Next Article in Journal
Insights from Expert Interviews on Navigating the Complexity of Prioritizing Chemicals for Human Biomonitoring in Latvia
Previous Article in Journal
Curcumin Can Inhibit Zearalenone-Induced Ferroptosis in Porcine Intestinal Epithelial Cells via the p53/SLC7A11/GPX4 Pathway
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Integration of “Omics”-Based Approaches in Environmental Risk Assessment to Establish Cause and Effect Relationships: A Review

1
Cawthron Institute, Private Bag 2, Nelson 7042, New Zealand
2
School of Biological Sciences, University of Auckland, Auckland 1142, New Zealand
3
Institute of Marine Science, University of Auckland, Private Bag 92019, Auckland 1142, New Zealand
4
Australian Rivers Institute, Griffith University Gold Coast, Building G51, Edmund Rice Drive, Southport, QLD 4215, Australia
5
Bioeconomy Science Institute, Lincoln 7640, New Zealand
*
Author to whom correspondence should be addressed.
Toxics 2025, 13(9), 714; https://doi.org/10.3390/toxics13090714
Submission received: 30 July 2025 / Revised: 18 August 2025 / Accepted: 23 August 2025 / Published: 24 August 2025
(This article belongs to the Special Issue Ecotoxicological Monitoring of Aquatic Systems)

Abstract

Marine and freshwater environments are under increasing pressure from anthropogenic stressors. The resulting impacts on exposed ecosystems are complex and challenging to characterise. The effects may be subtle and exhibited over long time periods. Effective and robust approaches are required to characterise the physiological and genetic processes that are impacted by pollutants to assess how populations and ecosystems may be adversely affected and at risk. The objective of the review is to provide an overview of “omics” methodologies used to assess the risk of stressors on exposed biota. This review covers the development of key omics approaches and how they have been used to contribute towards improved knowledge about the effects of environmental stressors, from molecular to whole-organism and community levels of biological organisation. We provide insights into how ecotoxicogenomics approaches can be used for various aspects of environmental risk assessment by characterising toxicological mechanisms of action. This information can be used to confirm cause-and-effect relationships required to better manage risks and protect the integrity and functionality of ecosystems.

1. Introduction

The integrity of ecosystems is vital for economic prosperity, environmental sustainability, and human and stock health [1]. Major changes in land use result in increasing anthropogenic inputs, such as urban, agricultural, and industrial runoff, that can result in environmental decline [2,3,4]. The presence of complex mixtures of legacy pollutants and emerging organic contaminants has been described as one of the key environmental issues facing humanity [5,6,7]. It has been established that anthropogenic chemicals, also described as novel entities, are now transgressing their planetary boundary at a threshold that threatens to impact biophysical and biochemical systems needed to maintain life on Earth [8,9,10]. Pollutants can impact various physiological processes of ecosystems, including disruption to reproductive processes that can potentially lead to population decline [6,11]. Contaminants can also induce sub-lethal biological responses that can lead to adverse effects at the population and community level [12]. Better information on whether anthropogenic pressures will result in adverse effects to an ecosystem and what specific stressors or anthropogenic activities are causing the harm are some of the challenges facing environmental managers [13].
The environmental risk assessment (ERA) framework uses chemical and biological methodologies to characterise the likely or actual adverse effects of natural and anthropogenic stressors on ecosystem health [14]. Biomarkers can provide a functional measure of an organism’s response to single and complex mixtures of chemical stressors that are bioavailable in the environment, helping inform ERA processes [15]. In vitro and in vivo bioassays are useful to characterise toxicity and predict bioavailability and biological effects of contaminants [16,17,18]. Such environmental data provide multiple lines of evidence for the weight-of-evidence process to inform on the impacts of stressors on ecosystems [19]. The adverse outcome pathway (AOP) concept also integrates knowledge concerning the linkage between a molecular initiating event (MIE; e.g., a molecular interaction between a pollutant and a specific biomolecule) and an adverse health effect relevant to the ERA [20].
Ecotoxicogenomics, or the integration of omics-derived data (e.g., genomics, transcriptomics, proteomics and metabolomics) into ecotoxicology, is a developing field that is revolutionising our ability to characterise responses to environmental stressors. These methodologies, known as omics, can characterise effects across levels of biological organisation, from the molecular to the whole community [21,22,23,24,25]. Technological advances have led to unprecedented sample throughput and analytical power that are redefining the fields of ecotoxicology [26], environmental monitoring [4,27] and ERA [28]. There is potential to transform regulatory frameworks and guide decision-making processes for the management of ecosystems and remediation actions [29,30,31]. While these approaches have their advantages, clear rationales for their use in ERA platforms are required. The aim of this review was to survey the omics literature on how omics can be used to characterise pollutant hazards and explore opportunities for their inclusion into ERA frameworks (e.g., Figure 1).

2. Genomics

Rapid progress in the field of genomics (the study of how an individual’s genome translates into biological functions) can assist in our understanding of the impacts of chemicals on human and ecosystem health [25,32]. Various novel high-throughput sequencing (HTS) approaches have been developed (e.g., Illumina (NextSeq, HiSeq, NovaSeq), Oxford Nanopore (e.g., MinION, PromethION), as well as Pacific Biosystems (e.g., Single Molecular Real-Time (SMRT) sequencing)). The technology has advanced with a range of platforms now available that range from 100 base pair reads to over 10 kb reads, facilitating whole genome sequencing and assembly. The evolution of HTS technologies has increased sensitivity and accuracy for genomics research and enabled the development of new biomarker assays from tissue, blood, and other sample types.
These technological advances in DNA sequencing have generated more accessible sequence information for ecologically relevant, non-model species, although the functional annotation of genes in these species can still remain a challenge [33]. This is particularly important as locally relevant species can be more effective in characterising environmental risk than standard test organisms. Functional annotation identifies genes with known characteristics such as molecular function, pathways, or biological processes and relies heavily on bioinformatic techniques to identify homology to genes with a characterised function in other organisms [32]. Additionally, gene function may also be determined by experimentation and exposure analyses in lab-based assessments, allowing for a stronger mechanistic understanding of anthropogenic stressors. Hypothesis-driven chemical exposures related to specific toxicity pathways will help detect the genes involved. Accurate gene annotations are essential for informative gene expression analyses (see transcriptomics section below). There is a need to understand both the response of genomes to genetic and environmental stimuli and, conversely, the impact the environment plays on the phenotype. Genome sequencing, in combination with the tools of functional genomics, offers opportunities to develop a better understanding of phenotypic evolution and understand the regulatory pathways involved in response to toxicants [25].
Pollutants in the environment can be at extremely low concentrations but still affect genetic diversity and the overall fitness of exposed ecosystems [34]. Genetic variation can be impacted at several levels, including nucleotide polymorphisms, copy-number variants, alternative splicing, and post-transcriptional and post-translational regulation. Characterisation of the complete genotype, including the identification of causal links between toxicant pressures and changes in genetic variability at the population level, is now feasible through relatively inexpensive whole-genome sequencing of many individuals, enabled by HTS technologies [35]. Our ability to clarify the complexity of the genomic landscape that underlies the response to pollutants is within reach. Population genomics scans can reveal both directly adaptive and compensatory loci, and quantitative trait locus (QTL) mapping may further clarify the mechanistic pathways and the response of specific phenotypes [36].

3. Epigenomics

In association with a rapid increase in whole genome characterisation, a revolution in our understanding of alternative non-DNA nucleotide sequence-based mechanisms, collectively termed epigenetics, which also contribute to phenotypic outcomes, has occurred [28]. Sources of epigenetic variation are mechanistically diverse but all alter a given genotype’s influence on an organism’s phenotype without changes in the underlying DNA nucleotide sequences [37,38]. Molecular-level epigenetic processes can activate, reduce or completely disable the activity of particular genes by a variety of processes, including (i) methylation of cytosine residues in the DNA, (ii) remodelling of chromatin structure through chemical modification of histone proteins, and (iii) altering regulatory processes mediated by small RNA molecules [39]. Epigenetic mechanisms have been shown to be altered by environmental changes, such as temperature [40], exposure to pollutants [41,42] and toxins [43], leading to alterations in gene expression and therefore phenotypic diversity [44]. These changes can be both transgenerational (i.e., germline transmission of information) and non-transgenerational (i.e., somatic, mitotic stability) factors, both of which can potentially alter the evolutionary pathway of future generations [45]. Epigenetic variation can be viewed as an extension of phenotypic plasticity across generations (i.e., trans-generational plasticity) [46].
Environmental contaminants have been shown to induce epigenetic changes to a wide range of ecologically relevant organisms, but there is a scarcity of information on the underlying mechanisms of epigenetic processes for most types of organisms [47]. The mechanisms of epigenetic inheritance appear to be well conserved across vertebrates, but are much more variable in invertebrate taxa, albeit with data from only a small number of species (e.g., Caenorhabditis elegans, Dahpnia magna, Crassostrea gigas) [48,49]. The concept of a persistent epigenetic signal over multiple generations is highly relevant to biomonitoring and ecological risk assessment [50]. Targeting epigenetic signatures may also be a useful approach for identifying ecotoxicological biomarkers. Epigenetic markers on specific genes could serve as biomarkers for exposure to specific stressors or toxic outcomes. More general markers, such as global DNA methylation, might prove to be broad indicators of accumulated stress throughout an organism’s lifetime [48].
Current epigenetics literature is primarily focused on DNA methylation, as it is a universal epigenetic marker that responds to environmental cues, and there are a range of methods available for its detection and quantification [51]. The use of methods to characterise genome-wide DNA methylation patterns (e.g., methylation-sensitive amplified fragment length polymorphism; MS-AFLP [52] or enzyme-linked immunosorbent assay (ELISA)-like spectrophotometric assays [53]) has dominated the ecological epigenetics literature. These methods can be limited, however, providing only global methylation patterns indicative of a response with no information regarding changes in specific regions of the genome, including functional genes [54]. Results can also be difficult to interpret due to simultaneous changes across many genes, and, therefore, identifying patterns in toxicity becomes difficult [55,56,57]. However, they offer affordable and approachable methods to gain insight into changes in global patterns (e.g., tissue-wide, or as a function of developmental stage) of DNA methylation between discrete samples [54]. For high-resolution analysis of DNA methylation at functional loci studies, new HTS techniques, e.g., reduced representative bisulphite sequencing (RRBS; [58]), bisulphite-converted restriction site associated with DNA sequencing (bsRADseq; [59]), methylation-specific PCR (MSP; [60]), whole-genome bisulphite sequencing (WGBS; [61]), coupled with gene expression analyses (e.g., quantitative PCR, RNA-seq), are likely to provide valuable insights into epigenetic changes in affected genes associated with functional outcomes. One promising method for the systematic analysis of epigenetic processes is using CRISPR (clustered regularly interspaced short palindromic repeats)-Cas9-based tools, which can be used for epigenetic editing at specific loci [62,63,64,65]. Until recently, epigenetic studies relied only on correlations between certain epigenetic modifications and gene regulation (i.e., activation or silencing). Epigenome editing now enables direct study of the functional relevance of certain epigenetic modifications at a specific locus or a genomic region [65].
Epigenetic biomarkers may provide early warnings of adverse outcomes associated with early and/or lifelong exposures to chemicals and other environmental stressors, that may have multigenerational effects. Apical endpoints of ecotoxicological relevance, such as growth, development, and reproduction, are known to be under epigenetic regulation [66]. With further research, the study of epigenetic markers may lead to an increased understanding of mechanisms of action of contaminants, and ultimately risk [48,50].

4. Transcriptomics

A transcriptome comprises the complete set of RNA transcripts that are actively being expressed by the genome, in a specific cell, tissue, organ, or organism at a specific point in time. Gene expression profiling to determine changes in up- or down-regulated genes as transcribed messenger RNA (mRNA) between different conditions is a powerful and sensitive method to characterise biological responses to environmental perturbations, such as exposure to toxic chemicals [26,67]. Determining these changes in gene expression in response to a foreign substance or compound aids in the elucidation of an organism’s molecular response and description of the potential mode of action (MOA) for a possible toxicant [68]. Virtually all responses to external stressors, including toxicants, involve changes in normal patterns of gene expression. Some are direct effects, for example, compounds can bind to a transcription factor (receptor), forming a complex that modulates transcription of specific genes, whereas other responses are compensatory and reflect the response of the organism to molecular damage or cellular dysfunction [69]. Different chemicals can generate specific patterns of gene expression, which can be used to generate hypotheses on the MOA. Through transcriptomic analyses, differential gene expression can be used to identify what classes of genes are responding to chemical contamination, providing sensitive and specific mechanistic insights into the underlying observed toxicity.
Vinken [70] identified two main areas where transcriptomic data are used within the field of ecotoxicology: (i) to define molecular initiating event(s) as either stand-alone data or to complement other types of data as input into AOPs, and (ii) to provide a set of biomarkers eligible for toxicity testing and hazard identification. Transcriptomic data can serve as first-level identifiers of chemical stress, and mechanisms can be confirmed using specific single-target methods at the proteomic, metabolomic, or organism level [70].
Transcriptomic experiments in aquatic toxicology have been diverse, encompassing various techniques for detecting changes in gene expression. Long-established methods include DNA microarrays, serial analysis of gene expression (SAGE), differential display (DD) and reverse transcription quantitative polymerase chain reaction (RT-qPCR), but have been largely replaced by HTS approaches. RT-qPCR permits the accurate and sensitive quantification of mRNA levels for expression profiling of specific genes. This technique has been established as a method of choice for the quantification of mRNA transcripts of a selected gene of interest in biological samples, but it is relatively low-throughput in comparison to microarrays. The resulting data can also be highly variable and difficult to reproduce without appropriate verification and validation of both samples and primers [71]. Subsequently, other forms of PCR have been developed, including digital PCR (dPCR) [72]. dPCR technology offers the advantage of direct and independent quantification of DNA without standard curves, giving more precise and reproducible data than qPCR. As dPCR is still an emerging technique in this field, additional work is necessary to demonstrate its efficacy compared to traditional qPCR assays. (e.g., [73,74]). Targeted gene expression techniques require that the targets (and thus the represented genes) are known “a priori”, and this has been a challenge for non-model species lacking a sequenced genome or transcriptome [25].
As with genomic techniques, advances in HTS methods have revolutionised RNA analyses [75]. Parallel sequencing of millions of nucleic acid molecules can provide mechanistic insights into toxicology and provide new avenues for biomarker discovery with growing relevance for risk assessment [24]. These technologies can identify transcriptional changes and genomic targets with base pair precision in response to chemical exposure [33]. Unlike targeted methods (e.g., RT-qPCR, dPCR), transcriptome sequencing can occur without prior genomic knowledge, although accurate alignment of transcripts is greatly enhanced by genome data assemblies [76]. Statistical confidence in differential transcript expression is increased by adequate ‘depth of coverage’ of the transcriptome or read number per sample, and the detection of rare transcripts requires significantly more read coverage [77]. The expense (although this is consistently decreasing), depth of transcriptome coverage, much greater data complexity, demands for computational analysis, and computational infrastructure are important considerations in transcriptome sequencing analysis. In addition, since results are only semiquantitative, validation through RT-qPCR/dPCR is often necessary.
Transcriptomic analyses can complement traditional ecotoxicology data by providing mechanistic insight and by identifying sub-lethal organismal responses and contaminant classes underlying observed toxicity. To apply transcriptomics in the context of ERA, the endpoints measured in the field or laboratory should be repeatedly and reliably associated with population-level responses. However, transcription of mRNA is only an intermediate step in converting genetic information into proteins, not all mRNA sequences are transcribed, many under-go post transcription modifications and proteins are modified (e.g., by phosphorylation, posttranslational cleavage) before becoming physiologically active which can create a complex relationship between specific responses and biomarkers to ecological adverse events [20]. Field-based studies that use transcriptomics profiling in conjunction with health indices of relevance to the monitoring programmes represent a solid direction for future research.

5. Proteomics

Proteomics is the study of the complete set of proteins present in a biological system [78]. To conduct proteomic analyses, proteins are digested to peptides and then detected using sodium dodecyl sulphate (SDS) gel electrophoresis and/or liquid chromatography with high-resolution tandem mass spectrometry [79]. Identification of the cognate intact protein is achieved by querying the mass spectrometry (MS) data against a database containing the amino acid sequences of proteins [80]. Proteomics databases are typically generated from previous genome or transcriptome characterisation of the biological system [81,82]. Methods also exist for estimating the relative abundance of each of the proteins from the MS data.
Proteomes are generally more complex and dynamic than the corresponding transcriptome from the same system. This increased diversity is the result of biological modulation of the translation process, post-translational modification of proteins such as glycosylation, phosphorylation, and linkage to lipids, and variable degradation through modulation of protease activity [83]. This complexity provides a rich and highly dynamic dataset from which to evaluate the health of the biological system being analysed, such as particular organs or fluid from a sentinel species living in an ecosystem being impacted by environmental toxins.
As with other omics technical approaches, proteomics is a non-targeted approach, reducing the requirement for selection of specific biomarkers at the outset [84]. Proteomics offers the opportunity to discover novel biomarkers for which specific low-cost assays can be developed, for example, antibody-based tests [85]. However, like other omics approaches, proteomics is dependent on pre-existing databases, and this can be a limitation for non-model species. While proteomics provides the opportunity to detect and monitor biological responses to toxicants with greater selectivity of response compared with analysis of the transcriptome, this comes with the added burden of higher analytical costs and greater complexity in data analysis compared with transcriptomics [85].
A range of methodological improvements and variations have been adapted from other areas of proteomics research and applied to ecotoxicology, and the use of proteomics in the field of ecotoxicology is still being established. One challenge is the limited extent of existing protein sequence databases for many of the most important species of interest in aquatic ecosystems [86]. Meeting this challenge will require leveraging continuing advances in high-throughput sequencing and its associated data processing technologies to establish genome resources, at least in draft form, or alternatively high-quality transcriptomes, from which proteome sequence databases can be derived. Some biological responses to chemical contaminants are generic, while others may be specific to particular compounds or classes of chemicals. For example, a meta-analysis of proteomic responses in zebrafish to environmental contaminants has shown that many of the same responses occur, irrespective of the compound being used [87]. In addition, such generic biological responses to compounds have also been recognised as a challenge to delineating the mechanism of action for ecological toxins [88]. Laboratory-based experimentation using model compounds and sentinel species provides only a simplified picture of the complex interactions occurring in nature, but nevertheless provides an important starting point towards a full understanding of biological effects at the molecular level in real-world ecosystems. The application of statistical tools such as Principal Components Analysis (PCA) and improved resolution and sensitivity of detection of perturbations of the proteome in species response to environmental stress provides a means for interpreting complex responses. Ultimately, a thorough understanding of the ecotoxicology of an ecosystem may require integration of proteomics with other omics approaches, such as transcriptomics and metabolomics, as well as combining data from several sentinel species. Advances such as these may in the future produce useful biomarkers for the early detection of stress on environments, opening the potential for mitigation at an early stage, before environmental impacts become profound or irreversible.
Ecotoxicological proteomics is now moving beyond the theoretical and into the lab, and its technical viability is established. However, most studies are largely descriptive and relatively few report mechanistic insight, but continued advances in databases and knowledge of the physiology of target species will facilitate progress.

6. Metabolomics

Metabolomics strives for the simultaneous measurement of the comprehensive suite of small endogenous molecules occurring in a biological sample [89,90]. The term metabolomics is generally used to describe small polar metabolites, the so-called metabolome, which includes various amino acids, sugars, nucleic acids, and some polar fatty acids and their derivatives [91]. Non-polar metabolites, including various classes of lipid, also fall under the general umbrella definition of metabolomics [92]; however, lipids are in many cases categorised as a separate omics discipline (lipidomics). The distinction is worth acknowledging from the perspective of molecular structure and chemical behaviour, but less so biologically since both polar and non-polar metabolites play key roles in diverse physiological and biochemical processes [93,94]. Indeed, polar and non-polar metabolites are involved with the synthesis and degradation of larger molecules, cell signalling, and contribute toward maintaining cellular homeostasis [95,96,97]. Where genomics, transcriptomics, and proteomics (i.e., the ‘upstream’ omics cascade) reflect epigenetic, post-transcriptional or post-translational modifications that may manifest as a tangible response in an organism, metabolites offer a closer representation of what has or is happening [62,98]. This relates to metabolites being tightly connected with gene expression but also strongly influenced by immediate environmental conditions [99]. This has led to metabolomics being viewed as the omics technique most closely related to phenotype, which makes it an important focus for bridging our understanding of interactions between the genetic and environmental forces acting on an organism [100].
Metabolites are most commonly measured via liquid- or gas-chromatography paired with mass spectrometry (LC-MS and GC-MS, respectively), or using Nuclear Magnetic Resonance (NMR) spectroscopy. There are advantages and disadvantages to each analytical platform, which have been discussed and debated elsewhere (e.g., [101]) and largely condense down to greater coverage of metabolites by MS countered by better high-throughput capability, greater reproducibility, and reduced costs for NMR [102]. With the recognised strength of metabolomics spanning medical and environmental applications, considerable research has been and is being directed towards developing technological advancements for both MS and NMR analytical platforms; for MS, a big focus is on reducing analytical time and costs, and, for NMR, the focus is on improved sensitivity to extend metabolite coverage [102].
Modern untargeted tools for measuring metabolites promote greater replication, extended temporal sampling and, in the case of laboratory studies, the inclusion of concentration gradients of contaminants [103], thereby facilitating quantification of the environmental exposure and determination of non-linearity or effect thresholds. Importantly, these upfront advantages facilitating robust study design are further complemented by interpretive strength once the data are obtained, due to the highly conserved nature of metabolites and biochemical pathways across species [104], making metabolomics workflows readily transferable across a wide range of biological samples, including plants [105], animals [100,106], and cell cultures [107].
One challenge with applying metabolomics to environmental toxicology is that uncontrolled environmental variables may confound the interpretation of field-scale studies [108]. For example, metabolomics was shown to offer a robust platform to qualitatively evaluate the physiological effects of metal(loid)s in fish inhabiting natural aquatic systems, but seasonal changes to environmental conditions were also observed to modify the response [99]. This is not unique to metabolomics and has been discussed in relation to many analytical techniques [109]. Thus, a simple recommendation for field-based metabolomics, and arguably for any focus, is to include temporal sampling representing a wide range of environmental conditions to best characterise differences between sites [94,99,110]. This lack of well-defined best practices and reporting standards for metabolomics data in ecotoxicology has resulted in limited translation of studies into reporting data for risk assessments [91]. However, recent initiatives have aimed to integrate metabolomics workflows and reporting to support decision-making about chemical compounds based on their mechanistic behaviour. An international expert group has developed the MERIT initiative (MEtabolomics standaRds Initiative in Toxicology) with the aim of describing the most relevant best practice guidelines, method performance standards, and minimal reporting standards for the acquisition, processing and statistical analysis of metabolomics data within the context of regulatory toxicology [111]. The establishment of robust workflows and methodologies will enable the progressive adoption of metabolomics in regulatory decision-making [91].

7. Ecogenomics

Ecological genomics (ecogenomics) applies advanced molecular technologies to study organismal responses to environmental challenges in natural settings, integrating ecology and molecular biology to uncover adaptation mechanisms at various levels and interactions between environment and phenotype [112,113,114,115,116]. Meta-omics techniques, such as metagenomics, metabarcoding, metatranscriptomics, and metaproteomics, are increasingly used to analyse genetic material directly from environmental samples [117,118].

8. Metabarcoding or Metagenetics

Metabarcoding has rapidly become an established PCR-based method for characterising the biodiversity of ecological communities in environmental samples [4,119,120,121,122]. Metabarcoding is the application of the “DNA barcoding” approach, but it utilises HTS to amplify a target gene that allows the simultaneous identification of many taxa from a bulk or environmental sample. Metabarcoding assays can be designed to encompass broad taxonomic groups, for example, viruses, bacteria, fungi, protists, or eukaryotes [123,124,125,126,127,128], or target specific taxa, such as foraminifera, ciliates, diatoms, fish, etc. [129,130,131,132,133], depending on primer selection and the aims of the study. Three key considerations when applying metabarcoding for characterising entire communities are the taxonomic resolution provided by the target gene, the ‘universality’ of the primers (i.e., will they amplify the target gene from a wide variety of taxa), and the requirement for robust reference databases [134,135,136]. Metabarcoding is highly effective for characterising aquatic communities and holds great potential for routine biomonitoring and environmental management [30,137,138,139,140]. Compared to traditional methods, it is faster, more cost-effective, and significantly improves sample throughput, accuracy, and taxonomic range. Recent studies highlight its promising applications in marine monitoring, particularly in aquaculture, offshore drilling, and coastal development (Table 1). In particular, the strong correlations obtained between traditional indices and recently developed DNA-based metrics/indices across a variety of pollution gradients (e.g. [128,141,142,143,144] suggest that metabarcoding has the potential to complement or even replace current biomonitoring techniques in the near future (reviewed in [27,145,146,147,148]. Finally, another promising avenue is the analysis of functional metabolic pathways derived from environmental DNA bacterial 16S ribosomal DNA metabarcoding to estimate the extent of functional redundancy within and among impacted biological communities [143].
Several genetic methods that do not rely on PCR can be used to characterise biological communities through metabarcoding. Non-PCR techniques are beneficial because PCR introduces bias by preferentially binding primers to certain DNA templates, increasing the likelihood of detecting some taxa while missing others. Methods like shotgun sequencing [172] and mitochondrial enrichment via centrifugation [173] avoid PCR but require ultra-deep sequencing to recover useful data, which drives up sequencing costs, data volume, and bioinformatics complexity. As a result, these methods are not yet practical for routine non-microbial environmental biomonitoring.
A third method that avoids the need for PCR is gene enrichment, which uses short synthetic DNA probes designed from reference sequences complementary to target genes. These probes capture sequences from a shotgun library, allowing for sequencing without PCR [174,175]. However, this technique requires a robust database of reference DNA sequences to create the probes. While extensively used in biomedical research and ancient DNA studies [176,177], its potential for biomonitoring macro-invertebrates has been demonstrated [149].

9. Meta-Omics

Meta-omics is a relatively novel field which aims to simultaneously analyse the taxonomy and function of all species from a biological sample. Meta-omics includes all the approaches described above but applied to a community rather than a single organism (i.e., metagenomics, metatranscriptomics, metaproteomics and community metabolomics). To date, the overwhelming majority of meta-omics studies have focused on microbial communities, mostly bacteria but also fungi, archaea, viruses, and microeukaryotes (e.g., foraminifera) [110]. Medical and clinical studies of microbiomes have driven the development of meta-omics approaches, but they are increasingly being used for environmental and ecotoxicological applications (e.g., [178,179,180]).
Meta-omics enable the inference of the complex microbial community and its relationship or interactions with the environment (and/or hosts), enabling characterisation of biochemical function and systems-level microbial interactions in situ [181,182]. However, the challenges associated with omic studies are even more complex with meta-omics, including assembly and identification of DNA/RNA sequences, metabolites and proteins of non-model organisms and the huge amount of computing power required for analyses [180,183].

10. Omics and the Development of Biomarkers

Omics demonstrate great capability to discover new toxicological biomarkers as omics profiling enables a comprehensive characterisation of a biological sample within a single analysis [184]. These techniques have the advantage of being applicable to a large diversity of samples, and, over the past decade, high-throughput technologies, such as DNA sequencing, proteomics, and metabolomic mass spectrometry, have been used to generate large amounts of data that allow for global comparison of changes in molecular profiles that underlie cellular or organism responses. As a result, the omics-based approaches, coupled with computational and bioinformatics methods, provide unprecedented opportunities to speed up biomarker discovery and are now widely used in the field of ecotoxicology to facilitate novel biomarker development. Within the AOP framework, omics datasets also enable a more precise definition of the MIE and the selection of biomarkers relevant for assessing effects and/or exposure [20,185]. Omics data provide both gene- and pathway-level read-outs that can be used for selecting measurable and relevant biomarkers [186,187]. However, changes at the molecular level do not necessarily result in the development of adverse effects at the organism level and may reflect inherent biological variability or compensatory, reversible changes. The discrimination of these effects requires biological understanding and interpretation [187]. However, under defined conditions of cellular location, time, and biological context, these changes can provide meaningful information about biological responses to a toxicant exposure.
Although omics can still be expensive and require substantial expertise to analyse and interpret the tremendous amount of data generated, systems biology is expanding at a fast rate, and the corresponding computational tools are continuously being developed [188]. The combined use of multiple omics to perform global characterisation of genes, epigenetic marks, mRNA, proteins, and metabolites from a specific biological sample will overcome the limitation of potentially examining the uninformative biological level of organisation or biological targets (see section below) [189]. Moreover, multi-omics should be able to explain complex biological phenomena not only for a class of contaminants but for multiple stressors. Consequently, untargeted omics analyses (universal methods), at least at the outset, should be favoured to identify relevant biological pathways disrupted by contaminants, and their corresponding sets of effect biomarkers [88].
Recent international assessments of the promises and challenges of omics techniques in chemical risk assessments have concluded that the major contribution lies in the classification of substances and definition of similarity, the elucidation of the mode of action of substances, and the identification of species-specific effects and the demonstration of human health relevance [187,190]. Importantly, consistent protocols for generating, storing, processing, and interpreting omics data are required for the outcomes of omics-based studies to be reliably verified and confidently integrated into regulatory hazard and risk assessment [26,190].

11. Integration of Omics Approaches for Ecotoxicology

Omics technologies can generate relevant information efficiently and accurately on substance-induced perturbations in the biochemistry and physiology of cells and organisms that are associated with adverse outcomes [189]. These technologies have been adopted in the ecotoxicology research space in the hope that they would provide tools to identify an array of biomarkers of adverse effects and mechanisms of toxicity to improve substance hazard assessments and would contribute to the development of alternative methods to animal testing [187]. However, the transfer of omics methodologies into the regulatory domain remains cautious [191]. In addition to technical issues, there are complex legal issues remaining to be addressed prior to achieving the incorporation of omics data into regulatory processes [187].
A single-omics technique will detect biomolecules of one type and thus capture changes only for a small subset of the components of a particular molecular pathway. The application of single-omics analyses can lead to the identification of targeted physiological processes for specific exposure scenarios, but they have limitations for providing a systemic understanding of toxicity or adverse outcome pathways [189]. The integration of multiple omics approaches can substantially improve the characterisation of responses to a pollutant [192], and this need has been identified (e.g., [190]). Multi-omics have particular relevance in ecotoxicology for determining the association of the molecular response with certain key events of an AOP and for the refinement of AOPs [189].
To date, multi-omics approaches to toxicological research have largely consisted of two levels of omics datasets, and studies with three or more layers are rare. The selection of the molecular level for investigation strongly depends on the research question and the model system. Examples of multi-omics approaches in toxicology studies include investigation of the effects of nanomaterials [193,194], biocides [195], trace metals [196], neurotoxins [197], and forever chemicals like perfluorooctanoic acid (PFOA) [198]. Recent advancements in high-throughput omics techniques to analyse biological samples have meant that each approach can easily generate from giga- to tera-byte-sized data files. The size of resulting data files together with differences in nomenclature among data types, accurate annotation of biomolecules, sample replication and statistical power, and data sharing, represent substantial challenges, making the integration of these multi-dimensional omics data into a biologically meaningful context challenging [199]. Many powerful omics integration methods have been published and reviewed (e.g., [200,201,202]. However, no single approach fulfils all the requirements from a toxicological perspective [189] to fully integrate the four or more critical omics approaches outlined in this review. The development of these integration methods is also limited by a lack of experiments with a multi-omics design focus using up-to-date omics techniques. These types of studies will allow a more thorough evaluation of multi-omics approaches and different integration methods [189]. The application of all single omics techniques can provide important information on biological response, but these data can be strengthened even more by correlations with other omics techniques through the formation of more extensive databases, accurate annotation and assembly of datasets.

12. Conclusions

Omics technologies are rapidly evolving and are being applied to a range of fields. These technologies are proving useful in ecotoxicology and could have a role as part of ERA frameworks by providing insights into the hazards of pollutants across the levels of biological organisation. Aspects of the technology and data interpretation remain complex to incorporate into hazard assessment, but there is enormous potential for gaining a much more nuanced understanding of the molecular mechanisms leading to the biological and ecological effects of pollutants. Better characterisation of the MoAs can lead to the identification of biomarkers of adverse effects linked to validated and confirmed AOPs. Many omics are new approach methodologies (NAMs) with low ethical costs and could be used as part of the regulatory process for the registration of chemicals and as part of routine monitoring programmes to assess perturbations or recovery following remedial actions. Ongoing validation and standardisation of omics will continue to demonstrate their use in ecotoxicological research and environmental management.
The implementation and incorporation of omics into ERA frameworks will require ongoing dialogue across disciplines and the involvement of environmental managers and regulators. This is needed to ensure that the information generated by omics can be validated to bring added value. Interpretation of omics data requires specialised bioinformatic expertise and extensive knowledge of the molecular physiology of the organism studied. The application of omics to ecotoxicology is currently better suited to model organisms with an extensive dataset and knowledge base. The application of omics approaches to non-model organisms is more challenging and requires species-specific molecular and physiological knowledge. During the development of approaches incorporating omics, we must consider the relevance of the information they provide that is fit for purpose for the protection of target species and ecosystems.

Author Contributions

Conceptualisation, K.F.S., X.P., S.D.M., T.T.W. and L.A.T.; methodology, K.F.S., X.P., S.D.M., T.T.W. and L.A.T.; investigation, K.F.S., X.P., S.D.M., T.T.W. and L.A.T.; writing—original draft preparation, K.F.S., X.P., S.D.M., T.T.W. and L.A.T.; writing—review and editing, K.F.S., X.P., S.D.M., T.T.W. and L.A.T.; project administration, L.A.T.; funding acquisition, L.A.T. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by the New Zealand Ministry of Business Innovation and Employment (MBIE) Endeavour grant “Emerging organic contaminants—managing risk for a safer NZ environment and economy”, grant number CAWX1708 and MBIE grant number C03X1802.

Acknowledgments

The authors thank Jacqui Stuart (Cawthron Institute) and Nicolette Faville (Bioeconomy Science Institute) for assistance with the figures.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Bell, C. Branding New Zealand: The National Green-wash. Br. Rev. N. Z. Stud. 2006, 15, 13–28. [Google Scholar]
  2. Gillespie, P.A.; Forrest, R.W.; Peake, B.M.; Basher, L.R.; Clement, D.M.; Dunmore, R.A.; Hicks, D.M. Spatial delineation of the depositional footprint of the Motueka River outwelling plume in Tasman Bay, New Zealand. N. Z. J. Mar. Freshw. Res. 2011, 45, 455–475. [Google Scholar] [CrossRef]
  3. Tremblay, L.A.; Trought, K.; Sheehan, T.J.; Holmes, R.J.P.; Barrick, A.; Young, R.G. Induction of metallothionein in the common bully (Gobiomorphus cotidianus) from the Motueka River. N. Z. J. Mar. Freshw. Res. 2021, 55, 497–503. [Google Scholar] [CrossRef]
  4. Wood, S.A.; Smith, K.F.; Banks, J.C.; Tremblay, L.A.; Rhodes, L.; Mountfort, D.; Cary, S.C.; Pochon, X. Molecular genetic tools for environmental monitoring of New Zealand’s aquatic habitats, past, present and the future. N. Z. J. Mar. Freshw. Res. 2013, 47, 90–119. [Google Scholar] [CrossRef]
  5. Kumar, A.; Xagoraraki, I. Pharmaceuticals, personal care products and endocrine-disrupting chemicals in U.S. surface and finished drinking waters: A proposed ranking system. Sci. Total Environ. 2010, 408, 5972–5989. [Google Scholar] [CrossRef]
  6. Schwarzenbach, R.P.; Escher, B.I.; Fenner, K.; Hofstetter, T.B.; Johnson, C.A.; von Gunten, U.; Wehrli, B. The challenge of micropollutants in aquatic systems. Science 2006, 313, 1072–1077. [Google Scholar] [CrossRef] [PubMed]
  7. Wang, F.; Xiang, L.L.; Leung, K.S.Y.; Elsner, M.; Zhang, Y.; Guo, Y.M.; Pan, B.; Sun, H.W.; An, T.C.; Ying, G.G.; et al. Emerging contaminants: A One Health perspective. Innovation 2024, 5, 100612. [Google Scholar] [CrossRef]
  8. Matlin, S.A.; Cornell, S.E.; Krief, A.; Hopf, H.; Mehta, G. Chemistry must respond to the crisis of transgression of planetary boundaries. Chem. Sci. 2022, 13, 11710–11720. [Google Scholar] [CrossRef]
  9. Richardson, K.; Steffen, W.; Lucht, W.; Bendtsen, J.; Cornell, S.E.; Donges, J.F.; Drüke, M.; Fetzer, I.; Bala, G.; von Bloh, W.; et al. Earth beyond six of nine planetary boundaries. Sci. Adv. 2023, 9, eadh2458. [Google Scholar] [CrossRef]
  10. Rockström, J.; Donges, J.F.; Fetzer, I.; Martin, M.A.; Wang-Erlandsson, L.; Richardson, K. Planetary Boundaries guide humanity’s future on Earth. Nat. Rev. Earth Environ. 2024, 5, 773–788. [Google Scholar] [CrossRef]
  11. Halpern, B.S.; Walbridge, S.; Selkoe, K.A.; Kappel, C.V.; Micheli, F.; D’Agrosa, C.; Bruno, J.F.; Casey, K.S.; Ebert, C.; Fox, H.E.; et al. A global map of human impact on marine ecosystems. Science 2008, 319, 948–952. [Google Scholar] [CrossRef]
  12. Dallas, L.J.; Jha, A.N. Applications of biological tools or biomarkers in aquatic biota: A case study of the Tamar estuary, South West England. Mar. Pollut. Bull. 2015, 95, 618–633. [Google Scholar] [CrossRef] [PubMed]
  13. Lam, P.K.S.; Gray, J.S. The use of biomarkers in environmental monitoring programmes. Mar. Pollut. Bull. 2003, 46, 182–186. [Google Scholar] [CrossRef]
  14. Van der Oost, R.; Beyer, J.; Vermeulen, N.P.E. Fish bioaccumulation and biomarkers in environmental risk assessment: A review. Environ. Toxicol. Pharmacol. 2003, 13, 57–149. [Google Scholar] [CrossRef]
  15. Adams, S.M.; Giesy, J.P.; Tremblay, L.A.; Eason, C.T. The use of biomarkers in ecological risk assessment: Recommendations from the Christchurch conference on Biomarkers in Ecotoxicology. Biomarkers 2001, 6, 1–6. [Google Scholar] [CrossRef]
  16. Boehler, S.; Strecker, R.; Heinrich, P.; Prochazka, E.; Northcott, G.L.; Ataria, J.M.; Leusch, F.D.L.; Braunbeck, T.; Tremblay, L.A. Assessment of urban stream sediment pollutants entering estuaries using chemical analysis and multiple bioassays to characterise biological activities. Sci. Total Environ. 2017, 593–594, 498–507. [Google Scholar] [CrossRef]
  17. Charry, M.P.; Keesing, V.; Costello, M.; Tremblay, L.A. Assessment of the ecotoxicity of urban estuarine sediment using benthic and pelagic copepod bioassays. PeerJ 2018, 6, e4936. [Google Scholar] [CrossRef] [PubMed]
  18. Leusch, F.D.L.; Allen, H.; De Silva, N.A.L.; Hodson, R.; Johnson, M.; Neale, P.A.; Stewart, M.; Tremblay, L.A.; Wilde, T.; Northcott, G.L. Effect-based monitoring of two rivers under urban and agricultural influence reveals a range of biological activities in sediment and water extracts. J. Environ. Manag. 2024, 351, 119692. [Google Scholar] [CrossRef] [PubMed]
  19. Burton, G.A., Jr.; Chapman, P.M.; Smith, E.P. Weight-of-evidence approaches for assessing ecosystem impairment. Hum. Ecol. Risk Assess. 2002, 8, 1657–1673. [Google Scholar] [CrossRef]
  20. Ankley, G.T.; Bennett, R.S.; Erickson, R.J.; Hoff, D.J.; Hornung, M.W.; Johnson, R.D.; Mount, D.R.; Nichols, J.W.; Russom, C.L.; Schmieder, P.K.; et al. Adverse outcome pathways: A conceptual framework to support ecotoxicology research and risk assessment. Environ. Toxicol. Chem. 2010, 29, 730–741. [Google Scholar] [CrossRef]
  21. Cheifet, B. Where is genomics going next? Genome Biol. 2019, 20, 17. [Google Scholar] [CrossRef]
  22. Gutleben, J.; De Mares, M.C.; van Elsas, J.D.; Smidt, H.; Overmann, J.; Sipkema, D. The multi-omics promise in context: From sequence to microbial isolate. Crit. Rev. Microbiol. 2018, 44, 212–229. [Google Scholar] [CrossRef]
  23. Hudson, M.E. Sequencing breakthroughs for genomic ecology and evolutionary biology. Mol. Ecol. Resour. 2008, 8, 3–17. [Google Scholar] [CrossRef]
  24. Prat, O.; Degli-Esposti, D. 6—New Challenges: Omics Technologies in Ecotoxicology. In Ecotoxicology; Gross, E., Garric, J., Eds.; Elsevier: Amsterdam, The Netherlands, 2019; pp. 181–208. [Google Scholar]
  25. Snape, J.R.; Maund, S.J.; Pickford, D.B.; Hutchinson, T.H. Ecotoxicogenomics: The challenge of integrating genomics into aquatic and terrestrial ecotoxicology. Aquat. Toxicol. 2004, 67, 143–154. [Google Scholar] [CrossRef]
  26. Zhang, X.; Xia, P.; Wang, P.; Yang, J.; Baird, D.J. Omics Advances in Ecotoxicology. Environ. Sci. Technol. 2018, 52, 3842–3851. [Google Scholar] [CrossRef] [PubMed]
  27. Pawlowski, J.; Kelly-Quinn, M.; Altermatt, F.; Apotheloz-Perret-Gentil, L.; Beja, P.; Boggero, A.; Borja, A.; Bouchez, A.; Cordier, T.; Domaizon, I.; et al. The future of biotic indices in the ecogenomic era: Integrating (e) DNA metabarcoding in biological assessment of aquatic ecosystems. Sci. Total Environ. 2018, 637, 1295–1310. [Google Scholar] [CrossRef] [PubMed]
  28. Van Aggelen, G.; Ankley, G.T.; Baldwin, W.S.; Bearden, D.W.; Benson, W.H.; Chipman, J.K.; Collette, T.W.; Craft, J.A.; Denslow, N.D.; Embry, M.R.; et al. Integrating omic technologies into aquatic ecological risk assessment and environmental monitoring: Hurdles, achievements, and future outlook. Environ. Health Perspect. 2010, 118, 1–5. [Google Scholar] [CrossRef]
  29. Campos, B.; Colbourne, J.K.; Brown, J.B.; Viant, M.R.; Biales, A.D.; Gallagher, K.; Henry, T.R.; Sappington, K.G.; Marshall, S.; Whale, G. How Omics Technologies can Enhance Chemical Safety Regulation: Perspectives from Academia, Government, and Industry. Environ. Toxicol. Chem. 2018, 37, 1252–1259. [Google Scholar] [CrossRef]
  30. Hering, D.; Borja, A.; Jones, J.I.; Pont, D.; Boets, P.; Bouchez, A.; Bruce, K.; Drakare, S.; Hänfling, B.; Kahlert, M.; et al. Implementation options for DNA-based identification into ecological status assessment under the European Water Framework Directive. Water Res. 2018, 138, 192–205. [Google Scholar] [CrossRef]
  31. Tong, W.D.; Ostroff, S.; Blais, B.; Silva, P.; Dubuc, M.; Healy, M.; Slikker, W. Genomics in the land of regulatory science. Regul. Toxicol. Pharmacol. 2015, 72, 102–106. [Google Scholar] [CrossRef] [PubMed]
  32. Garcia-Reyero, N.; Perkins, E.J. Systems biology: Leading the revolution in ecotoxicology. Environ. Toxicol. Chem. 2011, 30, 265–273. [Google Scholar] [CrossRef] [PubMed]
  33. Merrick, B.A. Next generation sequencing data for use in risk assessment. Curr. Opin. Toxicol. 2019, 18, 18–26. [Google Scholar] [CrossRef]
  34. Breitholtz, M.; Ruden, C.; Hansson, S.O.; Bengtsson, B.E. Ten challenges for improved ecotoxicological testing in environmental risk assessment. Ecotoxicol. Environ. Saf. 2006, 63, 324–335. [Google Scholar] [CrossRef]
  35. Ffrench-Constant, R.H. The molecular genetics of insecticide resistance. Genetics 2013, 194, 807–815. [Google Scholar] [CrossRef]
  36. Whitehead, A.; Clark, B.W.; Reid, N.M.; Hahn, M.E.; Nacci, D. When evolution is the solution to pollution: Key principles, and lessons from rapid repeated adaptation of killifish (Fundulus heteroclitus) populations. Evol. Appl. 2017, 10, 762–783. [Google Scholar] [CrossRef]
  37. Bird, A. Perceptions of epigenetics. Nature 2007, 447, 396–398. [Google Scholar] [CrossRef]
  38. Landry, C.R.; Aubin-Horth, N. Recent advances in ecological genomics: From phenotypic plasticity to convergent and adaptive evolution and speciation. In Ecological Genomics. Advances in Experimental Medicine and Biology; Landry, C., Aubin-Horth, N., Eds.; Springer: Dordrecht, The Netherlands, 2013; Volume 781. [Google Scholar] [CrossRef]
  39. Bossdorf, O.; Richards, C.L.; Pigliucci, M. Epigenetics for ecologists. Ecol. Lett. 2008, 11, 106–115. [Google Scholar] [CrossRef]
  40. Hawes, N.A.; Tremblay, L.A.; Pochon, X.; Dunphy, B.; Fidler, A.E.; Smith, K.F. Effects of temperature and salinity stress on DNA methylation in a highly invasive marine invertebrate, the colonial ascidian Didemnum vexillum. Peerj 2018, 6, e5003. [Google Scholar] [CrossRef]
  41. Bromer, J.G.; Zhou, Y.P.; Taylor, M.B.; Doherty, L.; Taylor, H.S. Bisphenol-A exposure in utero leads to epigenetic alterations in the developmental programming of uterine estrogen response. FASEB J. 2010, 24, 2273–2280. [Google Scholar] [CrossRef] [PubMed]
  42. Fragou, D.; Fragou, A.; Kouidou, S.; Njau, S.; Kovatsi, L. Epigenetic mechanisms in metal toxicity. Toxicol. Mech. Methods 2011, 21, 343–352. [Google Scholar] [CrossRef] [PubMed]
  43. Jeremias, G.; Goncalves, F.J.M.; Pereira, J.L.; Asselman, J. Prospects for incorporation of epigenetic biomarkers in human health and environmental risk assessment of chemicals. Biol. Rev. 2020, 95, 822–846. [Google Scholar] [CrossRef]
  44. Hawes, N.A.; Fidler, A.E.; Tremblay, L.A.; Pochon, X.; Dunphy, B.J.; Smith, K.F. Understanding the role of DNA methylation in successful biological invasions: A review. Biol. Invasions 2018, 20, 2285–2300. [Google Scholar] [CrossRef]
  45. Skinner, M.K.; Guerrero-Bosagna, C. Environmental signals and transgenerational epigenetics. Epigenomics 2009, 1, 111–117. [Google Scholar] [CrossRef]
  46. Bonduriansky, R.; Crean, A.J.; Day, T. The implications of nongenetic inheritance for evolution in changing environments. Evol. Appl. 2012, 5, 192–201. [Google Scholar] [CrossRef] [PubMed]
  47. Shaw, J.L.; Judy, J.D.; Kumar, A.; Bertsch, P.; Wang, M.-B.; Kirby, J.K. Incorporating transgenerational epigenetic inheritance into ecological risk assessment frameworks. Environ. Sci. Technol. 2017, 51, 9433–9445. [Google Scholar] [CrossRef] [PubMed]
  48. Head, J.A.; Dolinoy, D.C.; Basu, N. Epigenetics for ecotoxicologists. Environ. Toxicol. Chem. 2012, 31, 221–227. [Google Scholar] [CrossRef]
  49. Srut, M. Ecotoxicological epigenetics in invertebrates: Emerging tool for the evaluation of present and past pollution burden. Chemosphere 2021, 282, 131026. [Google Scholar] [CrossRef]
  50. Vandegehuchte, M.B.; Janssen, C.R. Epigenetics and its implications for ecotoxicology. Ecotoxicology 2011, 20, 607–624. [Google Scholar] [CrossRef]
  51. Plongthongkum, N.; Diep, D.H.; Zhang, K. Advances in the profiling of DNA modifications: Cytosine methylation and beyond. Nat. Rev. Genet. 2014, 15, 647–661. [Google Scholar] [CrossRef]
  52. Schrey, A.W.; Alvarez, M.; Foust, C.M.; Kilvitis, H.J.; Lee, J.D.; Liebl, A.L.; Martin, L.B.; Richards, C.L.; Robertson, M. Ecological Epigenetics: Beyond MS-AFLP. Integr. Comp. Biol. 2013, 53, 340–350. [Google Scholar] [CrossRef]
  53. Guyon, A.; Smith, K.F.; Charry, M.P.; Champeau, O.; Tremblay, L.A. Effects of chronic exposure to benzophenone and diclofenac on DNA methylation levels and reproductive success in a marine copepod. J. Xenobiot. 2018, 8, 7674. [Google Scholar] [CrossRef]
  54. Hofmann, G.E. Ecological Epigenetics in Marine Metazoans. Front. Mar. Sci. 2017, 4, 4. [Google Scholar] [CrossRef]
  55. Oppold, A.; Kreß, A.; Vanden Bussche, J.; Diogo, J.B.; Kuch, U.; Oehlmann, J.; Vandegehuchte, M.B.; Müller, R. Epigenetic alterations and decreasing insecticide sensitivity of the Asian tiger mosquito Aedes albopictus. Ecotoxicol. Environ. Saf. 2015, 122, 45–53. [Google Scholar] [CrossRef]
  56. Schrey, A.W.; Coon, C.A.; Grispo, M.T.; Awad, M.; Imboma, T.; McCoy, E.D.; Mushinsky, H.R.; Richards, C.L.; Martin, L.B. Epigenetic Variation May Compensate for Decreased Genetic Variation with Introductions: A Case Study Using House Sparrows (Passer domesticus) on Two Continents. Genet. Res. Int. 2012, 2012, 979751. [Google Scholar] [CrossRef]
  57. Verhoeven, K.J.F.; Jansen, J.J.; van Dijk, P.J.; Biere, A. Stress-induced DNA methylation changes and their heritability in asexual dandelions. New Phytol. 2010, 185, 1108–1118. [Google Scholar] [CrossRef]
  58. Chatterjee, A.; Ozaki, Y.; Stockwell, P.A.; Horsfield, J.A.; Morison, I.M.; Nakagawa, S. Mapping the zebrafish brain methylome using reduced representation bisulfite sequencing. Epigenetics 2013, 8, 979–989. [Google Scholar] [CrossRef]
  59. Trucchi, E.; Mazzarella, A.B.; Gilfillan, G.D.; Lorenzo, M.T.; Schönswetter, P.; Paun, O. BsRADseq: Screening DNA methylation in natural populations of non-model species. Mol. Ecol. 2016, 25, 1697–1713. [Google Scholar] [CrossRef] [PubMed]
  60. Ramalho-Carvalho, J.; Henrique, R.; Jerónimo, C. Methylation-Specific PCR. In DNA Methylation Protocols; Tost, J., Ed.; Springer: New York, NY, USA, 2018; pp. 447–472. [Google Scholar]
  61. Cokus, S.J.; Feng, S.; Zhang, X.; Chen, Z.; Merriman, B.; Haudenschild, C.D.; Pradhan, S.; Nelson, S.F.; Pellegrini, M.; Jacobsen, S.E. Shotgun bisulphite sequencing of the Arabidopsis genome reveals DNA methylation patterning. Nature 2008, 452, 215–219. [Google Scholar] [CrossRef] [PubMed]
  62. Jurkowski, T.P.; Ravichandran, M.; Stepper, P. Synthetic epigenetics—Towards intelligent control of epigenetic states and cell identity. Clin. Epigenetics 2015, 7, 18. [Google Scholar] [CrossRef] [PubMed]
  63. Laufer, B.I.; Singh, S.M. Strategies for precision modulation of gene expression by epigenome editing: An overview. Epigenetics Chromatin 2015, 8, 34. [Google Scholar] [CrossRef]
  64. Puchta, H. Using CRISPR/Cas in three dimensions: Towards synthetic plant genomes, transcriptomes and epigenomes. Plant J. 2016, 87, 5–15. [Google Scholar] [CrossRef]
  65. Vojta, A.; Dobrinić, P.; Tadić, V.; Bočkor, L.; Korać, P.; Julg, B.; Klasić, M.; Zoldoš, V. Repurposing the CRISPR-Cas9 system for targeted DNA methylation. Nucleic Acids Res. 2016, 44, 5615–5628. [Google Scholar] [CrossRef]
  66. Major, K.M.; DeCourten, B.M.; Li, J.; Britton, M.; Settles, M.L.; Mehinto, A.C.; Connon, R.E.; Brander, S.M. Early Life Exposure to Environmentally Relevant Levels of Endocrine Disruptors Drive Multigenerational and Transgenerational Epigenetic Changes in a Fish Model. Front. Mar. Sci. 2020, 7, 471. [Google Scholar] [CrossRef]
  67. Baettig, C.G.; Laroche, O.; Ockenden, A.; Smith, K.F.; Lear, G.; Tremblay, L.A. Characterization of the transcriptional effects of the plastic additive dibutyl phthalate alone and in combination with microplastic on the green-lipped mussel (Perna canaliculus). Environ. Toxicol. Chem. 2024, 43, 1604–1614. [Google Scholar] [CrossRef]
  68. Schirmer, K.; Fischer, B.B.; Madureira, D.J.; Pillai, S. Transcriptomics in ecotoxicology. Anal. Bioanal. Chem. 2010, 397, 917–923. [Google Scholar] [CrossRef]
  69. Ankley, G.T.; Daston, G.P.; Degitz, S.J.; Denslow, N.D.; Hoke, R.A.; Kennedy, S.W.; Miracle, A.L.; Perkins, E.J.; Snape, J.; Tillitt, D.E. Toxicogenomics in regulatory ecotoxicology. Environ. Sci. Technol. 2006, 40, 4055–4065. [Google Scholar] [CrossRef] [PubMed]
  70. Vinken, M. Omics-based input and output in the development and use of adverse outcome pathways. Curr. Opin. Toxicol. 2019, 18, 8–12. [Google Scholar] [CrossRef]
  71. Taylor, S.C.; Laperriere, G.; Germain, H. Droplet Digital PCR versus qPCR for gene expression analysis with low abundant targets: From variable nonsense to publication quality data. Sci. Rep. 2017, 7, 2409. [Google Scholar] [CrossRef]
  72. Hindson, B.J.; Ness, K.D.; Masquelier, D.A.; Belgrader, P.; Heredia, N.J.; Makarewicz, A.J.; Bright, I.J.; Lucero, M.Y.; Hiddessen, A.L.; Legler, T.C. High-throughput droplet digital PCR system for absolute quantitation of DNA copy number. Anal. Chem. 2011, 83, 8604–8610. [Google Scholar] [CrossRef]
  73. Baettig, C.G.; Zirngibl, M.; Smith, K.F.; Lear, G.; Tremblay, L.A. Comparison between droplet digital PCR and reverse transcription-quantitative PCR methods to measure ecotoxicology biomarkers. Mar. Pollut. Bull. 2023, 190, 114829. [Google Scholar] [CrossRef]
  74. Fabrello, J.; Grapputo, A.; Munari, M.; Marin, M.G.; Masiero, L.; Pacchioni, B.; Millino, C.; Matozzo, V. Molecular and biochemical responses of vitellogenin in the mussel Mytilus galloprovincialis exposed to the glyphosate-based herbicide Roundup® Power 2.0. Environ. Sci. Pollut. Res. 2020, 27, 26543–26553. [Google Scholar] [CrossRef] [PubMed]
  75. Kircher, M.; Kelso, J.J.B. High-throughput DNA sequencing–concepts and limitations. Bioessays 2010, 32, 524–536. [Google Scholar] [CrossRef] [PubMed]
  76. Martin, J.A.; Wang, Z. Next-generation transcriptome assembly. Nat. Rev. Genet. 2011, 12, 671–682. [Google Scholar] [CrossRef] [PubMed]
  77. Sims, D.; Sudbery, I.; Ilott, N.E.; Heger, A.; Ponting, C.P. Sequencing depth and coverage: Key considerations in genomic analyses. Nat. Rev. Genet. 2014, 15, 121–132. [Google Scholar] [CrossRef]
  78. Aebersold, R.; Mann, M. Mass spectrometry-based proteomics. Nature 2003, 422, 198–207. [Google Scholar] [CrossRef]
  79. Cox, J.; Mann, M. Quantitative, High-Resolution Proteomics for Data-Driven Systems Biology. Annu. Rev. Biochem. 2011, 80, 273–299. [Google Scholar] [CrossRef]
  80. Heijne, W.H.M.; Kienhuis, A.S.; van Ommen, B.; Stierum, R.H.; Groten, J.P. Systems toxicology: Applications of toxicogenomics, transcriptomics, proteomics and metabolomics in toxicology. Expert Rev. Proteom. 2005, 2, 767–780. [Google Scholar] [CrossRef]
  81. Cagney, G.; Emili, A. De novo peptide sequencing and quantitative profiling of complex protein mixtures using mass-coded abundance tagging. Nat. Biotechnol. 2002, 20, 163–170. [Google Scholar] [CrossRef]
  82. Mann, M.; Wilm, M. Error tolerant identification of peptides in sequence databases by peptide sequence tags. Anal. Chem. 1994, 66, 4390–4399. [Google Scholar] [CrossRef]
  83. Altelaar, A.F.M.; Munoz, J.; Heck, A.J.R. Next-generation proteomics: Towards an integrative view of proteome dynamics. Nat. Rev. Genet. 2013, 14, 35–48. [Google Scholar] [CrossRef]
  84. Liang, X.; Martyniuk, C.J.; Simmons, D.B.D. Are we forgetting the “proteomics” in multi-omics ecotoxicology? Comp. Biochem. Physiol. Part D Genom. Proteom. 2020, 36, 100751. [Google Scholar] [CrossRef]
  85. Gouveia, D.; Almunia, C.; Cogne, Y.; Pible, O.; Degli-Esposti, D.; Salvador, A.; Cristobal, S.; Sheehan, D.; Chaumot, A.; Geffard, O.; et al. Ecotoxicoproteomics: A decade of progress in our understanding of anthropogenic impact on the environment. J. Proteom. 2019, 198, 66–77. [Google Scholar] [CrossRef]
  86. Trapp, J.; Armengaud, J.; Salvador, A.; Chaumot, A.; Geffard, O. Next-Generation Proteomics: Toward Customized Biomarkers for Environmental Biomonitoring. Environ. Sci. Technol. 2014, 48, 13560–13572. [Google Scholar] [CrossRef] [PubMed]
  87. Groh, K.J.; Dalkvist, T.; Piccapietra, F.; Behra, R.; Suter, M.J.F.; Schirmer, K. Critical influence of chloride ions on silver ion-mediated acute toxicity of silver nanoparticles to zebrafish embryos. Nanotoxicology 2015, 9, 81–91. [Google Scholar] [CrossRef] [PubMed]
  88. Martins, C.; Dreij, K.; Costa, P.M. The state-of-the art of environmental toxicogenomics: Challenges and perspectives of "omics" approaches directed to toxicant mixtures. Int. J. Environ. Res. Public Health 2019, 16, 4718. [Google Scholar] [CrossRef]
  89. Khoo, S.H.G.; Al-Rubeai, M. Metabolomics as a complementary tool in cell culture. Biotechnol. Appl. Biochem. 2007, 47, 71–84. [Google Scholar] [CrossRef]
  90. Nalbantoglu, S.; Abu-Asab, M.; Suy, S.; Collins, S.; Amri, H. Metabolomics-Based Biosignatures of Prostate Cancer in Patients Following Radiotherapy. Omics-A J. Integr. Biol. 2019, 23, 214–223. [Google Scholar] [CrossRef]
  91. Olesti, E.; González-Ruiz, V.; Wilks, M.F.; Boccard, J.; Rudaz, S. Approaches in metabolomics for regulatory toxicology applications. Analyst 2021, 146, 1820–1834. [Google Scholar] [CrossRef]
  92. German, J.B.; Gillies, L.A.; Smilowitz, J.T.; Zivkovic, A.M.; Watkins, S.M. Lipidomics and lipid profiling in metabolomics. Curr. Opin. Lipidol. 2007, 18, 66–71. [Google Scholar] [CrossRef] [PubMed]
  93. Chen, T.; Liu, Y.; Li, M.H.; Xu, H.D.; Sheng, J.Y.; Zhang, L.; Wang, J.S. Integrated 1H NMR-based metabolomics analysis of earthworm responses to sub-lethal Pb exposure. Environ. Chem. 2016, 13, 792–803. [Google Scholar] [CrossRef]
  94. Melvin, S.D.; Habener, L.J.; Leusch, F.D.L.; Carroll, A.R. 1H NMR-based metabolomics reveals sub-lethal toxicity of a mixture of diabetic and lipid-regulating pharmaceuticals on amphibian larvae. Aquat. Toxicol. 2017, 184, 123–132. [Google Scholar] [CrossRef] [PubMed]
  95. Johnson, C.H.; Ivanisevic, J.; Siuzdak, G. Metabolomics: Beyond biomarkers and towards mechanisms. Nat. Rev. Mol. Cell Biol. 2016, 17, 451–459. [Google Scholar] [CrossRef]
  96. Sunshine, H.; Iruela-Arispe, M.L. Membrane lipids and cell signaling. Curr. Opin. Lipidol. 2017, 28, 408–413. [Google Scholar] [CrossRef]
  97. Van Ravenzwaay, B.; Herold, M.; Kamp, H.; Kapp, M.D.; Fabian, E.; Looser, R.; Krennrich, G.; Mellert, W.; Prokoudine, A.; Strauss, V.; et al. Metabolomics: A tool for early detection of toxicological effects and an opportunity for biology based grouping of chemicals-From QSAR to QBAR. Mutat. Res. Genet. Toxicol. Environ. Mutagen. 2012, 746, 144–150. [Google Scholar] [CrossRef]
  98. Patti, G.J.; Yanes, O.; Siuzdak, G. Metabolomics: The apogee of the omics trilogy. Nat. Rev. Mol. Cell Biol. 2012, 13, 263–269. [Google Scholar] [CrossRef]
  99. Melvin, S.D.; Lanctot, C.M.; Doriean, N.J.C.; Carroll, A.R.; Bennett, W.W. Untargeted NMR-based metabolomics for field-scale monitoring: Temporal reproducibility and biomarker discovery in mosquitofish (Gambusia holbrooki) from a metal(loid)-contaminated wetland. Environ. Pollut. 2018, 243, 1096–1105. [Google Scholar] [CrossRef] [PubMed]
  100. Bundy, J.G.; Davey, M.P.; Viant, M.R. Environmental metabolomics: A critical review and future perspectives. Metabolomics 2009, 5, 3–21. [Google Scholar] [CrossRef]
  101. Emwas, A.H.; Luchinat, C.; Turano, P.; Tenori, L.; Roy, R.; Salek, R.M.; Ryan, D.; Merzaban, J.S.; Kaddurah-Daouk, R.; Zeri, A.C.; et al. Standardizing the experimental conditions for using urine in NMR-based metabolomic studies with a particular focus on diagnostic studies: A review. Metabolomics 2015, 11, 872–894. [Google Scholar] [CrossRef]
  102. Miggiels, P.; Wouters, B.; van Westen, G.J.P.; Dubbelman, A.C.; Hankemeier, T. Novel technologies for metabolomics: More for less. TrAC Trends Anal. Chem. 2019, 120, 115323. [Google Scholar] [CrossRef]
  103. Bouhifd, M.; Hartung, T.; Hogberg, H.T.; Kleensang, A.; Zhao, L. Review: Toxicometabolomics. J. Appl. Toxicol. 2013, 33, 1365–1383. [Google Scholar] [CrossRef] [PubMed]
  104. Peregrin-Alvarez, J.M.; Sanford, C.; Parkinson, J. The conservation and evolutionary modularity of metabolism. Genome Biol. 2009, 10, R63. [Google Scholar] [CrossRef]
  105. Griffiths, L.L.; Melvin, S.D.; Connolly, R.M.; Pearson, R.M.; Brown, C.J. Metabolomic indicators for low-light stress in seagrass. Ecol. Indic. 2020, 114, 106316. [Google Scholar] [CrossRef]
  106. Denslow, N.D.; Griffitt, R.J.; Martyniuk, C.J. Advancing the Omics in aquatic toxicology: SETAC North America 31st Annual Meeting. Ecotoxicol. Environ. Saf. 2012, 76, 1–2. [Google Scholar] [CrossRef] [PubMed]
  107. Alecu, I.; Sosa-Miranda, C.D.; Sandhu, J.K.; Bennett, S.A.L.; Cuperlovic-Culf, M. Chapter 12—Cell culture metabolomics and lipidomics. In Metabolomics Perspectives – From Theory to Practical Application; Troisi, J., Ed.; Elsevier: Amsterdam, The Netherlands, 2022; pp. 415–456. [Google Scholar]
  108. Martyniuk, C.J.; Simmons, D.B. Spotlight on environmental omics and toxicology: A long way in a short time. Comp. Biochem. Physiol. D Genom. Proteom. 2016, 19, 97–101. [Google Scholar] [CrossRef] [PubMed]
  109. Bidwell, J.R.; Heath, A.G. An in-situ study of rock bass (Ambloplites-rupestris) physiology—Effect of season and mercury contamination. Hydrobiologia 1993, 264, 137–152. [Google Scholar] [CrossRef]
  110. Beale, D.J.; Jones, O.A.H.; Bose, U.; Broadbent, J.A.; Walsh, T.K.; van de Kamp, J.; Bissett, A. Omics-based ecosurveillance for the assessment of ecosystem function, health, and resilience. Emerg. Top. Life Sci. 2022, 6, 185–199. [Google Scholar] [CrossRef]
  111. Viant, M.R.; Ebbels, T.M.D.; Beger, R.D.; Ekman, D.R.; Epps, D.J.T.; Kamp, H.; Leonards, P.E.G.; Loizou, G.D.; MacRae, J.I.; van Ravenzwaay, B.; et al. Use cases, best practice and reporting standards for metabolomics in regulatory toxicology. Nat. Commun. 2019, 10, 3041. [Google Scholar] [CrossRef]
  112. Beja, O. To BAC or not to BAC: Marine ecogenomics. Curr. Opin. Biotechnol. 2004, 15, 187–190. [Google Scholar] [CrossRef]
  113. Dicke, M.; van Loon, J.J.A.; de Jong, P.W. Ecogenomics benefits community ecology. Science 2004, 305, 618–619. [Google Scholar] [CrossRef]
  114. Maphosa, F.; de Vos, W.M.; Smidt, H. Exploiting the ecogenomics toolbox for environmental diagnostics of organohalide-respiring bacteria. Trends Biotechnol. 2010, 28, 308–316. [Google Scholar] [CrossRef] [PubMed]
  115. Ouborg, N.J.; Vriezen, W.H. An ecologist’s guide to ecogenomics. J. Ecol. 2007, 95, 8–16. [Google Scholar] [CrossRef]
  116. Snoeren, T.A.L.; De Jong, P.W.; Dicke, M. Ecogenomic approach to the role of herbivore-induced plant volatiles in community ecology. J. Ecol. 2007, 95, 17–26. [Google Scholar] [CrossRef]
  117. Faure, D.; Joly, D. 10—Structure and functioning of microbial ecosystems: Metagenomics and integration of omics. In Insight on Environmental Genomics High-Throughput Sequencing; Faure, D., Joly, D., Eds.; Elsevier: Amsterdam, The Netherlands, 2016; pp. 103–113. [Google Scholar]
  118. Pattnaik, S.S.; Busi, S. Rhizospheric Fungi: Diversity and Potential Biotechnological Applications. In Recent Advancement in White Biotechnology Through Fungi, Volume 1: Diversity and Enzymes Perspectives; Yadav, A.N., Mishra, S., Singh, S., Gupta, A., Eds.; Fungal Biology-US; Springer: Cham, Switzerland, 2019; pp. 63–84. [Google Scholar]
  119. Baird, D.J.; Hajibabaei, M. Biomonitoring 2.0: A new paradigm in ecosystem assessment made possible by next-generation DNA sequencing. Mol. Ecol. 2012, 21, 2039–2044. [Google Scholar] [CrossRef]
  120. Ruppert, K.M.; Kline, R.J.; Rahman, M.S. Past, present, and future perspectives of environmental DNA (eDNA) metabarcoding: A systematic review in methods, monitoring, and applications of global eDNA. Global Ecol. Conserv. 2019, 17, e00547. [Google Scholar] [CrossRef]
  121. Shokralla, S.; Spall, J.L.; Gibson, J.F.; Hajibabaei, M. Next-generation sequencing technologies for environmental DNA research. Mol. Ecol. 2012, 21, 1794–1805. [Google Scholar] [CrossRef] [PubMed]
  122. Zimmermann, H.H.; Haròardóttir, S.; Ribeiro, S. Assessing the performance of short 18S rDNA markers for environmental DNA metabarcoding of marine protists. Environ. DNA 2024, 6, e580. [Google Scholar] [CrossRef]
  123. De Vargas, C.; Audic, S.; Henry, N.; Decelle, J.; Mahé, F.; Logares, R.; Lara, E.; Berney, C.; Bescot, N.L.; Probert, I.; et al. Eukaryotic plankton diversity in the sunlit ocean. Science 2015, 348, 1261605. [Google Scholar] [CrossRef]
  124. Lanzen, A.; Lekang, K.; Jonassen, I.; Thompson, E.M.; Troedsson, C. DNA extraction replicates improve diversity and compositional dissimilarity in metabarcoding of eukaryotes in marine sediments. PLoS ONE 2017, 12, e0179443. [Google Scholar] [CrossRef]
  125. Li, W.; Wang, M.M.; Wang, X.G.; Cheng, X.L.; Guo, J.J.; Bian, X.M.; Cai, L. Fungal communities in sediments of subtropical Chinese seas as estimated by DNA metabarcoding. Sci. Rep. 2016, 6, 26528. [Google Scholar] [CrossRef]
  126. Li, Y.; Hingamp, P.; Watai, H.; Endo, H.; Yoshida, T.; Ogata, H. Degenerate PCR primers to reveal the diversity of giant viruses in coastal waters. Viruses 2018, 10, 496. [Google Scholar] [CrossRef]
  127. Pawlowski, J.; Bruce, K.; Panksep, K.; Aguirre, F.I.; Amalfitano, S.; Apothéloz-Perret-Gentil, L.; Baussant, T.; Bouchez, A.; Carugati, L.; Cermakova, K.; et al. Environmental DNA metabarcoding for benthic monitoring: A review of sediment sampling and DNA extraction methods. Sci. Total Environ. 2022, 818, 151783. [Google Scholar] [CrossRef]
  128. Pawlowski, J.; Esling, P.; Lejzerowicz, F.; Cedhagen, T.; Wilding, T.A. Environmental monitoring through protist next-generation sequencing metabarcoding: Assessing the impact of fish farming on benthic foraminifera communities. Mol. Ecol. Resour. 2014, 14, 1129–1140. [Google Scholar] [CrossRef]
  129. Girard, E.B.; Didaskalou, E.A.; Pratama, A.M.A.; Rattner, C.; Morard, R.; Renema, W. Quantitative assessment of reef foraminifera community from metabarcoding data. Mol. Ecol. Resour. 2024, 24, e14000. [Google Scholar] [CrossRef]
  130. Lecroq, B.; Lejzerowicz, F.; Bachar, D.; Christen, R.; Esling, P.; Baerlocher, L.; Osteras, M.; Farinelli, L.; Pawlowski, J. Ultra-deep sequencing of foraminiferal microbarcodes unveils hidden richness of early monothalamous lineages in deep-sea sediments. Proc. Natl. Acad. Sci. USA 2011, 108, 13177–13182. [Google Scholar] [CrossRef]
  131. Miya, M.; Sato, Y.; Fukunaga, T.; Sado, T.; Poulsen, J.Y.; Sato, K.; Minamoto, T.; Yamamoto, S.; Yamanaka, H.; Araki, H.; et al. MiFish, a set of universal PCR primers for metabarcoding environmental DNA from fishes: Detection of more than 230 subtropical marine species. R. Soc. Open Sci. 2015, 2, 150088. [Google Scholar] [CrossRef]
  132. Stoeck, T.; Kochems, R.; Forster, D.; Lejzerowicz, F.; Pawlowski, J. Metabarcoding of benthic ciliate communities shows high potential for environmental monitoring in salmon aquaculture. Ecol. Indic. 2018, 85, 153–164. [Google Scholar] [CrossRef]
  133. Zimmermann, J.; Glockner, G.; Jahn, R.; Enke, N.; Gemeinholzer, B. Metabarcoding vs. morphological identification to assess diatom diversity in environmental studies. Mol. Ecol. Resour. 2015, 15, 526–542. [Google Scholar] [CrossRef] [PubMed]
  134. Ficetola, G.F.; Taberlet, P. Towards exhaustive community ecology via DNA metabarcoding. Mol. Ecol. 2023, 32, 6320–6329. [Google Scholar] [CrossRef]
  135. McGee, K.M.; Robinson, C.V.; Hajibabaei, M. Gaps in DNA-Based Biomonitoring Across the Globe. Front. Ecol. Evol. 2019, 7, 337. [Google Scholar] [CrossRef]
  136. Zaiko, A.; Pochon, X.; Garcia-Vazquez, E.; Olenin, S.; Wood, S.A. Advantages and limitations of environmental DNA/RNA tools for marine biosecurity: Management and surveillance of non-indigenous species. Front. Mar. Sci. 2018, 5, 322. [Google Scholar] [CrossRef]
  137. Darling, J.A.; Galil, B.S.; Carvalho, G.R.; Rius, M.; Viard, F.; Piraino, S. Recommendations for developing and applying genetic tools to assess and manage biological invasions in marine ecosystems. Mar. Policy 2017, 85, 54–64. [Google Scholar] [CrossRef]
  138. Darling, J.A.; Pochon, X.; Abbott, C.L.; Inglis, G.J.; Zaiko, A. The risks of using molecular biodiversity data for incidental detection of species of concern. Divers. Distrib. 2020, 26, 1116–1121. [Google Scholar] [CrossRef]
  139. Kelly, R.P.; Port, J.A.; Yamahara, K.M.; Crowder, L.B. Using environmental DNA to census marine fishes in a large mesocosm. PLoS ONE 2014, 9, e86175. [Google Scholar] [CrossRef]
  140. Zhang, M.L.; Zou, Y.T.; Xiao, S.; Hou, J. Environmental DNA metabarcoding serves as a promising method for aquatic species monitoring and management: A review focused on its workflow, applications, challenges and prospects. Mar. Pollut. Bull. 2023, 194, 115430. [Google Scholar] [CrossRef] [PubMed]
  141. Birrer, S.C.; Dafforn, K.A.; Simpson, S.L.; Kelaher, B.P.; Potts, J.; Scanes, P.; Johnston, E.L. Interactive effects of multiple stressors revealed by sequencing total (DNA) and active (RNA) components of experimental sediment microbial communities. Sci. Total Environ. 2018, 637, 1383–1394. [Google Scholar] [CrossRef] [PubMed]
  142. Keeley, N.; Wood, S.A.; Pochon, X. Development and preliminary validation of a multi-trophic metabarcoding biotic index for monitoring benthic organic enrichment. Ecol. Indic. 2018, 85, 1044–1057. [Google Scholar] [CrossRef]
  143. Laroche, O.; Pochon, X.; Tremblay, L.A.; Ellis, J.I.; Lear, G.; Wood, S.A. Incorporating molecular-based functional and co-occurrence network properties into benthic marine impact assessments. FEMS Microbiol. Ecol. 2018, 94, fiy167. [Google Scholar] [CrossRef] [PubMed]
  144. Laroche, O.; Wood, S.A.; Tremblay, L.A.; Ellis, J.I.; Lear, G.; Pochon, X. A cross-taxa study using environmental DNA/RNA metabarcoding to measure biological impacts of offshore oil and gas drilling and production operations. Mar. Pollut. Bull. 2018, 127, 97–107. [Google Scholar] [CrossRef]
  145. Compson, Z.G.; McClenaghan, B.; Singer, G.A.C.; Fahner, N.A.; Hajibabaei, M. Metabarcoding from microbes to mammals: Comprehensive bioassessment on a global scale. Front. Ecol. Evol. 2020, 8, 581835. [Google Scholar] [CrossRef]
  146. Cordier, T.; Alonso-Saez, L.; Apothéloz-Perret-Gentil, L.; Aylagas, E.; Bohan, D.A.; Bouchez, A.; Chariton, A.; Creer, S.; Frühe, L.; Keck, F.; et al. Ecosystems monitoring powered by environmental genomics: A review of current strategies with an implementation roadmap. Mol. Ecol. 2021, 30, 2937–2958. [Google Scholar] [CrossRef]
  147. Pawlowski, J.; Lejzerowicz, F.; Apotheloz-Perret-Gentil, L.; Visco, J.; Esling, P. Protist metabarcoding and environmental biomonitoring: Time for change. Eur. J. Protistol. 2016, 55, 12–25. [Google Scholar] [CrossRef]
  148. Takahashi, M.; Saccò, M.; Kestel, J.H.; Nester, G.; Campbell, M.A.; van der Heyde, M.; Heydenrych, M.J.; Juszkiewicz, D.J.; Nevill, P.; Dawkins, K.L.; et al. Aquatic environmental DNA: A review of the macro-organismal biomonitoring revolution. Sci. Total Environ. 2023, 873, 162322. [Google Scholar] [CrossRef]
  149. Dowle, E.J.; Pochon, X.; Banks, J.C.; Shearer, K.; Wood, S.A. Targeted gene enrichment and high-throughput sequencing for environmental biomonitoring: A case study using freshwater macroinvertebrates. Mol. Ecol. Resour. 2016, 16, 1240–1254. [Google Scholar] [CrossRef]
  150. Frühe, L.; Dully, V.; Forster, D.; Keeley, N.B.; Laroche, O.; Pochon, X.; Robinson, S.; Wilding, T.A.; Stoeck, T. Global trends of benthic bacterial diversity and community composition along organic enrichment gradients of salmon farms. Front. Microbiol. 2021, 12, 637811. [Google Scholar] [CrossRef] [PubMed]
  151. Pochon, X.; Wood, S.A.; Keeley, N.B.; Lejzerowicz, F.; Esling, P.; Drew, J.; Pawlowski, J. Accurate assessment of the impact of salmon farming on benthic sediment enrichment using foraminiferal metabarcoding. Mar. Pollut. Bull. 2015, 100, 370–382. [Google Scholar] [CrossRef]
  152. He, X.P.; Sutherland, T.F.; Pawlowski, J.; Abbott, C.L. Responses of foraminifera communities to aquaculture-derived organic enrichment as revealed by environmental DNA metabarcoding. Mol. Ecol. 2019, 28, 1138–1153. [Google Scholar] [CrossRef]
  153. Lejzerowicz, F.; Esling, P.; Pillet, L.; Wilding, T.A.; Black, K.D.; Pawlowski, J. High-throughput sequencing and morphology perform equally well for benthic monitoring of marine ecosystems. Sci. Rep. 2015, 5, 13932. [Google Scholar] [CrossRef]
  154. Leontidou, K.; Rubel, V.; Stoeck, T. Comparing quantile regression spline analyses and supervised machine learning for environmental quality assessment at coastal marine aquaculture installations. Peerj 2023, 11, e15425. [Google Scholar] [CrossRef] [PubMed]
  155. Smith, M.B.; Rocha, A.M.; Smillie, C.S.; Olesen, S.W.; Paradis, C.; Wu, L.Y.; Campbell, J.H.; Fortney, J.L.; Mehlhorn, T.L.; Lowe, K.A.; et al. Natural bacterial communities serve as quantitative geochemical biosensors. Mbio 2015, 6, 10–1128. [Google Scholar] [CrossRef]
  156. Dutra, J.; García, G.; Gomes, R.; Cardoso, M.; Côrtes, A.; Silva, T.; de Jesus, L.; Rodrigues, L.; Freitas, A.; Waldow, V.; et al. Effective Biocorrosive Control in Oil Industry Facilities: 16S rRNA Gene metabarcoding for monitoring microbial communities in produced water. Microorganisms 2023, 11, 846. [Google Scholar] [CrossRef] [PubMed]
  157. Laroche, O.; Wood, S.A.; Tremblay, L.A.; Ellis, J.I.; Lejzerowicz, F.; Pawlowski, J.; Lear, G.; Atalah, J.; Pochon, X. First evaluation of foraminiferal metabarcoding for monitoring environmental impact from an offshore oil drilling site. Mar. Environ. Res. 2016, 120, 225–235. [Google Scholar] [CrossRef]
  158. Lanzén, A.; Lekang, K.; Jonassen, I.; Thompson, E.M.; Troedsson, C. High-throughput metabarcoding of eukaryotic diversity for environmental monitoring of offshore oil-drilling activities. Mol. Ecol. 2016, 25, 4392–4406. [Google Scholar] [CrossRef]
  159. Laroche, O.; Wood, S.A.; Tremblay, L.A.; Lear, G.; Ellis, J.I.; Pochon, X. Metabarcoding monitoring analysis: The pros and cons of using co-extracted environmental DNA and RNA data to assess offshore oil production impacts on benthic communities. Peerj 2017, 5, e3347. [Google Scholar] [CrossRef] [PubMed]
  160. Mauffrey, F.; Cordier, T.; Apothéloz-Perret-Gentil, L.; Cermakova, K.; Merzi, T.; Delefosse, M.; Blanc, P.; Pawlowski, J. Benthic monitoring of oil and gas offshore platforms in the North Sea using environmental DNA metabarcoding. Mol. Ecol. 2021, 30, 3007–3022. [Google Scholar] [CrossRef] [PubMed]
  161. Pawlowski, J.; Cermakova, K.; Cordier, T.; Frontalini, F.; Apothéloz-Perret-Gentil, L.; Merzi, T. Assessing the potential of nematode metabarcoding for benthic monitoring of offshore oil platforms. Sci. Total Environ. 2024, 933, 173092. [Google Scholar] [CrossRef]
  162. Alexander, J.B.; Marnane, M.J.; Elsdon, T.S.; Bunce, M.; Sitaworawet, P.; Songploy, S.; Chaiyakul, S.; Harvey, E.S. Using environmental DNA to better inform decision making around decommissioning alternatives for offshore oil and gas infrastructure. Sci. Total Environ. 2023, 901, 165991. [Google Scholar] [CrossRef]
  163. Chariton, A.A.; Stephenson, S.; Morgan, M.J.; Steven, A.D.L.; Colloff, M.J.; Court, L.N.; Hardy, C.M. Metabarcoding of benthic eukaryote communities predicts the ecological condition of estuaries. Environ. Pollut. 2015, 203, 165–174. [Google Scholar] [CrossRef]
  164. Tagliabue, A.; Matterson, K.O.; Ponti, M.; Turicchia, E.; Abbiati, M.; Costantini, F. Sediment and bottom water eDNA metabarcoding to support coastal management. Ocean Coast. Manag. 2023, 244, 106785. [Google Scholar] [CrossRef]
  165. Avó, A.P.; Daniell, T.J.; Neilson, R.; Oliveira, S.; Branco, J.; Adao, H. DNA barcoding and morphological identification of benthic nematodes assemblages of estuarine intertidal sediments: Advances in molecular tools for biodiversity assessment. Front. Mar. Sci. 2017, 4, 66. [Google Scholar] [CrossRef]
  166. Aylagas, E.; Borja, A.; Muxika, I.; Rodríguez-Ezpeleta, N. Adapting metabarcoding-based benthic biomonitoring into routine marine ecological status assessment networks. Ecol. Indic. 2018, 95, 194–202. [Google Scholar] [CrossRef]
  167. Rey, A.; Viard, F.; Lizé, A.; Corre, E.; Valentini, A.; Thiriet, P. Coastal rocky reef fish monitoring in the context of the Marine Strategy Framework Directive: Environmental DNA metabarcoding complements underwater visual census. Ocean Coast. Manag. 2023, 241, 106625. [Google Scholar] [CrossRef]
  168. Kapshyna, I.; Veit-Koehler, G.; Hoffman, L.; Khodami, S. Impact of a coastal protection measure on sandy-beach meiofauna at Ahrenshoop (Baltic Sea, Germany): Results from metabarcoding and morphological approaches are similar. Metabarcoding Metagenomics 2024, 8, e127688. [Google Scholar] [CrossRef]
  169. Aylagas, E.; Borja, A.; Rodríguez-Ezpeleta, N. Environmental status assessment using DNA metabarcoding: Towards a genetics based marine biotic index (gAMBI). PLoS ONE 2014, 9, e90529. [Google Scholar] [CrossRef]
  170. Wangensteen, O.S.; Palacín, C.; Guardiola, M.; Turon, X. DNA metabarcoding of littoral hard-bottom communities: High diversity and database gaps revealed by two molecular markers. Peerj 2018, 6, e4705. [Google Scholar] [CrossRef] [PubMed]
  171. Castro-Cubillos, M.L.; Taylor, J.D.; Mastretta-Yanes, A.; Benítez-Villalobos, F.; Islas-Villanueva, V. Monitoring of benthic eukaryotic communities in two tropical coastal lagoons through eDNA metabarcoding: A spatial and temporal approximation. Sci. Rep. 2022, 12, 10089. [Google Scholar] [CrossRef] [PubMed]
  172. Taberlet, P.; Bonin, A.; Zinger, L.; Coissac, E. Environmental DNA: For Biodiversity Research and Monitoring; Oxford University Press: Oxford, UK, 2018. [Google Scholar]
  173. Zhou, X.; Li, Y.Y.; Liu, S.L.; Yang, Q.; Su, X.; Zhou, L.L.; Tang, M.; Fu, R.B.; Li, J.G.; Huang, Q.F. Ultra-deep sequencing enables high-fidelity recovery of biodiversity for bulk arthropod samples without PCR amplification. Gigascience 2013, 2, 2047-217X-2-4. [Google Scholar] [CrossRef]
  174. Mertes, F.; ElSharawy, A.; Sauer, S.; van Helvoort, J.; van der Zaag, P.J.; Franke, A.; Nilsson, M.; Lehrach, H.; Brookes, A.J. Targeted enrichment of genomic DNA regions for next-generation sequencing. Brief. Funct. Genom. 2011, 10, 374–386. [Google Scholar] [CrossRef]
  175. Nota, K.; Orlando, L.; Marchesini, A.; Girardi, M.; Bertilsson, S.; Vernesi, C.; Parducci, L. Enriching barcoding markers in environmental samples utilizing a phylogenetic probe design: Insights from mock communities. Environ. DNA 2024, 6, e593. [Google Scholar] [CrossRef]
  176. Giarla, T.C.; Esselstyn, J.A. The challenges of resolving a rapid, recent radiation: Empirical and simulated phylogenomics of philippine shrews. Syst. Biol. 2015, 64, 727–740. [Google Scholar] [CrossRef]
  177. Horn, S. Target enrichment via DNA hybridization capture. In Ancient DNA: Methods and Protocols; Shapiro, B., Hofreiter, M., Eds.; Humana Press: Totowa, NJ, USA, 2012; pp. 177–188. [Google Scholar]
  178. Borsetto, C.; Raguideau, S.; Travis, E.; Kim, D.W.; Lee, D.H.; Bottrill, A.; Stark, R.; Song, L.J.; Cha, C.J.; Pearson, J.; et al. Impact of sulfamethoxazole on a riverine microbiome. Water Res. 2021, 201, 117382. [Google Scholar] [CrossRef]
  179. Chen, W.J.; Wang, Z.G.; Xu, W.H.; Tian, R.M.; Zeng, J. Dibutyl phthalate contamination accelerates the uptake and metabolism of sugars by microbes in black soil. Environ. Pollut. 2020, 262, 114332. [Google Scholar] [CrossRef]
  180. Yu, K.; Yi, S.; Li, B.; Guo, F.; Peng, X.X.; Wang, Z.P.; Wu, Y.; Alvarez-Cohen, L.; Zhang, T. An integrated meta-omics approach reveals substrates involved in synergistic interactions in a bisphenol A (BPA)-degrading microbial community. Microbiome 2019, 7, 16. [Google Scholar] [CrossRef]
  181. Schiml, V.C.; Delogu, F.; Kumar, P.; Kunath, B.; Batut, B.; Mehta, S.; Johnson, J.E.; Grüning, B.; Pope, P.B.; Jagtap, P.D.; et al. Integrative meta-omics in Galaxy and beyond. Environ. Microbiome 2023, 18, 56. [Google Scholar] [CrossRef]
  182. Segata, N.; Boernigen, D.; Tickle, T.L.; Morgan, X.C.; Garrett, W.S.; Huttenhower, C. Computational meta’omics for microbial community studies. Mol. Syst. Biol. 2013, 9, 666. [Google Scholar] [CrossRef]
  183. Michealsamy, A.; Thangamani, L.; Manivel, G.; Kumar, P.; Sundar, S.; Piramanayagam, S.; Natarajan, J. Current research and applications of meta-omics stratagems in bioremediation: A bird’s-eye view. J. Appl. Biotechnol. Rep. 2021, 8, 109–115. [Google Scholar] [CrossRef]
  184. Jeddi, M.Z.; Hopf, N.B.; Viegas, S.; Price, A.B.; Paini, A.; van Thriel, C.; Benfenati, E.; Ndaw, S.; Bessems, J.; Behnisch, P.A.; et al. Towards a systematic use of effect biomarkers in population and occupational biomonitoring. Environ. Int. 2021, 146, 106257. [Google Scholar] [CrossRef]
  185. Brockmeier, E.K.; Hodges, G.; Hutchinson, T.H.; Butler, E.; Hecker, M.; Tollefsen, K.E.; Garcia-Reyero, N.; Kille, P.; Becker, D.; Chipman, K.; et al. The role of omics in the application of Adverse Outcome Pathways for chemical risk assessment. Toxicol. Sci. 2017, 158, 252–262. [Google Scholar] [CrossRef] [PubMed]
  186. Gatzidou, E.T.; Zira, A.N.; Theocharis, S.E. Toxicogenomics: A pivotal piece in the puzzle of toxicological research. J. Appl. Toxicol. 2007, 27, 302–309. [Google Scholar] [CrossRef] [PubMed]
  187. Sauer, U.G.; Deferme, L.; Gribaldo, L.; Hackermüller, J.; Tralau, T.; van Ravenzwaay, B.; Yauk, C.; Poole, A.; Tong, W.D.; Gant, T.W. The challenge of the application of ‘omics technologies in chemicals risk assessment: Background and outlook. Regul. Toxicol. Pharmacol. 2017, 91, S14–S26. [Google Scholar] [CrossRef]
  188. Pinu, F.R.; Beale, D.J.; Paten, A.M.; Kouremenos, K.; Swarup, S.; Schirra, H.J.; Wishart, D. Systems biology and multi-omics integration: Viewpoints from the metabolomics research community. Metabolites 2019, 9, 76. [Google Scholar] [CrossRef] [PubMed]
  189. Canzler, S.; Schor, J.; Busch, W.; Schubert, K.; Rolle-Kampczyk, U.E.; Seitz, H.; Kamp, H.; von Bergen, M.; Buesen, R.; Hackermuller, J. Prospects and challenges of multi-omics data integration in toxicology. Arch. Toxicol. 2020, 94, 371–388. [Google Scholar] [CrossRef] [PubMed]
  190. Buesen, R.; Chorley, B.N.; da Silva Lima, B.; Daston, G.; Deferme, L.; Ebbels, T.; Gant, T.W.; Goetz, A.; Greally, J.; Gribaldo, L.; et al. Applying ‘omics technologies in chemicals risk assessment: Report of an ECETOC workshop. Regul. Toxicol. Pharmacol. 2017, 91, S3–S13. [Google Scholar] [CrossRef]
  191. Tralau, T.; Oelgeschläger, M.; Gürtler, R.; Heinemeyer, G.; Herzler, M.; Höfer, T.; Itter, H.; Kuhl, T.; Lange, N.; Lorenz, N.; et al. Regulatory toxicology in the twenty-first century: Challenges, perspectives and possible solutions. Arch. Toxicol. 2015, 89, 823–850. [Google Scholar] [CrossRef]
  192. Escher, B.I.; Hackermuller, J.; Polte, T.; Scholz, S.; Aigner, A.; Altenburger, R.; Bohme, A.; Bopp, S.K.; Brack, W.; Busch, W.; et al. From the exposome to mechanistic understanding of chemical-induced adverse effects. Environ. Int. 2017, 99, 97–106. [Google Scholar] [CrossRef]
  193. Scala, G.; Kinaret, P.; Marwah, V.; Sund, J.; Fortino, V.; Greco, D. Multi-omics analysis of ten carbon nanomaterials effects highlights cell type specific patterns of molecular regulation and adaptation. NanoImpact 2018, 11, 99–108. [Google Scholar] [CrossRef] [PubMed]
  194. Wang, P.; Ng, Q.X.; Zhang, B.; Wei, Z.; Hassan, M.; He, Y.; Ong, C.N. Employing multi-omics to elucidate the hormetic response against oxidative stress exerted by nC60 on Daphnia pulex. Environ. Pollut. 2019, 251, 22–29. [Google Scholar] [CrossRef] [PubMed]
  195. Larras, F.; Billoir, E.; Scholz, S.; Tarkka, M.; Wubet, T.; Delignette-Muller, M.L.; Schmitt-Jansen, M. A multi-omics concentration-response framework uncovers novel understanding of triclosan effects in the chlorophyte Scenedesmus vacuolatus. J. Hazard. Mater. 2020, 397, 122727. [Google Scholar] [CrossRef]
  196. Jamers, A.; Blust, R.; De Coen, W.; Griffin, J.L.; Jones, O.A.H. An omics based assessment of cadmium toxicity in the green alga Chlamydomonas reinhardtii. Aquat. Toxicol. 2013, 126, 355–364. [Google Scholar] [CrossRef]
  197. Wang, J.; Li, C.-L.; Tu, B.-J.; Yang, K.; Mo, T.-T.; Zhang, R.-Y.; Cheng, S.-Q.; Chen, C.-Z.; Jiang, X.-J.; Han, T.-L.; et al. Integrated Epigenetics, Transcriptomics, and Metabolomics to Analyze the Mechanisms of Benzo[a]pyrene Neurotoxicity in the Hippocampus. Toxicol. Sci. 2018, 166, 65–81. [Google Scholar] [CrossRef]
  198. Zhao, X.; Jiang, W.Y.; Liu, J.J.; Yan, W.J.; Zhang, Y.H.; Zhang, Z.; Zhang, P.D. Unraveling the impact of PFOA toxicity on Zostera marina using a multi-omics approach: Insights from growth, physiological, transcriptomic, and metabolomic signatures. J. Hazard. Mater. 2025, 486, 137024. [Google Scholar] [CrossRef]
  199. Misra, B.B.; Langefeld, C.; Olivier, M.; Cox, L.A. Integrated omics: Tools, advances and future approaches. J. Mol. Endocrinol. 2019, 62, R21–R45. [Google Scholar] [CrossRef] [PubMed]
  200. Bersanelli, M.; Mosca, E.; Remondini, D.; Giampieri, E.; Sala, C.; Castellani, G.; Milanesi, L. Methods for the integration of multi-omics data: Mathematical aspects. BMC Bioinform. 2016, 17, S15. [Google Scholar] [CrossRef]
  201. Buescher, J.M.; Driggers, E.M. Integration of omics: More than the sum of its parts. Cancer Metab. 2016, 4, 4. [Google Scholar] [CrossRef] [PubMed]
  202. Huang, S.; Chaudhary, K.; Garmire, L.X. More is better: Recent progress in multi-omics data integration methods. Front. Genet. 2017, 8, 84. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Main criteria for a framework to conduct field bioassessment must include a range of responses across levels of biological organisation. Omics approaches can generate information on toxicological mechanisms to confirm cause-and-effect relationships and better characterise the ecological impacts linked to exposures to stressors. Omics can provide information on the hazards of pollutants from molecules to whole organisms and community levels.
Figure 1. Main criteria for a framework to conduct field bioassessment must include a range of responses across levels of biological organisation. Omics approaches can generate information on toxicological mechanisms to confirm cause-and-effect relationships and better characterise the ecological impacts linked to exposures to stressors. Omics can provide information on the hazards of pollutants from molecules to whole organisms and community levels.
Toxics 13 00714 g001
Table 1. A list of selected studies focused on marine biomonitoring using metabarcoding to assess the impacts of industry/anthropologically-derived stressors since 2015.
Table 1. A list of selected studies focused on marine biomonitoring using metabarcoding to assess the impacts of industry/anthropologically-derived stressors since 2015.
Stressor(s)Biological Group(s)Marker(s) (Marker Region)Application(s)/CountryReference
Salmon AquacultureBacteria16S (V4 region) ribosomal DNA (rDNA)Monitoring benthic impacts/New Zealand[149]
Bacteria16S (V4) rDNAMonitoring benthic impacts/Norway[132]
Bacteria16S (V3-V4) rDNAMonitoring benthic impacts/international[150]
Foraminifera18S (27F) rDNAMonitoring benthic impacts/New Zealand[151]
Foraminifera18S (27F) rDNAMonitoring benthic impacts/Norway[146]
Foraminifera18S (27F) rDNAMonitoring benthic impacts/Canada[152]
Ciliates18S (V9) rDNAMonitoring benthic impacts/Scotland[132]
Eukaryotes18S (V4) rDNAMonitoring benthic impacts/Scotland[153]
Multi-trophic16S (V4) rDNA, 18S (27F) rDNA, 18S (V4) rDNAMonitoring benthic impacts/New Zealand[142]
Multi-trophic16S (V3-V4) rDNA, 18S (V3-V4) rDNAMonitoring benthic impacts/Norway[154]
Offshore Oil and GasBacteria16S (V4) rDNAOil spill impacts/Gulf of Mexico[155]
Bacteria16S (V3-V4) rDNABiocorrosive control/Brazil[156]
Foraminifera18S (27F region) rDNAMonitoring benthic impacts/New Zealand[157]
Prokaryotes, Eukaryotes18S (V4-V5) rDNAMonitoring benthic impacts/Norway[158]
Multi-trophic16S (V4) rDNA, 18S (V4) rDNAMonitoring benthic impacts/New Zealand[159]
Multi-trophic18S (V1-V2; V9) rDNA, COIMonitoring benthic impacts/North Sea (Denmark)[160]
Multi-trophic18S (V1-V2; V9) rDNAMonitoring benthic impacts/North Sea (Denmark)[161]
Multi-trophicITS2, mitochondrial 16S, COIMonitoring old platforms/Gulf of Thailand[162]
Coastal DevelopmentEukaryotes18S (V1-V2) rDNAExcess organic enrichment Estuaries/Australia[163]
EukaryotesCOIMetabarcoding for management/Italy[164]
Nematodes18S (V1-V2) rDNA, COIMorpho-Molecular comparison/Estuary (Portugal)[165]
MacroinvertebratesCOIMonitoring network/Basque Coast (Spain)[166]
Teleost FishMitochondrial 16SCoastal rocky reef fish monitoring/France[167]
Meiofauna18S (V1-V2) rDNAProtected sandy beach/Germany[168]
Multi-trophic18S rDNA and COIMetabarcoding-based gAMBI index/Spain[169]
Multi-trophic18S (V7) rDNA, COIHard-Bottom Impacts/Spain[170]
Multi-trophic16S (V1-V3) rDNA, 18S (V9) rDNAEffects of multiple stressors/Australia[141]
Multi-trophic18S (V4) rDNA, COITropical coastal lagoon/Mexico[171]
rDNA: ribosomal DNA; COI: cytochrome oxidase I; ITS2: internal transcribed spacer.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Smith, K.F.; Pochon, X.; Melvin, S.D.; Wheeler, T.T.; Tremblay, L.A. Integration of “Omics”-Based Approaches in Environmental Risk Assessment to Establish Cause and Effect Relationships: A Review. Toxics 2025, 13, 714. https://doi.org/10.3390/toxics13090714

AMA Style

Smith KF, Pochon X, Melvin SD, Wheeler TT, Tremblay LA. Integration of “Omics”-Based Approaches in Environmental Risk Assessment to Establish Cause and Effect Relationships: A Review. Toxics. 2025; 13(9):714. https://doi.org/10.3390/toxics13090714

Chicago/Turabian Style

Smith, Kirsty F., Xavier Pochon, Steven D. Melvin, Thomas T. Wheeler, and Louis A. Tremblay. 2025. "Integration of “Omics”-Based Approaches in Environmental Risk Assessment to Establish Cause and Effect Relationships: A Review" Toxics 13, no. 9: 714. https://doi.org/10.3390/toxics13090714

APA Style

Smith, K. F., Pochon, X., Melvin, S. D., Wheeler, T. T., & Tremblay, L. A. (2025). Integration of “Omics”-Based Approaches in Environmental Risk Assessment to Establish Cause and Effect Relationships: A Review. Toxics, 13(9), 714. https://doi.org/10.3390/toxics13090714

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop