 | |
| Turritopsis rubra | Genome assembly: GCA_039566895.2 | 326 | Yes | https://doi.org/10.1038/s41467-024-49848-z | |
| Scyphozoa | Aurelia aurita | Genome assembly: GCA_004194415.1 | 2709 | No | N/A |
| Cassiopea xamachana | Genome assembly: GCA_964235115.1 | 561 | No | N/A | |
| Chrysaora fuscescens | Genome assembly: GCA_009936425.2 | 295,560 | No | N/A | |
| Rhizostoma pulmo | none | - | No | N/A | |
| Cubozoa | Tripedalia cystophora | none | - | No | N/A |
| Alatina alata | Genome assembly: GCA_008930755.2 | 2,005,718 | No | N/A | |
| Chironex flecker | none | - | No | N/A | |
| Carybdea rastonii | none | - | No | N/A | |
| Staurozoa | Haliclystus sanjuanensis | none | - | No | N/A |
| Lucernaria quadricornis | none | - | No | N/A | |
| Craterolophus convolvulus | none | - | No | N/A | |
| Calvadosia cruxmelitensis | Genome assembly: GCA_900245855.1 | 50,999 | No | N/A |
It remains unknown whether life cycle complexities affect regeneration competency. Cnidarians have diverse life cycles, which may influence their ability and their method of regeneration (Collins 2002). To our knowledge, all cnidarians are capable of asexual reproduction, which may facilitate the ability to regenerate during some developmental stages. Gaps in knowledge include whether stem cells are present across cnidarians (Gold and Jacobs 2013) and the extent to which medusae can regenerate. A comparison of cnidarian species commonly used in the field of biology reveals differences in their life cycles (Fig. 2). Nematostella and Exaiptaisia are anthozoan species that have external fertilization that proceeds to a planula larva. However, while Exaiptaisia can produce copies of itself from pedal lacerates that grow into a young anemone (Presnell et al. 2022), Nematostella is capable of asexual fission by physal pinching and polarity reversal (Reitzel et al. 2007). Within the group Hydrozoa, two commonly studied species include Hydra and Clytia. Under favorable conditions, Hydra reproduces asexually by budding, but under stressful conditions hermaphroditic Hydra undergoes gamete production. In this case, eggs are fertilized within the female, and an embryo is released that hatches into a young Hydra (Galliot and Schmid 2002; Steele et al. 2019). On the contrary, Clytia has a medusa adult stage, external fertilization, a planula stage, and a polyp colony (Leclère et al. 2019). In addition to Hydrozoa, the group Medusozoa has two other classes: Cubozoa (box jellyfish) and Scyphozoa (true jellyfish). The box jellyfish Tripedalia has a life cycle that includes internal fertilization, and each polyp undergoes metamorphosis to become an adult medusa (Werner et al. 1971). Unlike Tripedalia, the moon jellyfish Aurelia undergoes spawning, planula settle to produce a structure called a strobila, from which each segment that buds off becomes a juvenile medusa (Brekhman et al. 2015). In this brief comparison, it seems as though the medusae are more limited in their regenerative abilities. It is possible that this life stage is not capable of healing and regenerating in the traditional sense due to its more complex structure. The full extent of variation in life history and how it contributes to regeneration remains to be determined, but the diversity within Cnidaria makes it a unique group in which to investigate this potential link.
Fig. 2.
Cnidarian life cycles. Nematostella can reproduce sexually, asexually by physal pinching and polarity reversal, and are capable of full body regeneration, drawing adapted from Kelava 2014. Exaipatasia can reproduce sexually, asexually by pedal lacerates, and can regenerate their oral disc, drawing adapted from the Guse lab website. Hydra can reproduce sexually, asexually by budding, and are capable of full body regeneration, drawing adapted from Schaible et al. 2017. Clytia have external fertilization that proceeds by a polyp colony to produce medusae and are capable of umbrella and organ regeneration, drawing adapted from Wikimedia Commons by Munro. Aurelia has external fertilization that proceeds by a strobila structure. They are capable of regeneration of polyp from tentacles and of ephyra symmetrization, drawing adapted from Matveev et al. 2012. Tripedalia polyps reproduce asexually, and each polyp undergoes metamorphosis into an individual medusa. They are capable of fully regenerating their sensory structure called a rhopalium, drawing adapted from Gurska and Gram 2014. Stars indicate known life stages where regeneration has been investigated. Question marks denote life stages where regeneration is possible, but the extent of regenerative capacity is yet unresolved.
The extent of regeneration competency across cnidarians is not yet well characterized, but recent work suggests regeneration is possible in polyps, sea anemones, and to some extent even medusae. Hydra are well known and have long been studied as a model for regeneration (Galliot and Schmid 2002; Bode 2003; Galliot 2012). Hydra have an incredible regenerative capability, being able to regenerate head and foot when bisected and even capable of generating full polyps from cell aggregates (Trembley 1744; Gierer et al. 1972; Technau et al. 2000; Vogg et al. 2019). Similar to Hydra, Nematostella are another cnidarian species that have been widely studied and are also capable of full body regeneration (Layden et al. 2016). Recent research has expanded to investigate the tentacle and oral disc regeneration of sea anemones Calliactis and Exaiptaisia (Stewart et al. 2017; van der Burg et al. 2020). It is hypothesized that the regenerative abilities of sea anemones are due to their lack of a medusa stage and capacity to reproduce asexually (van der Burg and Prentis 2021). While medusae are seemingly more complex organisms, they may also possess the ability to regenerate. As an example, juvenileAurelia use their muscular network to rearrange existing body parts after injury to achieve symmetry (Abrams et al. 2015). On the other hand,Clytia medusae are capable of healing and regenerating their umbrella and organs by tissue remodeling, cell proliferation, and cell migration (Sinigaglia et al. 2020). The immortal jellyfish Turritopsis dohrnii, when stressed or damaged, reverts from a medusa to a polyp stage through cell transdifferentiation and changes in expression of genes associated with aging, development, tissue differentiation, transposable elements (TEs), and undescribed functions (Matsumoto et al. 2019). Lastly, Tripedalia can regenerate a sensory structure called a rhopalium, which contains eyes, gravity sensors, and neurons (Stamatis et al. 2018). The extent of whole organisms or organ regeneration across the phylum remains an open question and a fruitful field for future investigation.
To advance our understanding of regeneration, it is imperative to expand investigations into comparative studies such as within Cnidaria. Comparisons of species within this phylum will reveal similarities and differences in the features and mechanisms used for regeneration. While the phylogenetic relationships within the speciose phylum are not fully resolved, major relationships are known, and the phylogenetic placement of widely studied species has also been identified (DeBiasse et al. 2024). Using a phylogenetic framework, researchers interested in regeneration can seek to address questions such as: (1) What are the cell types used for regeneration? (2) What are the cell mechanisms of regeneration? (Do cells dedifferentiate? Transdifferentiate? Is there an organizer cell that arises?) (3) What genetic pathways are used for healing and repatterning? and (4) What are the gene regulatory networks that underlie regeneration? (Fig. S1). These studies will provide fundamental knowledge as to the nuances of this process. By characterizing the genomic features that underlie life histories, development, and regeneration, we can identify components that are shared within the phylum. Subsequently, expanding these comparisons to Bilateria, we can determine features that are conserved across regenerating animals giving insight into the evolutionary history of regeneration.
Determining functional genomics of regeneration in Cnidaria
The advancement of sequencing technologies achieved during the last decade allowed researchers to investigate genome-wide effects on traits of interest. Such studies have revealed additional genes involved in regeneration than were previously known. As an example, in Hydra, a fundamental study using in situ hybridization of candidate developmental genes uncovered a role of Wnt3 signaling in head regeneration (Hobmayer et al. 2000). Transcriptomic studies expanded on this work by characterizing the expression, timing, and regulation of pathways involved in animal response to injury and patterning (Petersen et al. 2015; Cazet et al. 2021; Murad et al. 2021). In addition to Wnt signaling, we now have a better understanding of the potential contributions of factors such as Jun, Fos, Fox, Otx, Creb, EGR, Rfx, Pax, and Zic (Petersen et al. 2015; Cazet et al. 2021; Murad et al. 2021). Similarly, a transcriptomic study in Nematostella characterized the expression of homeobox genes and transcription factors during oral and aboral regeneration and highlighted the overexpression of genes associated with developmental pathways (Schaffer et al. 2016). A comparison of genes involved in regeneration between planaria and Nematostella found Otx and Six involved in planaria oral and Nematostella aboral regeneration (Schaffer et al. 2016). Meanwhile, SoxB, Wnt2, and FoxD were associated with head regeneration in both (Schaffer et al. 2016). A similar pattern arises when we look at regeneration genes in Hydra and the acoel Hofstenia. In Hofstenia, Wnt3, Brachyury, Sp5, and FoxA1 associated with Hydra head regeneration were found to function in posterior regeneration (Ramirez et al. 2020). These comprehensive studies provide a wide suite of genes and transcription factors to investigate to decipher the components and molecular mechanisms of regeneration. To determine the extent to which these patterns of gene expression are shared across organisms, we need to sample more broadly and use a comparative framework.
Comparative studies seek to identify conserved features of regeneration. Thus, a focus on known injury response and developmental pathways is intuitive. Yet, transcriptomic studies have demonstrated that, while similar pathways may be used, the processes may take different trajectories. As an example, a study comparing Hydra head regeneration and head development during budding found that gene expression was more dynamic during regeneration (Murad et al. 2021). This study found that the head organizer gene, Wnt3, increased in expression throughout budding, but during regeneration, Wnt3 is lowly expressed early on and peaks around 12 h post-bisection and then decreases in expression again (Murad et al. 2021). Similarly, 298 genes had different expression profiles and different genetic trajectories during budding and regeneration (Murad et al. 2021). These results suggest regeneration does not recapitulate development, and that there may be more unexplored distinctions to this process. While a focus on known developmental genes has increased our understanding of regeneration, it has also limited our attention to a handful of pathways. The conservation of gene regulatory networks and underlying molecular processes of comparative regeneration remain to be determined (Srivastava 2021). Genome-wide approaches have discovered genes important to regeneration that are species-specific, uncharacterized, or novel (Petersen et al. 2015; Bryant et al. 2017; Stewart et al. 2017; Matsumoto et al. 2019). Future studies should begin to decipher the actions of these uncharacterized and novel genes, which may carry out unique functions during regeneration. Uncovering the functions of these novel and species-specific genes will help us better understand the mechanisms of regeneration that are currently limited due to a focus on identifying conserved genetic pathways for regeneration. Moreover, this understanding may help resolve the evolutionary history of regeneration.
Using multi-omics approaches to shed light on regeneration
Multi-omics is the practice of combining multiple “omics” datasets to better understand a pathway or biological process. Some examples of these -omics include genomics (genome content), transcriptomics (gene expression), proteomics (protein), metabolomics (metabolites), and microbiomics (microbiome) (Hasin et al. 2017). The approach of combining these data types gives information about complex processes from multiple levels of biological organization. Multi-omics studies have the potential to bridge the knowledge gap between genotype and phenotype relationships. As an example, RNA-seq (transcriptomics) studies tell us which genes are expressed differentially between treatments or tissue types. Combining RNA-seq with ChIP-seq or ATAC-seq (chromatin accessibility) reveals areas of cis-regulation, transcription factor binding, and the regulatory effects on gene expression. This combination allows us to create putative gene regulatory networks and to visualize how pathways proceed and interact. An added level of validation comes from using proteomics or metabolomics, which can confirm mRNA translation, protein structure, protein–protein interactions, and the physiological outcome of metabolic processes. Although a promising field, some limitations of multi-omics approaches include issues with normalization, sampling bias, and poorly designed experiments (Krassowski et al. 2020). Future studies should consider statistical power and reproducibility in designing experiments that may reveal additional genes, proteins, or pathways that contribute to their process of interest.
In Cnidaria, previous studies combined RNA-seq, ATAC-seq, and ChIP-seq to identify developmental pathways important for regeneration (Cazet et al. 2021; Murad et al. 2021). With advances in single-cell genomics, we can now determine gene regulation at the single-cell level by combining scRNA-seq and scATAC-seq. Peterson et al. combined time series transcriptomics and proteomics to identify genes and proteins important for injury response and repatterning (Petersen et al. 2015). By combining the two data types, the list of candidate genes more than doubled, and novel genes and proteins associated with regeneration were identified (Petersen et al. 2015). One thing to keep in mind when combining data sets is the time frames relevant to linking genomics and proteomics studies. For example, in mammals, an increase in mRNA results in a change of protein concentration anytime between minutes to 2–3 days (Hargrove et al. 1991). Metabolomics can be used to complement the above approaches to identify metabolic pathways that are being upregulated or downregulated during healing and regeneration. For example, research has shown that stress and immune response are important for regeneration, and the metabolic pathways for these processes are known in some model organisms. Proteomic studies found increased activity of metabolic pathways during planaria and mice liver regeneration (see review Franco et al. 2013). Metabolomics can be used to investigate the activity of metabolic pathways during regeneration to determine whether any facilitate the process of healing or injury response. Metabolic studies during regeneration may confirm the function and timing of these pathways and identify new ones important for regeneration. Furthermore, a rising amount of research is being done to investigate the role of microorganisms on cnidarian immunity, development, and regeneration (Bosch et al. 2014; Parisi et al. 2020; Klein et al. 2021). An open question is whether host/microbe relationships may enhance cnidarian immune systems thus contributing to healing during regeneration. A recent study found that anti-microbial treated Nematostella and Aiptasia regenerated slower than control animals, and treatment resulted in fewer and smaller regenerated tentacles suggesting a role for microorganisms in cnidarian regenerative ability (Da-Anoy et al. 2025). This integration of microbiomics has the potential to decipher the role of external factors in regeneration competency. These studies have an important role in understanding regeneration and are also important in the context of species resilience and conservation.
Identifying cell types of regenerating tissues
While some organisms have stem cells that can produce different cell types during regeneration, other regenerating organisms make use of multipotent or unipotent cells (Tanaka and Reddien 2011). The actions of organizing cells after injury, the constraints or malleability of cell identities, and the trajectories taken by differentiating cells during regeneration are understudied. We can now begin to address this gap in knowledge using single-cell sequencing and analyses. Advances in single-cell technologies now facilitate cell identification and lineage tracing. As an example, a study in Xenopus tadpoles identified a regeneration-organizing cell type that allowed for regeneration competence (Aztekin et al. 2019). These cells are found in the wound epidermis and express genes involved in regeneration signaling pathways such as FGF, Wnt, BMP, Msx, and Notch (Aztekin et al. 2019). On the other hand, single-cell analyses in Axolotl found that connective tissue cells dedifferentiate into a multipotent state and then redifferentiate during limb regeneration (Gerber et al. 2018). By tracking the movement and gene expression of connective tissue cells, researchers were able to detect a restriction in source cells recruited by different areas of the regenerating limb. Similar studies using single-cell technologies in other organisms can help identify cell types that may play a large role in regeneration. Determining the cell types that are used by different regenerating animals and the cell states or gene expression trajectories that they take will help identify potential homologies or convergence in the process.
Cnidarians are diploblastic animals, with two germ layers as opposed to the three found in bilaterian animals. Although seemingly simple in structure, they possess epithelial, muscle, gland, neuron, stem, and novel stinging cells (Sebé-Pedrós et al. 2018; Siebert et al. 2019). There is now an increasing amount of knowledge about the cell types and trajectories of some model cnidarians, including Nematostella, Hydra, Clytia, and Hydractinia (Sebé-Pedrós et al. 2018; Siebert et al. 2019; Chari et al. 2021; Salamanca-Díaz et al. 2025). These cell atlases were generated with adult organisms, but as technologies improve, there is a push to incorporate developmental time points and an opportunity to study regeneration dynamics at the single-cell level. This knowledge can be leveraged to investigate cell signaling, cell specification, and cell movement during regeneration. For example, Hydra have 3 stem cell lineages whose differentiation trajectories have been projected and described in detail (Siebert et al. 2019; Cazet et al. 2023). Yet, the movement of cells and whether the same differentiation trajectories are taken during regeneration and development remain to be discovered. Hydra also have a cluster of cells known as the head organizer that maintains axial patterning and controls head regeneration (Bode 2012). The cellular composition and signaling mechanisms of the head organizer remain elusive. Single-cell sequencing during a regeneration time course may help decipher the cellular substructure and signaling of the Hydra head organizer. This type of analysis will reveal similarities or differences between a “steady state” organizer and “regenerating” organizer. Similar studies can be done to compare the mechanisms of regeneration in other cnidarians where the presence and potency of stem cells are unknown (Gold and Jacobs 2013). Moreover, cell atlases and cell trajectory analyses from regenerating tissues can help identify potential organizing cells that arise after injury and may reveal whether dedifferentiation or transdifferentiation (mechanisms in regenerating vertebrate models) takes place in addition to the use of stem cells. By investigating the identity of cells deployed for regeneration and their molecular signaling, we can detect the elements that underlie malleability and constraints of cells to differentiate. For example, if organizer cells are detected in regenerating cnidarians, their gene expression profiles can be compared to those of the Xenopus cell organizer to determine whether there are any unifying principles. Similarly, if new cell states arise in cnidarians due to dedifferentiation, similar to axolotls, we can probe the characteristics of these cell types that make them primed to undergo this transition. This knowledge can be applied to engineer cells and manipulate regeneration competency in other animals.
Gene editing to validate candidate features of regeneration competency
Previous and future genetics studies will identify genomic components important for regeneration. Historically, gene knockdowns or chemical inhibitions allowed researchers to validate the association of some key pathways in regeneration. However, this was limited to a few genes and a few model organisms. The advent of CRISPR technology now allows researchers to identify and validate specific gene functions in many non-model organisms. For regeneration studies, CRISPR knock-ins can be used to apply fluorescent tags to genes that are cell-specific, injury-related, or candidate regeneration genes to visualize their expression. In addition, knockout animals can be generated and tested for regenerative ability. A potential limitation is that knocking down development associated genes may be detrimental to an organism. If organisms are malleable to knock-in approaches, this can be used to generate an inducible system to target developmental genes during regeneration (Cao et al. 2016). Alternatively, knockdown approaches using electroporation of shRNA and RNAi have been established in cnidarians (Karabulut et al. 2019; Quiroga-Artigas et al. 2020).
Cnidarians are a good system for gene editing protocols to uncover the genetic underpinnings of regeneration. Firstly, some cnidarians are small and easy to maintain in a lab. A few species are being established as model organisms for studies in stem cell biology, neurogenesis, aging, development, and regeneration. Secondly, many of them are capable of asexual reproduction, which allows for both large populations and genetically identical individuals. Currently, standardized protocols for care and husbandry have been published for Nematostella and Clytia (Lechable et al. 2020; Carvalho et al. 2023). Gene editing methods are also already in place for Nematostella, Hydractinia, and to some extent Hydra (Lommel et al. 2017; Nakanishi and Martindale 2018; Sanders et al. 2018; Carvalho et al. 2023). These standardized protocols can be used as a starting point to investigate regeneration in some of these lab reared cnidarians and then expanded to additional species. As we know, there are potentially different mechanisms, cell types, and competency of regeneration for cnidarians, comparative gene editing studies will reveal the exact function of genetic components and whether the same genes and gene networks are used for regeneration within the phylum.
Additional considerations
Genomic components that have been historically overlooked in regeneration due to limited genomic resources are TEs. Long thought of as “junk” DNA, recent studies have identified a potential role for TEs in mammal development (Low et al. 2021; Senft and Macfarlan 2021). In regenerating animals, TEs were dynamically expressed during regeneration in the sea cucumber Holothuria glaberrima, in Hydra, and in Turritopsis (Mashanov et al. 2012; Petersen et al. 2015; Matsumoto et al. 2019). In addition, it was recently hypothesized that regulation of TEs is important for regeneration competency across animals (Angileri et al. 2022). In Hydra, “active TEs” were found to be responsible for changes in genome length between two strains, and some active TEs were found to be inserted and expressed in stem cells (Kon-Nanjo et al. 2024). The specific actions of TEs during regeneration remain unknown. Specifically, if any families are under selection or whether TEs have a direct role in gene regulation during regeneration is an open question. Determining whether TEs have a functional role in regeneration is a tall task because TEs remain difficult to classify and study. Future research should aim to characterize TEs in cnidarian genomes, investigate their expression profiles (transcriptomics or in situ if possible), and attempt to decipher their specific functions with gene silencing approaches.
Conclusions
We are now entering an exciting new chapter for comparative genomics studies. Advances in sequencing technologies and bioengineering make it possible to expand out from model organisms into diverse species and traits. Cnidarians are a unique system for comparative studies on regeneration due to their phylogenetic placement, diverse life histories, and regenerative capacity. Cnidarians represent a growing field of interest with many open questions for investigation. Genome-wide approaches have identified components of the genome that have until now been under investigated and may have an important role in regeneration. Combination of multi-omics approaches can now reveal the missing links between genotype and phenotype relationships in regeneration. Single-cell genomics allow us to identify cell types and trace their differentiation trajectories. Lastly, transgenics in multiple organisms can be used to validate gene functions. A comparative approach of regenerating animals with different regeneration potentials will help decipher the mechanisms, cellular, and genetic components of regeneration. As we deepen our understanding of the mechanisms underlying regeneration, it is now crucial to elucidate its evolutionary history. Investigating the factors that underlie regeneration across animals will unravel homologies or instances of co-option. To do this, we must pivot away from a focus on a few model organisms to investigating regenerations in a broad range of taxa, including early branching organisms. Understanding the shared components of regenerating organisms, or unique adaptations, holds the potential to manipulate regenerative ability using gene therapy and bioengineering.
Supplementary Material
Acknowledgments
I am grateful for the guidance provided by Dr. Ali Mortazavi as I transitioned into the field of regeneration and for advice on this manuscript. I want to thank undergraduate and graduate students in my lab for discussing this paper, especially Ayanna Mays who provided comments and recommended edits. I also want to thank the reviewers who provided constructive advice that helped improve this manuscript.
Notes
From the symposium “Cnidarian sensory systems as comparative models for the evolution of complexity” presented at the annual meeting of the Society for Integrative and Comparative Biology, January 3-7th, 2025.
References
- Abrams MJ, Basinger T, Yuan W, Guo CL, Goentoro L. 2015. Self-repairing symmetry in jellyfish through mechanically driven reorganization. Proc Natl Acad Sci USA. 112:E3365–73. 10.1073/pnas.1502497112 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Agata K, Saito Y, Nakajima E. 2007. Unifying principles of regeneration I: epimorphosis versus morphallaxis. Dev Growth Differ. 49:73–8. 10.1111/j.1440-169X.2007.00919.x [DOI] [PubMed] [Google Scholar]
- Alvarado AS, Tsonis PA. 2006. Bridging the regeneration gap: genetic insights from diverse animal models. Nat Rev Genet. 7:873–84. 10.1038/nrg1923 [DOI] [PubMed] [Google Scholar]
- Angileri KM, Bagia NA, Feschotte C. 2022. Transposon control as a checkpoint for tissue regeneration. Development. 149:149. 10.1242/dev.191957 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Arendt D, Musser JM, Baker CVH, Bergman A, Cepko C, Erwin DH, Pavlicev M, Schlosser G, Widder S, Laubichler MD et al. 2016. The origin and evolution of cell types. Nat Rev Genet. 17:744–57. 10.1038/nrg.2016.127 [DOI] [PubMed] [Google Scholar]
- Aztekin C, Hiscock TW, Marioni JC, Gurdon JB, Simons BD, Jullien J. 2019. Identification of a regeneration-organizing cell in the Xenopus tail. Science. 364:653–8. 10.1126/science.aav9996 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Aztekin C, Storer MA. 2022. To regenerate or not to regenerate: vertebrate model organisms of regeneration-competency and -incompetency. Wound Repair Regen. 30:623–35. 10.1111/wrr.13000 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Bely AE, Nyberg KG. 2010. Evolution of animal regeneration: re-emergence of a field. Trends Ecol Evol. 25:161–70. 10.1016/j.tree.2009.08.005 [DOI] [PubMed] [Google Scholar]
- Berg HC, Brown DA. 1972. Regeneration of Hydra from reaggregated cells. Nature. 239:500–4. 10.1038/239500a0 [DOI] [PubMed] [Google Scholar]
- Bode HR. 2003. Head regeneration in Hydra. Dev Dyn. 226:225–36. 10.1002/dvdy.10225 [DOI] [PubMed] [Google Scholar]
- Bode HR. 2012. The head organizer in Hydra. Int J Dev Biol. 56:473–8. 10.1387/ijdb.113448hb [DOI] [PubMed] [Google Scholar]
- Bölük A, Yavuz M, Demircan T. 2022. Axolotl: a resourceful vertebrate model for regeneration and beyond. Dev Dyn. 251:1914–33. 10.1002/dvdy.520 [DOI] [PubMed] [Google Scholar]
- Bosch TCG, Adamska M, Augustin R, Domazet‐Loso T, Foret S, Fraune S, Funayama N, Grasis J, Hamada M, Hatta M et al. 2014. How do environmental factors influence life cycles and development? An experimental framework for early-diverging metazoans. BioEssays. 36:1185–94. 10.1002/bies.201400065 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Brekhman V, Malik A, Haas B, Sher N, Lotan T. 2015. Transcriptome profiling of the dynamic life cycle of the scypohozoan jellyfish Aurelia aurita. BMC Genomics. 16:74. 10.1186/s12864-015-1320-z [DOI] [PMC free article] [PubMed] [Google Scholar]
- Brockes JP. 1997. Regeneration : amphibian limb structure rebuilding a complex structure. Science. 276:81–7. [DOI] [PubMed] [Google Scholar]
- Brockes JP, Kumar A. 2008. Comparative aspects of animal regeneration. Annu Rev Cell Dev Biol. 24:525–49. 10.1146/annurev.cellbio.24.110707.175336 [DOI] [PubMed] [Google Scholar]
- Bryant DM, Johnson K, Ditommaso T, Tickle T, Couger MB, Payzin-Dogru D, Lee TJ, Leigh ND, Kuo T-H, Davis FG et al. 2017. A tissue-mapped axolotl de novo transcriptome enables identification of limb regeneration factors. Cell Rep. 18:762–76. 10.1016/j.celrep.2016.12.063 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Cao J, Wu L, Zhang S-M, Lu M, Cheung WKC, Cai W, Gale M, Xu Q, Yan Q 2016. An easy and efficient inducible CRISPR/Cas9 platform with improved specificity for multiple gene targeting. Nucleic Acids Res. 44:gkw660. 10.1093/nar/gkw660 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Carroll SB. 2008. Evo-devo and an expanding evolutionary synthesis: a genetic theory of morphological evolution. Cell. 134:25–36. 10.1016/j.cell.2008.06.030 [DOI] [PubMed] [Google Scholar]
- Carvalho JE, Burtin M, Detournay O, Amiel AR, Rottinger E. 2025. Optimized husbandry and targeted gene-editing for the cnidarian Nematostella vectensis. Development. 152:dev204387. 10.1242/dev.204387 [DOI] [PubMed] [Google Scholar]
- Cazet J, Cho A, Juliano C. 2021. Generic injuries are sufficient to induce ectopic wnt organizers in Hydra. Elife. 10:1–31. 10.7554/eLife.60562 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Cazet JF, Siebert S, Little HM, Bertemes P, Primack AS, Ladurner P, Achrainer M, Fredriksen MT, Moreland RT, Singh S et al. 2023. A chromosome-scale epigenetic map of the Hydra genome reveals conserved regulators of cell state. Genome Res. 33:283–98. 10.1101/gr.277040.122 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Chapman JA, Kirkness EF, Simakov O, Hampson SE, Mitros T, Weinmaier T, Rattei T, Balasubramanian PG, Borman J, Busam D et al. 2010. The dynamic genome of Hydra. Nature. 464:592–6. 10.1038/nature08830 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Chari T, Weissbourd B, Gehring J, Ferraioli A, Leclère L, Herl M, Gao F, Chevalier S, Copley RR, Houliston E et al. 2021. Whole-animal multiplexed single-cell RNA-seq reveals transcriptional shifts across Clytia medusa cell types. Sci Adv. 7:eabh1683. 10.1126/sciadv.abh1683 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Chera S, Ghila L, Dobretz K, Wenger Y, Bauer C, Buzgariu W, Martinou J-C, Galliot B. 2009. Apoptotic cells provide an unexpected source of Wnt3 signaling to drive Hydra head regeneration. Dev Cell. 17:279–89. 10.1016/j.devcel.2009.07.014 [DOI] [PubMed] [Google Scholar]
- Chera S, Ghila L, Wenger Y, Galliot B. 2011. Injury-induced activation of the MAPK/CREB pathway triggers apoptosis-induced compensatory proliferation in Hydra head regeneration. Dev Growth Differ. 53:186–201. 10.1111/j.1440-169X.2011.01250.x [DOI] [PubMed] [Google Scholar]
- Collins AG. 2002. Phylogeny of medusozoa and the evolution of cnidarian life cycles. J Evol Biol. 15:418–32. 10.1046/j.1420-9101.2002.00403.x [DOI] [Google Scholar]
- Da-Anoy J, Toyama K, Jasnos O, Audrey W, Gilmore TD, Davies SW. 2025. Microbial depletion is associated with slower cnidarian regeneration. Integr Comp Biol. icaf007. 10.1093/icb/icaf007 [DOI] [PubMed]
- Darnet S, Dragalzew AC, Amaral DB, Sousa JF, Thompson AW, Cass AN, Lorena J, Pires ES, Costa CM, Sousa MP et al. 2019. Deep evolutionary origin of limb and fin regeneration. Proc Natl Acad Sci USA. 116:15106–15. 10.1073/pnas.1900475116 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Debiasse MB, Buckenmeyer A, Macrander J, Babonis LS, Bentlage B, Cartwright P, Prada C, Reitzel AM, Stampar SN, Collins A et al. 2024. A cnidarian phylogenomic tree fitted with hundreds of 18S leaves. Bull Soc Syst Biol. 3:3. 10.18061/bssb.v3i2.9267 [DOI] [Google Scholar]
- Dolan CP, Dawson LA, Muneoka K. 2018. Digit Tip regeneration: merging regeneration biology with regenerative medicine. Stem Cells Trans Med. 7:262–70. 10.1002/sctm.17-0236 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Elchaninov A, Sukhikh G, Fatkhudinov T. 2021. Evolution of regeneration in animals: a tangled story. Front Ecol Evol. 9:1–14. 10.3389/fevo.2021.621686 [DOI] [Google Scholar]
- Endo T, Yokoyama H, Tamura K, Ide H. 1997. Shh expression in developing and regenerating limb buds of Xenopus laevis. Dev Dyn. 209:227–32. . [DOI] [PubMed] [Google Scholar]
- Franco C, Soares R, Pires E, Koci K, Almeida AM, Santos R, Coelho AV. 2013. Understanding regeneration through proteomics. Proteomics. 13:686–709. 10.1002/pmic.201200397 [DOI] [PubMed] [Google Scholar]
- Galliot B. 2012. Hydra, a fruitful model system for 270 years. Int J Dev Biol. 56:411–23. 10.1387/ijdb.120086bg [DOI] [PubMed] [Google Scholar]
- Galliot B, Schmid V. 2002. Cnidarians as a model system for understanding evolution and regeneration the developmental interest of Cnidarians. Int J Dev Biol. 46:39–48. [PubMed] [Google Scholar]
- Gehrke AR, Neverett E, Luo Y-J, Brandt A, Ricci L, Hulett RE, Gompers A, Ruby JG, Rokhsar DS, Reddien PW et al. 2019. Acoel genome reveals the regulatory landscape of whole-body regeneration. Science. 363:363. 10.1126/science.aau6173 [DOI] [PubMed] [Google Scholar]
- Gerber T, Murawala P, Knapp D, Masselink W, Schuez M, Hermann S, Gac-Santel M, Nowoshilow S, Kageyama J, Khattak S et al. 2018. Single-cell analysis uncovers convergence of cell identities during axolotl limb regeneration. Science. 362:362. 10.1126/science.aaq0681 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Gold DA, Jacobs DK. 2013. Stem cell dynamics in Cnidaria: are there unifying principles?. Dev Genes Evol. 223:53–66. 10.1007/s00427-012-0429-1 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Gold DA, Katsuki T, Li Y, Yan X, Regulski M, Ibberson D, Holstein T, Steele RE, Jacobs DK, Greenspan RJ. 2019. The genome of the jellyfish Aurelia and the evolution of animal complexity. Nat Ecol Evol. 3:96–104. 10.1038/s41559-018-0719-8 [DOI] [PubMed] [Google Scholar]
- Hargrove JL, Hulsey MG, Beale EG. 1991. The kinetics of mammalian gene expression. BioEssays. 13:667–74. 10.1002/bies.950131209 [DOI] [PubMed] [Google Scholar]
- Hasin Y, Seldin M, Lusis A. 2017. Multi-omics approaches to disease. Genome Biol. 18:1–15. 10.1186/s13059-017-1215-1 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hernroth B, Farahani F, Brunborg G, Dupont S, Dejmek A, Nilsson Sköld H. 2010. Possibility of mixed progenitor cells in sea star arm regeneration. J Exp Zool Pt B. 314B:457–68. 10.1002/jez.b.21352 [DOI] [PubMed] [Google Scholar]
- Hobmayer B, Rentzsch F, Kuhn K, Happel CM, Von Laue CC, Snyder P, Rothbächer U, Holstein TW. 2000. WNT signalling molecules act in axis formation in the diploblastic metazoan Hydra. Nature. 407:186–9. 10.1038/35025063 [DOI] [PubMed] [Google Scholar]
- Ivankovic M, Haneckova R, Thommen A, Grohme MA, Vila-Farré M, Werner S, Rink JC. 2019. Model systems for regeneration: planarians. Development. 146:146. 10.1242/dev.167684 [DOI] [PubMed] [Google Scholar]
- Karabulut A, He S, Chen CY, McKinney SA, Gibson MC. 2019. Electroporation of short hairpin RNAs for rapid and efficient gene knockdown in the starlet sea anemone, Nematostella vectensis. Dev Biol. 448:7–15. 10.1016/j.ydbio.2019.01.005 [DOI] [PubMed] [Google Scholar]
- Khadra YB. 2018. Regeneration in stellate Echinoderms: Crinoidea, Asteroidea and Ophiuroidea. Results Probl Cell Differ. 65:285–320. [DOI] [PubMed] [Google Scholar]
- Klein S, Frazier V, Readdean T, Lucas E, Diaz-Jimenez EP, Sogin M, Ruff ES, Echeverri K. 2021. Common environmental pollutants negatively affect development and regeneration in the sea anemone Nematostella vectensis holobiont. Front Ecol Evol. 9:1–15. 10.3389/fevo.2021.786037 [DOI] [Google Scholar]
- Klompen AML, Kayal E, Collins AG, Cartwright P. 2021. Phylogenetic and selection analysis of an expanded family of putatively pore-forming jellyfish toxins (Cnidaria: medusozoa). Genome Biol Evol. 13:1–17. 10.1093/gbe/evab081 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kon-Nanjo K, Kon T, Yu TC-TK, Rodriguez-Terrones D, Falcon F, Martínez DE, Steele RE, Tanaka EM, Holstein TW, Simakov O. 2024. The dynamic genomes of Hydra and the anciently active repeat complement of animal chromosomes. bioRxiv. 10.1101/2024.03.13.584568 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Krassowski M, Das V, Sahu SK, Misra BB. 2020. State of the field in multi-omics research: from computational needs to data mining and sharing. Front Genet. 11:1–17. 10.3389/fgene.2020.610798 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Layden MJ, Rentzsch F, Röttinger E. 2016. The rise of the starlet sea anemone Nematostella vectensis as a model system to investigate development and regeneration. WIREs Dev Biol. 5:408–28. 10.1002/wdev.222 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lechable M, Jan A, Duchene A, Uveira J, Weissbourd B, Gissat L, Collet S, Gilletta L, Chevalier S, Leclère L et al. 2020. An improved whole life cycle culture protocol for the hydrozoan genetic model Clytia hemisphaerica. Biol Open. 9:bio051268. 10.1242/BIO.051268 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Leclère L, Horin C, Chevalier S, Lapébie P, Dru P, Peron S, Jager M, Condamine T, Pottin K, Romano S et al. 2019. The genome of the jellyfish Clytia hemisphaerica and the evolution of the cnidarian life-cycle. Nat Ecol Evol. 3:801–10. 10.1038/s41559-019-0833-2 [DOI] [PubMed] [Google Scholar]
- Lehoczky JA, Robert B, Tabin CJ. 2011. Mouse digit tip regeneration is mediated by fate-restricted progenitor cells. Proc Natl Acad Sci USA. 108:20609–14. 10.1073/pnas.1118017108 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lommel M, Tursch A, Rustarazo-calvo L, Trageser B, Thomas W. 2017. Genetic knockdown and knockout approaches in Hydra. bioRxiv. 1–19. 10.1101/230300 [DOI]
- Low Y, Tan DEK, Hu Z, Tan SYX, Tee WW. 2021. Transposable element dynamics and regulation during zygotic genome activation in mammalian embryos and embryonic stem cell model systems. Stem Cells Int. 2021:1–17. 10.1155/2021/1624669 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Maden M. 2018. The evolution of regeneration—where does that leave mammals?. Int J Dev Biol. 62:369–72. 10.1387/ijdb.180031mm [DOI] [PubMed] [Google Scholar]
- Mashanov VS, Zueva OR, García‐Arrarás JE. 2012. Posttraumatic regeneration involves differential expression of long terminal repeat (LTR) retrotransposons. Dev Dyn. 241:1625–36. 10.1002/dvdy.23844 [DOI] [PubMed] [Google Scholar]
- Mashanov VS, Zueva OR, Heinzeller T. 2008. Regeneration of the radial nerve cord in a holothurian: a promising new model system for studying post-traumatic recovery in the adult nervous system. Tissue and Cell. 40:351–72. 10.1016/j.tice.2008.03.004 [DOI] [PubMed] [Google Scholar]
- Matsumoto Y, Piraino S, Miglietta MP. 2019. Transcriptome characterization of reverse development in Turritopsis dohrnii (Hydrozoa, Cnidaria). G3 (Bethesda). 9:4127–38. 10.1534/g3.119.400487 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Monteiro A, Podlaha O. 2009. Wings, horns, and butterfly eyespots: how do complex traits evolve?. PLoS Biol. 7:0209–16. 10.1371/journal.pbio.1000037 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Murad R, Macias-Muñoz A, Wong A, Ma X, Mortazavi A. 2021. Coordinated gene expression and chromatin regulation during Hydra head regeneration. Genome Biol Evol. 13:1–17. 10.1093/gbe/evab221 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Nakanishi N, Martindale MQ. 2018. CRISPR knockouts reveal an endogenous role for ancient neuropeptides in regulating developmental timing in a sea anemone. Elife. 7:1–16. 10.7554/eLife.39742 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ohdera A, Ames CL, Dikow RB, Kayal E, Chiodin M, Busby B, La S, Pirro S, Collins AG, Medina M et al. 2019. Box, stalked, and upside-down? Draft genomes from diverse jellyfish (Cnidaria, Acraspeda) lineages: Alatina alata (Cubozoa), Calvadosia cruxmelitensis (Staurozoa), and Cassiopea xamachana (Scyphozoa). Gigascience. 8:1–15. 10.1093/gigascience/giz069 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Oulhen N, Heyland A, Carrier TJ, Zazueta-Novoa V, Fresques T, Laird J, Onorato TM, Janies D, Wessel G. 2016. Regeneration in bipinnaria larvae of the bat star Patiria miniata induces rapid and broad new gene expression. Mech Dev. 142:10–21. 10.1016/j.mod.2016.08.003 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Parisi MG, Parrinello D, Stabili L, Cammarata M. 2020. Cnidarian immunity and the repertoire of defense mechanisms in anthozoans. Biology. 9:1–26. 10.3390/biology9090283 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Park HD, Ortmeyer AB, Blankenbaker DP. 1970. Cell division during regeneration in Hydra. Nature. 227:617–9. [DOI] [PubMed] [Google Scholar]
- Petersen HO, Höger SK, Looso M, Lengfeld T, Kuhn A, Warnken U, Nishimiya-Fujisawa C, Schnölzer M, Krüger M, Özbek S et al. 2015. A comprehensive transcriptomic and proteomic analysis of Hydra head regeneration. Mol Biol Evol. 32:1928–47. 10.1093/molbev/msv079 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Picciani N, Kerlin JR, Sierra N, Swafford AJM, Ramirez MD, Roberts NG, Cannon JT, Daly M, Oakley TH. 2018. Prolific origination of eyes in Cnidaria with co-option of non-visual opsins. Curr Biol. 28:2413–2419.e4. 10.1016/j.cub.2018.05.055 [DOI] [PubMed] [Google Scholar]
- Presnell JS, Wirsching E, Weis VM. 2022. Tentacle patterning during Exaiptasia diaphana pedal lacerate development differs between symbiotic and aposymbiotic animals. PeerJ. 10:1–24. 10.7717/peerj.12770 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Putnam NH, Srivastava M, Hellsten U, Dirks B, Chapman J, Salamov A, Terry A, Shapiro H, Lindquist E, Kapitonov VV et al. 2007. Sea anemone genome reveals ancestral eumetazoan gene repertoire and genomic organization. Science. 317:86–94. 10.1126/science.1139158 [DOI] [PubMed] [Google Scholar]
- Quiroga-Artigas G, Duscher A, Lundquist K, Waletich J, Schnitzler CE. 2020. Gene knockdown via electroporation of short hairpin RNAs in embryos of the marine hydroid Hydractinia symbiolongicarpus. Sci Rep. 10:10. 10.1038/s41598-020-69489-8 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ramirez AN, Loubet-Senear K, Srivastava M. 2020. A regulatory program for initiation of wnt signaling during posterior regeneration. Cell Rep. 32:108098. 10.1016/j.celrep.2020.108098 [DOI] [PubMed] [Google Scholar]
- Reinardy HC, Emerson CE, Manley JM, Bodnar AG. 2015. Tissue regeneration and biomineralization in sea urchins: role of Notch signaling and presence of stem cell markers. PLoS One. 10:1–15. 10.1371/journal.pone.0133860 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Reitzel AM, Burton PM, Krone C, Finnerty JR. 2007. Comparison of developmental trajectories in the starlet sea anemone Nematostella vectensis: embryogenesis, regeneration, and two forms of asexual fission. Invertebr Biol. 126:99–112. 10.1111/j.1744-7410.2007.00081.x [DOI] [Google Scholar]
- Salamanca-Díaz DA, Horkan HR, García-Castro H, Emili E, Salinas-Saavedra M, Pérez-Posada A, Rossi ME, Álvarez-Presas M, Gabhann RM, Hillenbrand P et al. 2025. The Hydractinia cell atlas reveals cellular and molecular principles of cnidarian coloniality. Nat Commun. 16:2121. 10.1038/s41467-025-57168-z [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sanders JL, Kent ML. 2014. The zebrafish as a model for complex tissue regeneration. Trends Genetics. 29:357–70. 10.1002/9781118395264.ch14 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sanders SM, Ma Z, Hughes JM, Riscoe BM, Gibson GA, Watson AM, Flici H, Frank U, Schnitzler CE, Baxevanis AD et al. 2018. CRISPR/Cas9-mediated gene knockin in the hydroid Hydractinia symbiolongicarpus. BMC Genomics. 19:1–17. 10.1186/s12864-018-5032-z [DOI] [PMC free article] [PubMed] [Google Scholar]
- Schaffer AA, Bazarsky M, Levy K, Chalifa-Caspi V, Gat U. 2016. A transcriptional time-course analysis of oral vs. aboral whole-body regeneration in the Sea anemone Nematostella vectensis. BMC Genomics. 17:1–22. 10.1186/s12864-016-3027-1 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Schnitzler CE, Chang ES, Waletich J, Quiroga-Artigas G, Wong WY, Nguyen A-D, Barreira SN, Doonan LB, Gonzalez P, Koren S et al. 2024. The genome of the colonial hydroid Hydractinia reveals that their stem cells use a toolkit of evolutionarily shared genes with all animals. Genome Res. 34:498–513. 10.1101/gr.278382.123 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Scimone ML, Cote LE, Rogers T, Reddien PW. 2016. Two FGFRL-Wnt circuits organize the planarian anteroposterior axis. Elife. 5:1–19. 10.7554/eLife.12845 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sebé-Pedrós A, Saudemont B, Chomsky E, Plessier F, Mailhé M-P, Renno J, Loe-Mie Y, Lifshitz A, Mukamel Z, Schmutz S et al. 2018. Cnidarian cell type diversity and regulation revealed by whole-organism single-cell RNA-seq. Cell. 173:1520–1534.e20. 10.1016/j.cell.2018.05.019 [DOI] [PubMed] [Google Scholar]
- Senft AD, Macfarlan TS. 2021. Transposable elements shape the evolution of mammalian development. Nat Rev Genet. 22:691–711. 10.1038/s41576-021-00385-1 [DOI] [PubMed] [Google Scholar]
- Shafer MER. 2019. Cross-species analysis of single-cell transcriptomic data. Front Cell Dev Biol. 7:1–9. 10.3389/fcell.2019.00175 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Shubin N, Tabin C, Carroll S. 2009. Deep homology and the origins of evolutionary novelty. Nature. 457:818–23. 10.1038/nature07891 [DOI] [PubMed] [Google Scholar]
- Siebert S. 2019. Stem cell differentiation trajectories in Hydra resolved at single-cell resolution. Science. 365:eaav9314. 10.1101/460154 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sinigaglia C, Peron S, Eichelbrenner J, Chevalier S, Steger J, Barreau C, Houliston E, Leclère L. 2020. Pattern regulation in a regenerating jellyfish. Elife. 9:1–33. 10.7554/ELIFE.54868 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Somorjai IML, Somorjai RL, Garcia-Fernàndez J, Escrivà H. 2012. Vertebrate-like regeneration in the invertebrate chordate amphioxus. Proc Natl Acad Sci USA. 109:517–22. 10.1073/pnas.1100045109 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Srivastava M. 2021. Beyond casual resemblance: rigorous frameworks for comparing regeneration across species. Annu Rev Cell Dev Biol. 37:415–40. 10.1146/annurev-cellbio-120319-114716 [DOI] [PubMed] [Google Scholar]
- Srivastava M, Mazza-Curll KL, Van Wolfswinkel JC, Reddien PW. 2014. Whole-body acoel regeneration is controlled by wnt and bmp-admp signaling. Curr Biol. 24:1107–13. 10.1016/j.cub.2014.03.042 [DOI] [PubMed] [Google Scholar]
- Stamatis SA, Worsaae K, Garm A. 2018. Regeneration of the rhopalium and the rhopalial nervous system in the box jellyfish Tripedalia cystophora. Biol Bull. 234:22–36. 10.1086/697071 [DOI] [PubMed] [Google Scholar]
- Steele RE, Updegrove MD, Kirolos SA, Mowery L, Martínez DE, Bryant PJ. 2019. Reproductive bet-hedging and existence in vernal pools as components of Hydra life history. Biol Bull. 237:111–8. 10.1086/705161 [DOI] [PubMed] [Google Scholar]
- Stewart ZK, Pavasovic A, Hock DH, Prentis PJ. 2017. Transcriptomic investigation of wound healing and regeneration in the cnidarian Calliactis polypus. Sci Rep. 7:1–11. 10.1038/srep41458 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Storer MA, Miller FD. 2020. Cellular and molecular mechanisms that regulate mammalian digit tip regeneration: mechanisms of digit tip regeneration. Open Biol. 10:200194. 10.1098/rsob.200194 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Tanaka E, Reddien PW. 2011. The cellular basis for animal regeneration sources of new cells in animal regeneration. Dev Cell. 21:172–85. 10.1016/j.devcel.2011.06.016 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Technau U, Cramer Von Laue C, Rentzsch F, Luft S, Hobmayer B, Bode HR, Holstein TW. 2000. Parameters of self-organization in Hydra aggregates. Proc Natl Acad Sci USA. 97:12127–31. 10.1073/pnas.97.22.12127 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Tiozzo S, Copley RR. 2015. Reconsidering regeneration in metazoans: an evo-devo approach. Front Ecol Evol. 3:1–12. 10.3389/fevo.2015.00067 [DOI] [Google Scholar]
- Trembley A. 1744. Mémoires pour Servir à L'histoire d'une Genre de Polypes D'eau Douce, à Bras En Forme de Cornes. Jean & Herman Verbeek: Leiden. [Google Scholar]
- Van Der Burg CA, Pavasovic A, Gilding EK, Pelzer ES, Surm JM, Smith HL, Walsh TP, Prentis PJ. 2020. The rapid regenerative response of a model sea anemone species Exaiptasia pallida is characterised by tissue plasticity and highly coordinated cell communication. Mar Biotechnol. 22:285–307. 10.1007/s10126-020-09951-w [DOI] [PubMed] [Google Scholar]
- van der Burg CA, Prentis PJ. 2021. The tentacular spectacular: evolution of regeneration in sea anemones. Genes. 12:1072. 10.3390/genes12071072 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Vogg MC, Galliot B, Tsiairis CD. 2019. Model systems for regeneration: Hydra. Development. 146:146. 10.1242/dev.177212 [DOI] [PubMed] [Google Scholar]
- Werner B, Cutress CE, Studebaker JP. 1971. Life cycle of Tripedalia cystophora Conant (Cubomedusae). Nature. 232:582–3. [DOI] [PubMed] [Google Scholar]
- Zhao A, Qin H, Fu X. 2016. What determines the regenerative capacity in animals?. Bioscience. 66:735–46. 10.1093/biosci/biw079 [DOI] [Google Scholar]
- Zheng M, Zueva O, Hinman VF. 2022. Regeneration of the larval sea star nervous system by wounding induced respecification to the sox2 lineage. Elife. 11:1–23. 10.7554/eLife.72983 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zimmermann B, Montenegro JD, Robb SMC, Fropf WJ, Weilguny L, He S, Chen S, Lovegrove-Walsh J, Hill EM, Chen C-Y et al. 2023. Topological structures and syntenic conservation in sea anemone genomes. Nat Commun. 14:8270. 10.1038/s41467-023-44080-7 [DOI] [PMC free article] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.


