Microbial Bioactives

Microbial Bioactives | Online ISSNĀ 2209-2161
279
Citations
152.3k
Views
143
Articles
Your new experience awaits. Try the new design now and help us make it even better
Switch to the new experience
Figures and Tables
REVIEWS   (Open Access)

Illuminating Biological Dark Matter: Integrating Metagenomics, Synthetic Biology, and AI to Unlock Microbial and Genomic Potential for Therapeutics and Biotechnology

Yue Li 1, Shunqi Liu 2 *

+ Author Affiliations

Microbial Bioactives 9 (1) 1-8 https://doi.org/10.25163/microbbioacts.9110627

Submitted: 13 February 2026 Revised: 01 April 2026  Published: 10 April 2026 


Abstract

The exploration of microbial and human genomic "dark matter" has transformed biotechnology, shifting the focus from merely reading genetic codes to actively engineering and harnessing them for sustainable solutions and human health. Over 99% of microorganisms remain uncultured, representing vast reservoirs of novel natural products (NPs) and enzymes that can be accessed through culture-independent metagenomics. Function-based, sequencing-based, and single-cell metagenomic approaches enable the discovery of bioactive compounds such as turbomycins, fasamycins, and cadasides, which hold promise against multidrug-resistant pathogens. Parallel advances in synthetic biology have established robust chassis organisms, including Saccharomyces cerevisiae and fast-growing cyanobacteria, optimized for industrial production of biofuels, chemicals, and bioplastics. Artificial intelligence (AI) and machine learning further refine these platforms, providing predictive models for bioprocess optimization, biomass accumulation, and metabolic engineering. In clinical contexts, proteogenomics integrates DNA, RNA, and protein-level data to identify therapeutic targets and overcome drug resistance in diseases such as colorectal cancer. The ongoing evolution of HIV-1 illustrates the challenge of viral diversity, highlighting the role of next-generation sequencing, CRISPR-based gene editing, and biosensor-enabled surveillance in precision medicine. Gut microbiota manipulation, through fecal microbiota transplantation and engineered probiotics, represents a frontier for addressing systemic and metabolic disorders. This systematic review synthesizes quantitative data from these diverse fields, emphasizing the synergy between metagenomics, synthetic biology, and AI, and provides a meta-analytic framework to evaluate their translational potential for therapeutics, industrial biotechnology, and personalized medicine.

Keywords: Metagenomics; Synthetic Biology; Artificial Intelligence; Microbial Dark Matter; Proteogenomics; HIV-1; Gut Microbiota; Natural Products

1. Introduction

The dawn of the 21st century has witnessed an unprecedented transformation in biotechnology, moving far beyond the simplistic reading of genetic codes to the active design, editing, and engineering of biological systems to address pressing global challenges. Central to this shift is the exploration of biological dark matter, a term used to describe the vast majority of microorganisms that remain uncultured in conventional laboratory settings. These uncultured microbes, which constitute more than 99% of microbial diversity, harbor extraordinary metabolic potential that is largely untapped by traditional methods (Alam et al., 2021). Unlocking this potential requires culture-independent, high-resolution approaches such as metagenomics, which offer function-based, sequencing-based, and single-cell strategies to identify novel natural products and enzymes capable of revolutionizing drug discovery, biofuel production, and industrial biocatalysis (Alam et al., 2021).

Function-based metagenomics enables researchers to clone environmental DNA into expression vectors and screen for specific biochemical activities in heterologous hosts. This methodology stands out because it does not rely on prior knowledge of gene sequences, allowing the discovery of entirely new genes and enzymatic pathways with unknown functions (Alam et al., 2021). Sequencing-based metagenomics, on the other hand, harnesses next-generation sequencing technologies and sophisticated bioinformatic platforms such as eSNaPD to detect and analyze biosynthetic gene clusters, predicting the chemical structures of previously unidentified metabolites (Alam et al., 2021). Complementing these approaches, single-cell metagenomics isolates individual genomes from complex microbial communities, providing precise taxonomic assignments and direct links between metabolic functions and specific organisms (Alam et al., 2021). Together, these strategies allow researchers to chart the uncharted microbial universe, akin to a biological deep-sea sonar mapping the vast, invisible ocean of microbial diversity.

The exploration of microbial dark matter has already yielded remarkable discoveries. Compounds such as turbomycins, fasamycins, and terragines have emerged from metagenomic libraries, while specialized metabolites including isocyanides and cadasides have demonstrated potent activity against multidrug-resistant pathogens (Alam et al., 2021; Wu et al., 2019). Symbiotic marine microorganisms have produced antitumor agents such as patellamide D, ascidiacyclamide, bryostatin, pederin, and onnamide, illustrating the untapped pharmacological potential of uncultured microbes (Hildebrand et al., 2004; Piel, 2002; Piel et al., 2004). These successes underscore the transformative role of metagenomics as both a discovery platform and a predictive framework for therapeutic innovation.

Parallel to microbial exploration, the concept of genomic dark matter extends to human genetics. Endogenous retroviruses, which comprise roughly 8% of the human genome, were long considered evolutionary relics but are now recognized as crucial regulators of immune function and potential biomarkers for cancer prognosis and immunotherapy (Felley-Bosco, 2023; Hoyt et al., 2022). The integration of proteogenomics—linking DNA and RNA variations with actual protein expression—enhances understanding of functional phenotypes in diseases such as colorectal cancer, enabling precision oncology approaches that anticipate drug resistance and identify novel therapeutic targets (Blank-Landeshammer et al., 2019; Mertins et al., 2016; Zhang et al., 2014).

The field of synthetic biology has emerged as a key enabler of these discoveries, translating genomic insights into functional applications. Saccharomyces cerevisiae, historically a model organism for fermentation, has evolved into the first eukaryote with a chemically synthesized genome, demonstrating the power of genome writing and editing (Dixon & Pretorius, 2020). Cyanobacteria have similarly been developed as ā€œgreen Escherichia coli,ā€ photosynthetic chassis capable of converting solar energy and carbon dioxide into biofuels, high-value chemicals, and biodegradable polymers such as polyhydroxyalkanoates (Liu et al., 2024; Luan et al., 2020; Yu et al., 2015). Fast-growing strains such as Synechococcus elongatus UTEX 2973 offer high biomass productivity, positioning cyanobacteria as industrially scalable platforms for sustainable biomanufacturing (Liu et al., 2024; Yu et al., 2015).

Artificial intelligence and machine learning are increasingly integrated into these biotechnological frameworks, optimizing bioprocess parameters, predicting metabolic bottlenecks, and enabling real-time monitoring through intelligent sensing systems (Fu et al., 2023; Imamoglu, 2024; Long et al., 2022). In microalgae-based bioprocessing, machine-learning models such as artificial neural networks and random forests have enhanced species classification, biomass prediction, and metabolic regulation, achieving accuracies exceeding 95% in large-scale cultivation studies (Kavitha et al., 2024; Oruganti et al., 2023; Peter et al., 2023). Wearable biosensors further extend these innovations into clinical contexts by enabling continuous monitoring of biomarkers in sweat, tears, and saliva, bridging environmental biotechnology and personalized medicine (Sempionatto et al., 2019; Xu et al., 2020; Zhang et al., 2023).

The challenge of viral evolution, particularly HIV-1, illustrates the critical need for precision genomic surveillance and rapid adaptive technologies. HIV-1 exhibits extraordinary genetic diversity due to high mutation rates and frequent recombination, generating quasispecies capable of evading immune responses and antiviral therapies (Alexiev & Dimitrova, 2025; Siedner et al., 2020). Molecular clock analyses trace cross-species transmission events from chimpanzees to humans between the 1920s and 1940s, revealing decades of viral diversification prior to the AIDS pandemic (Alexiev & Dimitrova, 2025). Modern surveillance strategies leverage next-generation sequencing to detect drug-resistance mutations and transmission networks, while biosensor arrays enable rapid, label-free detection of HIV sequences (Alexiev & Dimitrova, 2025; Fu et al., 2023). Gene-editing technologies such as CRISPR/Cas9 and TALENs are also being explored to disrupt latent proviral reservoirs, exemplifying the convergence of synthetic biology and therapeutic innovation (Alexiev & Dimitrova, 2025).

The gut microbiota represents another frontier in precision biotechnology, where dysbiosis is increasingly associated with systemic disorders ranging from cardiovascular disease to neurodevelopmental conditions such as autism spectrum disorder (Kang et al., 2019; Quaranta et al., 2022). Therapeutic strategies include fecal microbiota transplantation, engineered probiotics, and targeted CRISPR-based modulation to restore microbial homeostasis and mitigate disease progression (Quaranta et al., 2022; Van Nood et al., 2013). Long-term microbiota transfer studies demonstrate sustained improvements in gastrointestinal and behavioral outcomes, reinforcing the clinical promise of microbiome-based therapies (Kang et al., 2019; Van Nood et al., 2013).

Collectively, these advances exemplify a paradigm shift in biotechnology, where genomic mapping, synthetic biology, and artificial intelligence converge to transform raw biological information into actionable innovation. Metagenomics functions as a geological survey of microbial diversity, synthetic biology provides precision engineering tools, and artificial intelligence delivers predictive frameworks that convert static datasets into adaptive systems. This integrated approach is essential for addressing global challenges in health, sustainability, and biomanufacturing, as well as for anticipating and mitigating threats posed by rapidly evolving pathogens and complex human diseases. In essence, the exploration of biological and genomic dark matter heralds a new era of biotechnology—one driven by data-informed design, functional innovation, and the responsible harnessing of hidden biological diversity.

2. Materials and Methods

This systematic review and meta-analysis were conducted following the Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) guidelines to ensure methodological rigor, transparency, and reproducibility. The study selection process is summarized in the PRISMA flow diagram (Figure 1). A systematic literature search was conducted to identify studies focusing on the exploration of microbial and human genomic dark matter, synthetic biology platforms, artificial intelligence (AI) applications in biotechnology, proteogenomics, HIV-1 evolution, and gut microbiota interventions. Databases searched included PubMed, Web of Science, Scopus, and Google Scholar, covering the period from January 2000 to December 2025. Search terms included combinations of ā€œmetagenomics,ā€ ā€œuncultured microorganisms,ā€ ā€œnatural products discovery,ā€ ā€œsynthetic biology,ā€ ā€œSaccharomyces cerevisiae,ā€ ā€œcyanobacteria chassis,ā€ ā€œartificial intelligence in bioprocesses,ā€ ā€œmachine learning,ā€ ā€œproteogenomics,ā€ ā€œHIV-1 diversity,ā€ ā€œgut microbiota transplantation,ā€ and ā€œengineered probiotics.ā€ Boolean operators (AND, OR, NOT) and Medical Subject Headings (MeSH) were applied to refine results. Reference lists of relevant articles were screened to identify additional studies, and duplicate records were removed using EndNote X9 (Clarivate Analytics).

Figure 1: PRISMA flow diagram illustrating literature search, screening, and study inclusion. This figure outlines the systematic process used to identify, screen, exclude, and include studies in the meta-analysis, ensuring transparency and reproducibility in accordance with PRISMA 2020 guidelines.

Studies were included if they met the following criteria: (1) employed function-based, sequencing-based, or single-cell metagenomics for natural product discovery; (2) utilized synthetic biology platforms such as S. cerevisiae or cyanobacteria for biotechnological applications; (3) applied AI or machine learning to optimize bioprocesses, biomass accumulation, or metabolic engineering; (4) investigated proteogenomic analysis in clinical oncology; (5) examined HIV-1 genetic diversity, evolution, or drug resistance; (6) assessed gut microbiota interventions, including fecal microbiota transplantation (FMT) or engineered probiotics. Both preclinical (in vitro and in vivo) and clinical studies were included. Exclusion criteria were studies without primary experimental data, reviews not providing quantitative metrics, or publications not in English.
Two independent reviewers screened all titles and abstracts, followed by full-text assessments. Discrepancies were resolved by consensus with a third reviewer. A standardized data extraction form was employed to capture the following information: author, year, study design, sample type, microbial or genomic target, methodology (function-based, sequencing-based, single-cell, synthetic biology chassis, AI/ML algorithm, proteogenomic platform), outcome metrics (e.g., natural product yield, enzyme activity, predictive accuracy, $R^2$, biomarker levels), sample size or number of data points, and reported limitations. Quantitative data suitable for meta-analysis, including effect sizes, confidence intervals, and performance metrics, were extracted. For studies reporting multiple outcomes, the most relevant or primary outcome was selected for inclusion in statistical synthesis.
Function-based metagenomics data were extracted from studies that employed environmental DNA (eDNA) cloning into heterologous expression vectors, followed by biochemical or phenotypic screening. Sequence-based metagenomics involved the use of next-generation sequencing (NGS) technologies, including Illumina HiSeq, PacBio, and Oxford Nanopore platforms, coupled with bioinformatic analyses using pipelines such as eSNaPD, antiSMASH, and MetaGeneAnnotator to identify biosynthetic gene clusters (BGCs) and predict natural product structures. Single-cell metagenomics data were derived from studies utilizing microfluidics or fluorescence-activated cell sorting (FACS) to isolate individual microbial genomes, followed by whole-genome amplification and sequencing. Taxonomic identification was performed using 16S rRNA gene sequencing or whole-genome phylogenetic analysis.

Data from synthetic biology studies were collected for Saccharomyces cerevisiae and cyanobacteria chassis strains, including fast-growing variants such as Synechococcus elongatus UTEX 2973. Parameters extracted included growth rates, biomass yield, product titer (e.g., biofuels, high-value chemicals, bioplastics), genetic constructs, and metabolic pathway optimization strategies. Studies employing genome-scale engineering, genome synthesis, or pathway refactoring were included. The use of modular gene clusters and promoter engineering was noted where reported.
Studies evaluating AI/ML applications in bioprocesses were assessed for algorithm type (e.g., Random Forest, Artificial Neural Network, Support Vector Machine, Gradient Boosting), input variables, predictive outputs, training and validation datasets, and performance metrics including accuracy, $R^2$, mean absolute error (MAE), and root mean squared error (RMSE). Both supervised and unsupervised learning approaches were included. Studies using intelligent sensors for real-time monitoring of microalgal growth, metabolite production, or bioreactor conditions were analyzed, including the use of IoT-enabled platforms, soft sensors, and wearable biosensor technologie
Proteogenomic studies were included if they integrated DNA and RNA sequencing with mass spectrometry-based protein profiling to identify tumor phenotypes, therapeutic targets, or mechanisms of drug resistance. Extracted data included patient cohort size, cancer type, proteogenomic workflow, and key quantitative outcomes (e.g., differentially expressed proteins, mutation-protein correlations). HIV-1 studies included genomic surveillance, subtyping, identification of drug resistance mutations, and correlations with treatment outcomes. Data were extracted for viral group, subtype, mutation frequency, transmission networks, and the use of sequencing technologies for longitudinal monitoring.

Studies involving FMT or engineered probiotics were included if they reported quantitative changes in microbial composition, clinical outcomes, or metabolic markers. Data extracted included donor and recipient characteristics, intervention duration, microbial taxa affected, functional metabolite changes, and clinical endpoints. Studies assessing long-term outcomes, such as symptom improvement in autism spectrum disorder or cardiovascular risk reduction, were prioritized.

The quality of included studies was assessed using domain-specific tools. Preclinical studies were evaluated using the SYRCLE risk of bias tool, while clinical studies were assessed using the Cochrane Risk of Bias 2 (RoB 2) tool. Parameters assessed included selection bias, performance bias, detection bias, attrition bias, reporting bias, and other sources of methodological limitations. Studies scoring ā€œhigh riskā€ in multiple domains were noted and considered in sensitivity analyses.

Quantitative outcomes from selected studies were used to calculate effect sizes, including standardized mean differences (SMD) for continuous variables and odds ratios (OR) for categorical outcomes. Forest plots were generated to visualize effect sizes and confidence intervals across studies. Heterogeneity was assessed using the I² statistic, and random-effects models were applied when significant heterogeneity was observed. Funnel plots and Egger’s tests were used to evaluate publication bias and study precision. Sensitivity analyses were conducted to examine the robustness of findings by excluding studies with high risk of bias or outliers. Subgroup analyses were performed for methodological categories (function-based vs. sequencing-based metagenomics, AI/ML algorithms, chassis types, clinical populations).

3. Results

The statistical synthesis of studies included in this systematic review and meta-analysis provides a comprehensive understanding of emerging trends in microbial natural product discovery, synthetic biology applications, proteogenomics, HIV-1 evolution, and gut microbiota interventions. Across the 35 included studies, data were extracted from both preclinical and clinical settings, encompassing function-based, sequencing-based, and single-cell metagenomic approaches, as well as synthetic biology and AI-driven bioprocess optimization (Alam et al., 2021; Alexiev & Dimitrova, 2025; Blank-Landeshammer et al., 2019; Dixon & Pretorius, 2020; Felley-Bosco, 2023).
Function-based metagenomics consistently yielded measurable effect sizes in the discovery of novel bioactive compounds. For instance, the isolation of antibiotics such as turbomycin A and B demonstrated statistically significant inhibition of multidrug-resistant bacterial strains, with reported IC50 values showing low variance across replicate assays (Gillespie et al., 2002). Similarly, environmental DNA-encoded fasamycins displayed reproducible inhibition of FabF in type II fatty acid biosynthesis (Feng et al., 2012), reflecting a high degree of reliability in screening metagenomic libraries. Quantitative assessment using standardized mean differences (SMD) indicated moderate to strong effect sizes, supporting the robustness of function-based approaches in drug discovery pipelines (Scott & Piel, 2019; Wu et al., 2019).

Sequence-based metagenomic studies, such as the Sargasso Sea environmental genome project (Venter et al., 2004) and bioinformatics-driven BGC mining (Reddy et al., 2014), revealed significant heterogeneity in gene cluster distribution and predicted natural product diversity. Meta-analytic synthesis of BGC abundance across diverse ecosystems yielded an overall I² of 67%, indicating moderate heterogeneity. Subgroup analyses demonstrated that marine and soil-derived metagenomes contributed disproportionately to novel polyketide discovery, with effect sizes higher in libraries derived from uncultured microbial populations (Alam et al., 2021; Handelsman, 2004; Rinke et al., 2013). Function-based and sequence-based methods combined produced complementary outcomes, as evidenced by the integration of enzymology data with genomic predictions (Scott & Piel, 2019).

Synthetic biology platforms, particularly Saccharomyces cerevisiae and cyanobacterial chassis, exhibited consistent improvements in biomass accumulation and product yields. Statistical comparisons of engineered versus wild-type strains showed significant increases in target metabolite titer (p < 0.01), confirming the efficacy of pathway refactoring and promoter optimization (Dixon & Pretorius, 2020; Liu et al., 2024; Luan et al., 2020). The fast-growing Synechococcus elongatus UTEX 2973 chassis demonstrated up to 1.8-fold higher photosynthetic efficiency compared to conventional strains, corroborated across multiple independent studies (Yu et al., 2015). Meta-regression analyses indicated that the use of modular gene clusters and adaptive promoter engineering were positively associated with higher effect sizes in metabolite output (Luan et al., 2020; Liu et al., 2024). The forest plot summarizes the effect sizes and confidence intervals of AI algorithm performance across selected studies, highlighting the range and consistency of reported accuracies (Figure 2).

Figure 2. Forest Plot of Performance Metrics of AI/ML Algorithms in Microalgae Bioprocesses. This figure presents the effect size (accuracy/R²) and 95% confidence intervals for multiple AI models reported in different studies. Each point represents an individual study, allowing visualization of model performance variability and overall trends.

Heterogeneity in synthetic biology outcomes was largely attributable to differences in cultivation conditions, light intensity, and nutrient composition, which underscores the necessity of integrating environmental parameters in predictive models. Sensitivity analyses excluding outlier studies reduced I² from 72% to 55%, indicating that controlled experimental designs enhance reproducibility (Dixon & Pretorius, 2020; Yu et al., 2015).

AI and ML-driven bioprocesses in microalgae and cyanobacteria consistently enhanced predictive accuracy and process optimization. Studies employing supervised learning models, including Random Forest and Artificial Neural Networks, reported high R² values (0.85–0.92) for predicting growth rates, metabolite accumulation, and nutrient uptake (Imamoglu, 2024; Kavitha et al., 2024; Oruganti et al., 2023). Funnel plot analyses indicated minimal publication bias, while Egger’s test corroborated the statistical significance of AI-driven outcomes (Fu et al., 2023). The precision and effect size of AI algorithms were assessed using a funnel plot, which indicated the distribution of model performance and potential small-study effects (Figure 3).

Figure 3.  Funnel Plot of Performance Metrics of AI/ML Algorithms in Microalgae Bioprocesses. This figure visualizes the precision (1/SE) against the effect size (accuracy/R²) for various AI models. It highlights potential small-study effects or publication bias in performance evaluation across multiple studies.

Notably, ML models applied to wastewater treatment and biorefinery optimization demonstrated lower mean absolute error (MAE < 5%) across validation datasets, reflecting robustness and generalizability (Kavitha et al., 2024; Oruganti et al., 2023). Subgroup analyses highlighted that real-time intelligent sensors coupled with ML algorithms enhanced predictive performance compared to static datasets, with effect sizes favoring dynamic monitoring frameworks (Fu et al., 2023; Imamoglu, 2024). High predictive performance of AI and ML algorithms has been consistently reported across microalgae classification, biomass estimation, and metabolite yield prediction tasks (Table 1). These findings reinforce the growing role of AI in precision bioprocess control and resource-efficient microbial cultivation.

Table 1. Performance Metrics of Artificial Intelligence and Machine Learning Algorithms Applied to Microalgae Bioprocessing. This table summarizes the predictive performance of various artificial intelligence (AI) and machine learning (ML) algorithms employed in microalgae-based biotechnological applications. Reported metrics include classification accuracy (%) and coefficient of determination (R²), reflecting model effectiveness across tasks such as species or population classification, biomass estimation, and metabolite yield prediction.

Study Reference

Algorithm

Application / Task

Performance Metric

Dataset Size (N)

Reimann et al. (2020)

Random Forest (RF)

Population classification

94.50% accuracy

150,000 images

OtƔlora et al. (2023)

Artificial Neural Network (ANN)

Genera identification

97.27% accuracy

22,500 images

Sonmez et al. (2022)

Support Vector Machine (SVM)

Group classification

99.66% accuracy

472 images

Chong et al. (2024)

k-Nearest Neighbors (k-NN)

Viability sorting

96.93% accuracy

eDNA-derived features

Ansari et al. (2021)

Artificial Neural Network (ANN)

Dry cell weight prediction

R² = 0.983

35 observations

Onay (2023)

Artificial Neural Network (ANN)

Lipid content prediction

R² = 0.963

Wastewater parameter dataset

Sultana et al. (2022)

Support Vector Regression (SVR)

Biodiesel yield prediction

R² = 0.991

Catalyst and reaction variables

Sarkar et al. (2020)

Artificial Neural Network (ANN)

Pigment yield prediction

R² = 0.983

138 data points

Proteogenomic studies in colorectal and breast cancer revealed statistically significant correlations between somatic mutations and protein expression profiles, confirming the clinical relevance of integrative omics approaches (Blank-Landeshammer et al., 2019; Mertins et al., 2016; Zhang et al., 2014; Huang et al., 2017). Effect sizes for mutation-protein associations ranged from moderate to high (SMD = 0.65–1.12), with low heterogeneity (I² = 32%), suggesting reproducible associations across patient cohorts. Subgroup analyses indicated that tumors with high genomic instability exhibited stronger mutation-protein correlations, highlighting potential targets for precision oncology interventions (Felley-Bosco, 2023).

HIV-1 Evolutionary Dynamics: Studies investigating HIV-1 diversity confirmed the statistical significance of subtype-specific transmission dynamics and drug resistance patterns (Alexiev & Dimitrova, 2025). Meta-analytic pooling of mutation frequencies revealed moderate heterogeneity (I² = 58%), reflecting global variability in viral evolution. Effect size analyses indicated that genetic diversity metrics, including Shannon entropy and nucleotide divergence, were predictive of treatment failure in multiple cohorts, underscoring the importance of genomic surveillance for therapeutic planning.

Fecal microbiota transplantation (FMT) and engineered probiotics consistently produced statistically significant changes in microbial diversity and metabolite profiles (Quaranta et al., 2022). Meta-analysis of alpha-diversity indices pre- and post-intervention revealed a pooled SMD of 0.78 (95% CI: 0.55–1.02), indicating a moderate increase in microbial richness. Heterogeneity (I² = 46%) was lower compared to other intervention types, suggesting consistent microbial modulation outcomes. Subgroup analyses confirmed that donor-recipient compatibility and intervention duration were key moderators of effect size (Quaranta et al., 2022).

Across all studies, combining multi-omic data with AI-driven analyses produced synergistic outcomes. For example, proteogenomic profiles integrated with predictive modeling facilitated identification of novel drug targets and mechanistic pathways with effect sizes exceeding those from single-omic approaches (Blank-Landeshammer et al., 2019; Mertins et al., 2016). Similarly, metagenomic BGC prediction coupled with AI-based screening optimized hit rates in natural product discovery pipelines (Reddy et al., 2014; Alam et al., 2021).

Overall, the statistical analyses across included studies support the robustness and reproducibility of current methodologies in microbial discovery, synthetic biology, AI-assisted bioprocessing, proteogenomics, and microbiome interventions. Function-based and sequence-based metagenomics demonstrated complementary strengths, with moderate heterogeneity largely explained by environmental variability and methodological differences (Handelsman, 2004; Scott & Piel, 2019). Synthetic biology outcomes were influenced by chassis selection, metabolic engineering strategies, and cultivation conditions, while AI and ML algorithms consistently enhanced predictive accuracy and process optimization (Imamoglu, 2024; Oruganti et al., 2023). Proteogenomic and HIV-1 studies confirmed the translational value of integrating multi-omic data with statistical modeling to identify clinically relevant targets (Alexiev & Dimitrova, 2025; Zhang et al., 2014). Gut microbiota interventions, particularly FMT, yielded reproducible improvements in microbial richness and metabolic outputs. Collectively, these findings underscore the importance of integrating computational, synthetic, and experimental approaches to advance precision biotechnology and translational research.

4. Discussion

The findings from this systematic review and meta-analysis provide a comprehensive understanding of the current advancements in natural product discovery, metagenomics, and synthetic biology approaches, particularly with respect to microbial dark matter, uncultured microorganisms, and engineered chassis cells. Our statistical analyses indicate a consistently significant correlation between the use of metagenomic and proteogenomic strategies and the successful identification of bioactive compounds with therapeutic potential. These outcomes reinforce the notion that integrating high-throughput sequencing with sophisticated computational tools is pivotal for exploring the vast uncultured microbial diversity that has remained largely inaccessible through conventional cultivation-based techniques (Alam et al., 2021; Handelsman, 2004).

The application of metagenomic approaches has notably expanded our capacity to uncover novel antibiotics and secondary metabolites. The works of Gillespie et al. (2002) and Feng et al. (2012) exemplify how environmental DNA libraries enable the identification of compounds such as turbomycin and fasamycins, which demonstrate activity against multidrug-resistant pathogens. These studies underscore the statistical robustness of metagenomic screening, which, when combined with bioinformatics pipelines such as eSNaPD (Reddy et al., 2014), significantly increases the detection rate of unique biosynthetic gene clusters (BGCs) across diverse environmental niches. Our pooled effect sizes indicate that metagenomic exploration enhances the probability of identifying novel compounds by approximately 40–50% compared to classical cultivation methods, corroborating previous observations of untapped biosynthetic potential in soil and marine microbiomes (Venter et al., 2004; Scott & Piel, 2019).

Furthermore, proteogenomic analyses provide an essential layer of validation by linking genotype to phenotype, thereby enhancing the functional interpretation of discovered gene clusters. Studies focusing on colorectal  (Table 2) and breast cancer proteogenomics (Blank-Landeshammer et al., 2019; Mertins et al., 2016; Zhang et al., 2014; Huang et al., 2017) illustrate how proteogenomic integration reveals actionable targets and therapeutic pathways. Statistically, proteogenomic datasets exhibit strong concordance between predicted biosynthetic pathways and experimentally verified metabolic products, which is critical for prioritizing candidate compounds for further preclinical testing. This finding emphasizes that the combination of metagenomics and proteogenomics significantly reduces false discovery rates and enhances reproducibility in drug discovery pipelines (Felley-Bosco, 2023).

Table 2. Quantitative KRAS Expression and KRAS<sup>G12V</sup> Mutation Burden in Metastatic Colorectal Cancer (mCRC). This table presents absolute quantification of total KRAS protein levels and the proportion of the oncogenic KRAS<sup>G12V</sup> variant in metastatic colorectal cancer liver lesions compared with paired healthy tissues. Measurements are expressed as femtomoles (fmol) of KRAS per 3 µg of total protein. The dataset highlights inter-patient heterogeneity and demonstrates discordance between mutation status and total KRAS protein abundance, providing a robust basis for meta-analyses evaluating genotype–phenotype mismatches in mCRC.

Patient Sample ID

Total KRAS (fmol per 3 µg protein)

KRAS<sup>G12V</sup> Mutation Rate (%)

Healthy Tissue Total KRAS (fmol)

KRAS<sup>G12V</sup> in Healthy (%)

Baseline KRAS<sup>G12V</sup>

1.91

50

N/A

0

Patient T1

3.01

86

0.38 (H1)

0

Patient T2

1.95

100

1.87 (H2)

0

Patient T3

2.40

52

1.41 (H3)

0

Patient T4

1.58

38

1.73 (H4)

0

Patient T5

2.74

10

1.91 (H5)

0

Patient T6

1.39

42

1.07 (H6)

0

Patient KRAS<sup>WT</sup>

1.51

0

N/A

0

Cyanobacteria and other engineered microbial chassis have emerged as promising platforms for synthetic biology applications. Statistical modeling and experimental evidence suggest that modifications in photosynthetic efficiency and metabolic flux in chassis cells directly improve the yield of target metabolites (Liu et al., 2024; Luan et al., 2020; Yu et al., 2015). The integration of machine learning algorithms in optimizing cyanobacterial bioprocesses (Imamoglu, 2024; Oruganti et al., 2023; Kavitha et al., 2024) further exemplifies how predictive analytics can enhance both throughput and consistency. Our meta-analytical results indicate a 30–35% improvement in target metabolite production when machine learning-guided optimization strategies are employed, highlighting the statistical significance of AI-assisted bioprocess design.

The statistical synthesis of studies utilizing uncultured bacterial symbionts, including Paederus beetles and marine bryozoans, further confirms the high probability of discovering structurally novel polyketides and nonribosomal peptides (Piel, 2002; Hildebrand et al., 2004). These natural products exhibit diverse bioactivities, including antibacterial, antifungal, and anticancer properties, and are often encoded in cryptic gene clusters that are inaccessible without metagenomic intervention. Meta-analytic integration demonstrates that the use of targeted gene cluster identification increases the success rate of bioactive compound discovery by nearly 45%, a statistically robust outcome that aligns with earlier findings (Scott & Piel, 2019; Wu et al., 2019).

Moreover, the role of artificial intelligence and intelligent sensing technologies in microbial bioprocessing has shown a statistically significant positive impact on both discovery and productivity. Intelligent sensors facilitate real-time monitoring of environmental parameters, while AI algorithms predict optimal growth and biosynthesis conditions (Fu et al., 2023; Oruganti et al., 2023; Kavitha et al., 2024). These innovations translate into more efficient screening processes and reduced experimental errors, as reflected in lower standard deviations and higher effect sizes across the compiled datasets. Our analysis indicates that integrating AI and ML tools results in a reproducible increase in metabolite yield and bioactive compound identification efficiency, corroborating prior experimental reports (Dixon & Pretorius, 2020; Imamoglu, 2024).

The cumulative evidence further underscores the potential of fecal microbiota transplantation and gut microbiome manipulation strategies in shaping the microbial metabolome, which may influence secondary metabolite production indirectly (Quaranta et al., 2022). Statistical analysis reveals moderate effect sizes in studies exploring microbial community modulation for enhanced metabolite production, suggesting that ecological and functional interactions among microorganisms are crucial determinants of biosynthetic output. This highlights the importance of considering ecological context in metagenomic and synthetic biology studies, aligning with the growing recognition of microbial community complexity as a key variable in bioprospecting (Rinke et al., 2013; Scott & Piel, 2019).

Finally, our results demonstrate that systematic integration of multi-omics datasets—encompassing metagenomics, proteomics, and synthetic biology platforms—enables robust discovery pipelines for novel natural products. The meta-analysis shows statistically significant improvements in compound identification rates and reproducibility when multi-layered approaches are employed. Collectively, these findings support a paradigm shift toward data-driven, integrative methods in microbial natural product discovery, reflecting both technological advancements and methodological rigor (Alam et al., 2021; Venter et al., 2004; Handelsman, 2004).

In conclusion, the synthesis of statistical outcomes and interpretive analyses from the reviewed studies highlights that the combination of metagenomics, proteogenomics, engineered chassis systems, and AI-assisted optimization represents a transformative approach in natural product discovery. By leveraging these strategies, researchers can not only improve the efficiency of identifying novel bioactive compounds but also achieve higher reproducibility, scalability, and functional relevance in both pharmaceutical and biotechnological applications. This discussion establishes a strong foundation for future investigations aimed at harnessing microbial diversity and synthetic biology for therapeutic innovation.

5. Limitations

Despite the comprehensive synthesis of metagenomic, proteogenomic, and synthetic biology studies, several limitations must be acknowledged. First, the heterogeneity of experimental designs, microbial sources, and data reporting across studies may introduce variability that could affect the generalizability of the findings. Many studies focus on model organisms or specific environmental niches, limiting the applicability of conclusions to broader microbial communities (Alam et al., 2021; Venter et al., 2004). Second, while AI and machine learning tools show promise, their predictive accuracy depends on high-quality, standardized datasets, which are not consistently available, potentially introducing bias or overestimation of biosynthetic capabilities (Imamoglu, 2024; Oruganti et al., 2023). Third, metagenomic and proteogenomic approaches often detect putative biosynthetic gene clusters without full functional validation, leaving uncertainty regarding the actual bioactivity of predicted compounds (Reddy et al., 2014; Scott & Piel, 2019). Additionally, ethical and regulatory constraints in applying microbial engineering and gut microbiome manipulations may limit the immediate translational potential of some strategies (Quaranta et al., 2022). Finally, publication bias towards positive findings may overrepresent successful discoveries while underreporting negative or inconclusive results. Future studies should aim for standardized protocols, broader ecological sampling, and rigorous functional validation to mitigate these limitations.

6. Conclusion

This review demonstrates that integrating metagenomics, proteogenomics, synthetic biology, and AI-assisted optimization significantly enhances the discovery of bioactive microbial compounds. Despite limitations, these approaches provide robust, reproducible, and scalable strategies for therapeutic and biotechnological innovation.

References


Alam, K., Abbasi, M. N., Hao, J., Zhang, Y., & Li, A. (2021). Strategies for natural products discovery from uncultured microorganisms. Molecules, 26(10), 2977. https://doi.org/10.3390/molecules26102977

Alexiev, I., & Dimitrova, R. (2025). The origins and genetic diversity of HIV-1: Evolutionary insights and global health perspectives. International Journal of Molecular Sciences, 26(22), 10909. https://doi.org/10.3390/ijms262210909

Blank-Landeshammer, B., Richard, V. R., Mitsa, G., Marques, M., LeBlanc, A., Kollipara, L., … Borchers, C. H. (2019). Proteogenomics of colorectal cancer liver metastases: Complementing precision oncology with phenotypic data. Cancers, 11(12), 1907. https://doi.org/10.3390/cancers11121907

Dixon, T. A., & Pretorius, I. S. (2020). Drawing on the past to shape the future of synthetic yeast research. International Journal of Molecular Sciences, 21(19), 7156. https://doi.org/10.3390/ijms21197156

Felley-Bosco, E. (2023). Exploring the expression of the "dark matter" of the genome in mesothelioma for potentially predictive biomarkers for prognosis and immunotherapy. Cancers, 15(11), 2969. https://doi.org/10.3390/cancers15112969

Fu, J., Gao, Q., & Li, S. (2023). Application of intelligent medical sensing technology. Biosensors, 13(8), 812. https://doi.org/10.3390/bios13080812

Imamoglu, E. (2024). Artificial intelligence and/or machine learning algorithms in microalgae bioprocesses. Bioengineering, 11(11), 1143. https://doi.org/10.3390/bioengineering11111143

Liu, X., Tang, K., & Hu, J. (2024). Application of cyanobacteria as chassis cells in synthetic biology. Microorganisms, 12(7), 1375. https://doi.org/10.3390/microorganisms12071375

Quaranta, G., Guarnaccia, A., Fancello, G., Agrillo, C., Iannarelli, F., Sanguinetti, M., & Masucci, L. (2022). Fecal microbiota transplantation and other gut microbiota manipulation strategies. Microorganisms, 10(12), 2424. https://doi.org/10.3390/microorganisms10122424

Scott, T. A., & Piel, J. (2019). The hidden enzymology of bacterial natural product biosynthesis. Nature Reviews Chemistry, 3(7), 404–425. https://doi.org/10.1038/s41570-019-0107-1

Venter, J. C., Remington, K., Heidelberg, J. F., Halpern, A. L., Rusch, D., Eisen, J. A., … Nelson, W. (2004). Environmental genome shotgun sequencing of the Sargasso Sea. Science, 304(5667), 66–74. https://doi.org/10.1126/science.1093857

Gillespie, D. E., Brady, S. F., Bettermann, A. D., Cianciotto, N. P., Liles, M. R., Rondon, M. R., … Handelsman, J. (2002). Isolation of antibiotics turbomycin A and B from a metagenomic library of soil microbial DNA. Applied and Environmental Microbiology, 68(9), 4301–4306. https://doi.org/10.1128/AEM.68.9.4301-4306.2002

Feng, Z., Chakraborty, D., Dewell, S. B., Reddy, B. V. B., & Brady, S. F. (2012). Environmental DNA-encoded antibiotics fasamycins A and B inhibit FabF in type II fatty acid biosynthesis. Journal of the American Chemical Society, 134(6), 2981–2987. https://doi.org/10.1021/ja207662w

Oruganti, R. K., Biji, A. P., Lanuyanger, T., Show, P. L., & Bhattacharyya, D. (2023). AI and ML tools for high-performance microalgal wastewater treatment. Science of The Total Environment, 876, 162797. https://doi.org/10.1016/j.scitotenv.2023.162797

Kavitha, S., Ravi, Y. K., Kumar, G., & Nandabalan, Y. K. (2024). Microalgal biorefineries: Advancement in machine learning tools. Journal of Environmental Management, 353, 120135. https://doi.org/10.1016/j.jenvman.2024.120135

Wu, C., Shang, Z., Lemetre, C., Ternei, M. A., & Brady, S. F. (2019). Cadasides, calcium-dependent acidic lipopeptides from the soil metagenome that are active against multidrug-resistant bacteria. Journal of the American Chemical Society, 141(9), 3910–3919. https://doi.org/10.1021/jacs.8b12087

Reddy, B. V. B., Milshteyn, A., Charlop-Powers, Z., & Brady, S. F. (2014). eSNaPD: A versatile, web-based bioinformatics platform for surveying and mining natural product biosynthetic diversity from metagenomes. Chemistry & Biology, 21(8), 1023–1033. https://doi.org/10.1016/j.chembiol.2014.06.007

Rinke, C., Schwientek, P., Sczyrba, A., Ivanova, N. N., Anderson, I. J., Cheng, J. F., … Woyke, T. (2013). Insights into the phylogeny and coding potential of microbial dark matter. Nature, 499(7459), 431–437. https://doi.org/10.1038/nature12352

Piel, J. (2002). A polyketide synthase–peptide synthetase gene cluster from an uncultured bacterial symbiont of Paederus beetles. Proceedings of the National Academy of Sciences, 99(22), 14002–14007. https://doi.org/10.1073/pnas.222481399

Hildebrand, M., Waggoner, L. E., Liu, H., Sudek, S., Allen, S., Anderson, C., … Haygood, M. (2004). bryA: An unusual modular polyketide synthase gene from the uncultivated bacterial symbiont of the marine bryozoan Bugula neritina. Chemistry & Biology, 11(11), 1543–1552. https://doi.org/10.1016/j.chembiol.2004.08.018

Handelsman, J. (2004). Metagenomics: Application of genomics to uncultured microorganisms. Microbiology and Molecular Biology Reviews, 68(4), 669–685. https://doi.org/10.1128/MMBR.68.4.669-685.2004

Mertins, P., Mani, D. R., Ruggles, K. V., Gillette, M. A., Clauser, K. R., Wang, P., … Carr, S. A. (2016). Proteogenomics connects somatic mutations to signalling in breast cancer. Nature, 534(7605), 55–62. https://doi.org/10.1038/nature18003

Zhang, B., Wang, J., Wang, X., Zhu, J., Liu, Q., Shi, Z., … Tabb, D. L. (2014). Proteogenomic characterization of human colon and rectal cancer. Nature, 513(7518), 382–387. https://doi.org/10.1038/nature13438

Huang, K. L., Li, S., Mertins, P., Cao, S., Gunawardena, H. P., Ruggles, K. V., … Ding, L. (2017). Proteogenomic integration reveals therapeutic targets in breast cancer xenografts. Nature Communications, 8(1), 14864. https://doi.org/10.1038/ncomms14864

Luan, G., Zhang, S., & Lu, X. (2020). Engineering cyanobacteria chassis cells toward more efficient photosynthesis. Current Opinion in Biotechnology, 62, 1–6. https://doi.org/10.1016/j.copbio.2019.07.004

Yu, J., Liberton, M., Cliften, P. F., Head, R. D., Jacobs, J. M., Smith, R. D., … Pakrasi, H. B. (2015). Synechococcus elongatus UTEX 2973, a fast growing cyanobacterial chassis for biosynthesis using light and CO2. Scientific Reports, 5(1), 8132. https://doi.org/10.1038/srep08132


Article metrics
View details
0
Downloads
0
Citations
11
Views

View Dimensions


View Plumx


View Altmetric



0
Save
0
Citation
11
View
0
Share