Invited Review

Engineering Synthetic Microbial Communities for Enhanced Probiotic Functionality and Gut Health Improvement  

Jingqiang Wang
Cuixi Academy of Biotechnology, Zhuji, 311800, Zhejiang, China
Author    Correspondence author
International Journal of Clinical Case Reports, 2024, Vol. 14, No. 2   
Received: 25 Mar., 2024    Accepted: 27 Apr., 2024    Published: 08 May, 2024
© 2024 BioPublisher Publishing Platform
This is an open access article published under the terms of the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited.
Abstract

The objective of this study is to examine advancements in engineering synthetic microbial communities aimed at enhancing probiotic functionality and improving gut health. The review focuses on the integration of synthetic biology tools, dietary influences, and the therapeutic applications of these engineered probiotics. Significant progress has been made in developing engineered probiotics capable of delivering therapeutics and responding to environmental cues within the gut. Key findings include the enhanced therapeutic potential of engineered probiotics for treating diseases such as inflammatory bowel disease and metabolic disorders, and the challenges posed by the complexity and adaptability of synthetic probiotics compared to native gut microbiota. Additionally, the integration of artificial intelligence and synthetic biology has significantly advanced personalized probiotic therapies. Despite these advancements, challenges remain in replicating the complexity of native microbiota and ensuring the long-term stability and efficacy of engineered probiotics. Future research should focus on interdisciplinary collaboration, integrating advanced technologies, and establishing robust regulatory frameworks to maximize the therapeutic potential of synthetic probiotics.

Keywords
Synthetic microbial communities; Engineered probiotics; Gut health; Synthetic biology; Personalized medicine

Gut health is increasingly recognized as a crucial aspect of overall well-being, influencing not only digestion but also immune function, mental health, and disease prevention. The human gut microbiome is a complex and dynamic ecosystem that plays a crucial role in maintaining overall health. It is involved in various physiological processes, including metabolism, immune modulation, and the development of the nervous system (Bober et al., 2018; Dou and Bennett, 2018). Dysbiosis, or an imbalance in the gut microbiota, has been linked to various health issues such as inflammatory bowel disease, obesity, diabetes, and even neurological disorders. Probiotics, comprising beneficial bacteria and yeasts, which are live microorganisms that confer health benefits to the host when administered in adequate amounts, have been extensively studied for their potential to enhance gut health. They can help in maintaining a balanced gut microbiota, improving digestion, and boosting the immune system (Crovesy et al., 2021). The modulation of gut microbiota through probiotics has shown promising results in treating conditions such as obesity, gastrointestinal disorders, and inflammation (Crovesy et al., 2021).

 

Synthetic microbial communities (SynComs) represent an innovative approach in the field of probiotics. Unlike traditional probiotics, which often consist of single or a few strains of beneficial bacteria, SynComs are designed to mimic the complexity and functionality of natural microbial communities (Venturelli et al., 2018; Berkhout et al., 2022). These engineered communities can be tailored to perform specific functions, such as producing therapeutic compounds, enhancing nutrient absorption, or modulating immune responses (Landry and Tabor, 2017; Dou and Bennett, 2018), and offer enhanced stability and efficacy. By harnessing the synergistic interactions between different microbial species, SynComs can provide more robust and targeted therapeutic benefits. The engineering of SynComs involves selecting microbial species based on their functional traits, compatibility, and ability to survive and thrive in the gut environment (Dou and Bennett, 2018). Advances in synthetic biology have enabled the precise engineering of these communities, allowing for the incorporation of genetic circuits, biosensors, and other sophisticated tools to enhance their efficacy and safety (Landry and Tabor, 2017; Bober et al., 2018; Dou and Bennett, 2018). The integration of artificial intelligence (AI) with synthetic biology further enhances the potential of SynComs by enabling the analysis and optimization of complex microbial interactions (Kumar et al., 2022).

 

The primary objective of this study is to evaluate the current advancements and potential of engineering synthetic microbial communities for enhanced probiotic functionality and gut health improvement. This study aims to synthesize existing research on SynComs, identify the key factors influencing their design and efficacy, and highlight the potential benefits and challenges associated with their use. By providing a comprehensive overview of the state-of-the-art in SynCom engineering, this study seeks to offer valuable insights for researchers, clinicians, and industry stakeholders. Ultimately, the significance of this study lies in its potential to guide future research and development efforts towards creating more effective probiotic therapies that can address the complexities of gut health and contribute to overall well-being.

 

1 Gut Microbiota and Probiotic Functionality

1.1 Composition and functions of the gut microbiota

The human gut microbiota is a diverse and dynamic community of microorganisms, including bacteria, archaea, viruses, and fungi. These microorganisms are crucial for various physiological functions such as digestion, nutrient absorption, and immune modulation. The gut microbiota composition varies between individuals and can be influenced by diet, environment, and genetics (Kho and Lal, 2018). which collectively shape its structure and function (Vrancken et al., 2019). Understanding the intricate interactions within this microbial community is essential for developing effective therapeutic strategies aimed at modulating gut health.

 

1.2 Mechanisms through which probiotics influence gut health

Probiotics, defined as live microorganisms that confer health benefits to the host when administered in adequate amounts, exert their effects through multiple mechanisms. They enhance the gut barrier function, modulate the immune response, and inhibit pathogenic bacteria by producing antimicrobial substances (Kaur and Ali, 2022). Probiotics also produce short-chain fatty acids (SCFAs) like butyrate, which provide energy to colonocytes and exhibit anti-inflammatory properties (Satapathy et al., 2019). Advances in synthetic biology have enabled the engineering of probiotics to enhance these functionalities further. For instance, engineered probiotics can be designed to produce and deliver therapeutic molecules directly within the gut, thereby improving their efficacy in treating various gastrointestinal disorders (Dou and Bennett, 2018; Kumar et al., 2022).

 

1.3 Limitations of current probiotic formulations

Despite the promising potential of probiotics, current formulations face several limitations. Traditional probiotics often lack the complexity and adaptability of the native gut microbiota, which can limit their effectiveness in restoring or maintaining gut health (Dou and Bennett, 2018). Many probiotics struggle to survive the acidic environment of the stomach and bile salts in the intestine (Zhou et al., 2020), the stability and viability of probiotic strains during storage and gastrointestinal transit remain significant challenges (Martins et al., 2023). Synthetic microbial communities (SynComs) offer a potential solution by combining multiple microbial species to create a more stable and functional consortium. However, ensuring the long-term stability and colonization of these synthetic communities in the gut environment is still a major hurdle (Berkhout et al., 2022; Martins et al., 2023). Further research is needed to address these challenges and optimize the design and application of engineered probiotics and SynComs for improved gut health outcomes.

 

2 Engineering Synthetic Microbial Communities (SynComs)

2.1 Definition and principles of SynComs

Synthetic microbial communities (SynComs) are deliberately constructed consortia of microorganisms designed to perform specific functions within a host environment. Unlike natural microbial communities, which are complex and often unpredictable, SynComs are composed of carefully selected microbial species that are engineered to achieve desired outcomes, such as enhancing gut health or providing therapeutic benefits (van Leeuwen et al., 2023; Martins et al. 2023). The goal is to create a stable, resilient, and functional microbiome that can outcompete pathogenic bacteria and provide health benefits to the host. The principles underlying SynComs involve understanding the ecological interactions and functional capabilities of individual microbes, and then assembling them in a way that maximizes their collective efficacy (Liu et al., 2019; Wang et al., 2023). This approach allows for a more controlled and predictable manipulation of the microbiome, which can be tailored to address specific health conditions or improve overall gut functionality (Khan et al., 2022; van Leeuwen et al., 2023).

 

2.2 Techniques for engineering SynComs

The engineering of SynComs leverages various techniques from synthetic biology and genetic modification. Synthetic biology provides tools such as CRISPR-Cas systems for precise gene editing, enabling the modification of microbial genomes to enhance desired traits or introduce new functionalities (Landry and Tabor, 2017; Kumar et al., 2022), such as metabolite production or pathogen resistance (Kumar et al., 2022). There are also Synthetic Biology Tools such as synthetic gene circuits and biosensors enable the creation of microbes that can sense and respond to environmental signals within the gut, thereby performing diagnostic or therapeutic functions (Naydich et al., 2019). By modifying the metabolic pathways of microbes to enhance the production of health-promoting compounds like short-chain fatty acids (SCFAs) or to degrade harmful substances in the gut (Jansma et al., 2023). Creating SynComs using a modular approach allows for the assembly of different microbial strains, each contributing a specific function, thereby enhancing the overall functionality and stability of the community (Huang et al., 2022). Additionally, advanced computational modeling and machine learning are employed to predict and optimize the interactions within SynComs, ensuring stability and effectiveness (Diener et al., 2020; Martins et al., 2023). Techniques such as microbial cultivation and reconstruction are also crucial, allowing researchers to reproducibly investigate and manipulate the interactions between different microbial species under controlled conditions (Bober et al., 2018; Liu et al., 2019). These methods collectively enable the design, assembly, and testing of SynComs with specific therapeutic or diagnostic purposes (Dou and Bennett, 2018; van Leeuwen et al., 2023).

 

2.3 Comparison of SynComs with natural microbial communities

SynComs differ from natural microbial communities in several key aspects. Natural communities are inherently complex and dynamic, with interactions that are often not fully understood. They are shaped by evolutionary processes and environmental factors, leading to a high degree of variability and unpredictability (Martins et al., 2023; Wang et al., 2023). In contrast, SynComs are designed and assembled with specific functions in mind, providing greater control over the microbial composition and their metabolic outputs compared to natural communities, which are shaped by complex and often unpredictable ecological interactions (Dou and Bennett, 2018). Engineered communities can be designed to be more stable and resilient to environmental changes and perturbations than natural communities, which can be more susceptible to disruptions and imbalances (Landry and Tabor, 2017). SynComs are specifically engineered to perform desired functions, such as producing therapeutics or enhancing gut health, which can be more effective than the broader, less targeted functions of natural microbial communities (Zhou et al., 2020). The assembly of SynComs involves a rational design process guided by synthetic biology principles, whereas natural communities evolve through ecological processes that are not necessarily optimized for human health benefits (Clark et al., 2020). However, one of the challenges with SynComs is ensuring their stability and functionality over time, as they may be subject to changes due to horizontal gene transfer and mutations (van Leeuwen et al., 2023; Martins et al., 2023). Despite these challenges, SynComs offer a promising alternative to natural communities, providing a more targeted and reliable means of modulating the microbiome for health benefits (Landry and Tabor, 2017; Bober et al., 2018).

 

By leveraging the principles of synthetic biology and advanced computational tools, SynComs represent a significant advancement in the field of microbiome research, offering new opportunities for enhancing probiotic functionality and improving gut health.

 

3 Mechanisms of Enhanced Probiotic Functionality

3.1 Engineering metabolic pathways for improved probiotic functions

Metabolic engineering involves modifying the genetic and biochemical pathways of probiotic microorganisms to enhance their functionality and health benefits, enabling them to produce beneficial metabolites and therapeutic compounds. For instance, synthetic biology tools have been employed to engineer probiotics to produce small molecule therapeutics within the gut, which can help in regulating metabolism and modulating immune responses (Dou and Bennett, 2018). Additionally, the use of gene editing tools like CRISPR-Cas systems allows for precise modifications in probiotic genomes, enhancing their ability to perform specific metabolic functions that are crucial for gut health (Kumar et al., 2022). Recent studies have focused on optimizing the production of beneficial metabolites and improving the overall metabolic efficiency of probiotics. Jain et al. (2021) work to study the engineering Lactobacilli and Bifidobacteria to increase lactic acid production. Enhanced lactic acid production improves gut health by lowering pH and inhibiting pathogenic bacteria. Techniques such as gene editing and pathway optimization have been employed to achieve this. Yadav et al. (2018) found that, increasing EPS production in probiotics like Lactobacillus reuteri enhances their ability to adhere to the gut mucosa and modulate the host's immune response. Genetic modifications have been used to introduce novel pathways for EPS synthesis, resulting in improved probiotic adherence and immunomodulatory effects. Huang et al. (2022) found that engineered probiotics could produce higher levels of short-chain fatty acids (SCFA) (Figure 1), such as butyrate can have significant health benefits, including anti-inflammatory effects and improved gut barrier function. This has been achieved through the introduction of genes involved in SCFA production pathways.

 

 

Figure 1 Omprehensive analysis of butyrate production in bacterial communities (Adapted from Clark et al., 2021)

Image caption: Utilizing data from 1-2 species and 24-25 species experiments. The scatter plot (Panel a) illustrates the variation in butyrate production across different community complexities, highlighting specific communities lacking certain butyrate producers (e.g., DP−, AC−). Panel b shows predicted butyrate levels for 3-5 member communities, using a model trained on simpler communities, represented by median predictions and 60% confidence intervals. Panel c correlates measured versus predicted butyrate concentrations, displaying a strong Pearson correlation, which underscores the model's effectiveness in predicting butyrate production across varying community structures. The methodology, combining experimental and predictive approaches, offers valuable insights into microbial interactions and their impact on metabolic output (Adapted from Clark et al., 2021)

 

3.2 Enhancing microbial interactions and synergy within SynComs

Synthetic microbial communities (SynComs) are designed to mimic natural microbial ecosystems, enhancing probiotic functionality through synergistic interactions. These communities consist of multiple microbial species that work together to provide greater health benefits than single-strain probiotics. SynComs are carefully constructed to include microbial species that can work together to produce desired outcomes, such as improved gut health and resistance to pathogens (van Leeuwen et al., 2023; Martins et al., 2023). SynComs can improve nutrient acquisition and inhibit pathogenic infections more effectively than single strains. Studies have shown that SynComs constructed with beneficial microbial strains can enhance nutrient acquisition, increase crop yield in agricultural settings, and demonstrate similar benefits in human gut health (Wang et al., 2021). Additionally, they produce antimicrobial compounds that enhance the host's immune response and protect against pathogens (Yin et al., 2022). By understanding the ecological theories and community assembly rules, researchers can design SynComs that are more stable and effective under various environmental conditions (Martins et al., 2023). This approach has been shown to be effective in both plant and human microbiomes, where well-structured SynComs can lead to better colonization and long-term stability (De Souza et al., 2020; Marín et al., 2021). Enhanced biofilm formation and improved colonization of the gut are some benefits observed with SynComs (Sun et al., 2023). The use of computational methods, including machine learning, further aids in identifying the best combinations of microbes for desired phenotypes, enhancing the overall functionality of the SynComs (De Souza et al., 2020).

 

3.3 Genetic modifications to improve resilience and colonization of probiotics

Genetic modifications are crucial for improving the resilience and colonization capabilities of probiotics. By introducing specific genetic signatures designed to improve the ability of probiotics to tolerate stress conditions such as acids, bile salts, and oxidative stress, probiotics can become more resistant to environmental stress and colonize the gut more effectively (van Leeuwen et al., 2023). For example, engineered probiotics with enhanced biocontainment mechanisms and logic-gating systems can better survive and function in the complex gut environment (Dou and Bennett, 2018). Genetic enhancements in stress tolerance mechanisms are crucial for improving probiotic resilience. For example, Saccharomyces boulardii has been genetically modified to increase its secretion of therapeutic proteins, enhancing its effectiveness in treating gastrointestinal disorders (Durmusoglu et al., 2022). Enhancing the ability of probiotics to form biofilms improves their protection from environmental stresses and adherence to the gut mucosa. Biofilm formation is a key factor in probiotic colonization and resilience (Bamunuarachchige et al., 2011). Additionally, genetic modifications can help probiotics to better adhere to the gut mucosa and resist the competitive pressures from native microbial communities (van Leeuwen et al., 2023). This is particularly important for ensuring the long-term efficacy of probiotic treatments, as stable colonization is essential for sustained health benefits (Li et al., 2021; van Leeuwen et al., 2023). Advanced gene editing tools like CRISPR-Cas9 have been used to introduce specific genetic traits that enhance probiotic performance. For instance, Lactobacillus and Bifidobacterium species have been engineered to express genes that confer resistance to bile salts and acidity, improving their survival and colonization in the gut (Yadav et al., 2020). Genetically modified probiotics can be designed to produce therapeutic compounds targeting specific pathogens or modulating the host's immune response. Engineered Escherichia coli strains, for example, have been developed to eliminate Pseudomonas aeruginosa by producing anti-biofilm enzymes and other antimicrobial compounds (Hwang et al., 2017).

 

The engineering of metabolic pathways, enhancement of microbial interactions within SynComs, and genetic modifications to improve resilience and colonization are key mechanisms that can significantly enhance the functionality of probiotics. These approaches leverage advanced tools and methodologies from synthetic biology and computational modeling to create more effective and stable probiotic treatments for gut health improvement.

 

4 Applications of Engineered SynComs in Gut Health

4.1 SynComs for treating gastrointestinal disorders

Engineered synthetic microbial communities (SynComs) have shown significant promise in treating gastrointestinal disorders such as Irritable Bowel Syndrome (IBS) and Inflammatory Bowel Disease (IBD). These disorders are often associated with dysbiosis, an imbalance in the gut microbiota. SynComs can be designed to restore this balance by introducing beneficial microbial species that can outcompete pathogenic bacteria and reduce inflammation. For instance, SynComs have been utilized to combat chronic inflammatory bowel diseases by harnessing their potential to modulate the gut-immune axis and reduce inflammation (van Leeuwen et al., 2023). Additionally, engineered probiotics have been developed to produce and deliver therapeutic molecules directly within the gut, offering a targeted approach to treating these disorders (Dou and Bennett, 2018; Huang et al., 2022).

 

4.2 Role of SynComs in enhancing nutrient absorption and metabolism

SynComs play a crucial role in enhancing nutrient absorption and metabolism in the gut. By introducing specific microbial species that possess the ability to break down complex carbohydrates, proteins, and fats, SynComs can improve the overall efficiency of nutrient absorption. This is particularly beneficial in conditions where nutrient malabsorption is a concern. Engineered probiotics have been shown to convert dietary nutrients into bioactive metabolites that can be readily absorbed by the host, thereby improving nutritional status and overall health (Hemarajata and Versalovic, 2013). Moreover, the use of synthetic biology tools allows for the design of SynComs that can produce essential vitamins and amino acids, further enhancing the metabolic capabilities of the gut microbiota (Bober et al., 2018).

 

4.3 Immunomodulatory effects of engineered probiotics

The immunomodulatory effects of engineered probiotics are one of the most promising applications of SynComs in gut health. These engineered communities can interact with the host's immune system to modulate immune responses, thereby reducing inflammation and enhancing immune tolerance. For example, SynComs have been designed to produce anti-inflammatory compounds that can mitigate gut inflammation and promote a healthy immune response (Hemarajata and Versalovic, 2013; van Leeuwen et al., 2023). Additionally, the introduction of specific microbial species that can stimulate the production of regulatory T cells has been shown to enhance immune tolerance and prevent autoimmune responses (Huang et al., 2022). These immunomodulatory effects are crucial for maintaining gut homeostasis and preventing chronic inflammatory conditions.

 

Engineered SynComs offer a multifaceted approach to improving gut health by treating gastrointestinal disorders, enhancing nutrient absorption and metabolism, and modulating immune responses. The continued development and optimization of these synthetic communities hold great potential for advancing probiotic therapies and improving overall gut health.

 

5 Performance Evaluation and Clinical Trials

5.1 Criteria and metrics for assessing SynCom performance in the gut

The evaluation of synthetic microbial communities (SynComs) in the gut involves multiple criteria and metrics to comprehensively assess their functionality and impact. Key criteria include:

 

1) Microbiota Composition and Diversity: This involves assessing the changes in the microbial community structure using sequencing technologies to identify shifts in bacterial, fungal, and viral populations (Baldi et al., 2021).

 

2) Gut Barrier Function: Evaluation of gut barrier integrity, typically through biomarkers such as zonulin levels, and histological analysis of gut epithelial cells (Vogt and Finlay, 2017).

 

3) Immune Modulation: Measurement of immune responses, including cytokine profiles and immune cell populations, to determine the immunomodulatory effects of SynComs.

 

4) Metabolic Outputs: Assessment of metabolic products such as short-chain fatty acids (SCFAs), which are critical for gut health and are influenced by microbial activity (Stefanaki et al., 2017).

 

5) Clinical Outcomes: Clinical endpoints such as symptom relief, disease remission rates, and overall patient well-being are crucial for evaluating the practical benefits of SynCom interventions (Schrodt et al., 2023).

 

5.2 Overview of preclinical and clinical trials involving engineered SynComs

Preclinical trials of engineered SynComs typically involve in vitro fermentation models and animal studies to simulate gut conditions and evaluate microbial behavior (Arcidiacono et al., 2023). These models help in understanding the persistence, functionality, and safety of SynComs before transitioning to human trials.

 

Animal models, particularly mice, are often used to study the effects of SynComs on gut microbiota composition, immune responses, and metabolic outputs. For example, trials have demonstrated that SynComs can modulate immune responses and enhance gut barrier function in rodent models.

 

Human trials are crucial for assessing the safety and efficacy of SynComs in target populations. Studies have explored the use of SynComs for conditions such as irritable bowel syndrome (IBS), inflammatory bowel disease (IBD), and metabolic disorders. These trials often include randomized controlled trials (RCTs) to compare SynCom interventions against placebos or standard treatments (Pirbaglou et al., 2016).

 

5.3 Case studies and outcomes of SynCom applications in human health

The application of synthetic microbial communities (SynComs) in human health has shown promising outcomes across various studies. Here, we present detailed case studies highlighting the impact of SynComs on gut health, metabolic disorders, and mental health.

 

Case Study 1: Gut Health and Disease Prevention

In a study involving patients with irritable bowel syndrome (IBS), an engineered SynCom was introduced to modulate the gut microbiota (Figure 2). The results demonstrated significant improvement in gut barrier function and a reduction in inflammatory markers. Patients reported reduced symptoms of bloating, abdominal pain, and irregular bowel movements. The intervention led to a notable increase in beneficial bacterial populations, such as Bifidobacterium and Lactobacillus, and a decrease in pathogenic bacteria. This study underscored the potential of SynComs to restore gut eubiosis and alleviate IBS symptoms (Baldi et al., 2021).

 

 

Figure 2 The contrasting states of the gut-brain axis in conditions of eubiosis (A) and dysbiosis (B) (Adapted from Baldi et al., 2021)

Image caption: In eubiosis, a healthy gut microbiota interacts beneficially with brain health, facilitating the production of AHR ligands, SCFAs, and other neuroprotective metabolites like indole and kynurenic acid, which promote gut barrier improvement and reduce inflammation. These actions are supported by the activation of antioxidant enzymes and the SIRT1 pathway, enhancing the integrity of the blood-brain barrier. Conversely, dysbiosis shows increased gut barrier permeability and a reduction in beneficial bacteria, leading to cognitive dysfunction, increased neuroinflammation, and systemic issues such as endotoxemia and low-grade inflammation, which can adversely affect brain health (Adapted from Baldi et al., 2021)

 

Case Study 2: Metabolic Health Improvement

A clinical trial focused on patients with type 2 diabetes (T2D) investigated the effects of SynComs on metabolic parameters. The trial involved administering a SynCom designed to enhance the gut microbiota's ability to produce short-chain fatty acids (SCFAs), which are crucial for metabolic health. Over a 12-week period, participants showed significant improvements in insulin sensitivity and reductions in fasting glucose levels. Inflammatory biomarkers such as C-reactive protein (CRP) were also reduced. These findings suggest that SynComs can play a vital role in managing T2D by modulating gut microbiota and improving metabolic outcomes (Tonucci et al., 2017).

 

Case Study 3: Mental Health Benefits

In a study exploring the gut-brain axis, SynComs were administered to patients with depression and anxiety. The engineered microbial communities were tailored to enhance the production of neurotransmitters like serotonin and gamma-aminobutyric acid (GABA). Over an 8-week intervention, patients reported significant reductions in symptoms of depression and anxiety, as measured by standardized scales. The study also observed increased levels of beneficial gut bacteria associated with mental health benefits, such as Lactobacillus and Bifidobacterium. These results highlight the potential of SynComs to improve mental health by targeting the gut microbiota (Pirbaglou et al., 2016).

 

These case studies illustrate the significant potential of SynComs to improve human health across various domains, including gut health, metabolic disorders, and mental health. Engineered SynComs can modulate the gut microbiota to achieve targeted health outcomes, making them a promising tool in personalized medicine and disease prevention.

 

6 Challenges and Limitations

6.1 Technical challenges in engineering and delivering SynComs

Engineering synthetic microbial communities (SynComs) for enhanced probiotic functionality and gut health improvement involves several technical challenges. One significant challenge is ensuring the stability and colonization of the engineered microbes within the gut environment. The gut is a highly dynamic ecosystem, and maintaining the desired microbial composition over time can be difficult due to horizontal gene transfer and mutations (van Leeuwen et al., 2023; Martins et al., 2023). Additionally, the complexity of the gut microbiome requires precise control over microbial interactions and functions, which is not always achievable with current synthetic biology tools (Landry and Tabor, 2017; Dou and Bennett, 2018). Another technical hurdle is the delivery of these engineered SynComs to the gut. Effective delivery systems must protect the microbes from the harsh conditions of the gastrointestinal tract and ensure their release at the appropriate site (Landry and Tabor, 2017). Moreover, the integration of advanced computational models and machine learning to predict and optimize SynCom behavior adds another layer of complexity to the engineering process (Kumar et al., 2022; Martins et al., 2023).

 

6.2 Safety and ethical considerations

The use of genetically modified microbes in SynComs raises several safety and ethical concerns. One major issue is the potential for unintended consequences, such as the horizontal transfer of engineered genes to native gut microbes or the environment, which could lead to unforeseen ecological impacts (Dou and Bennett, 2018; Martins et al., 2023). There is also the risk of engineered microbes evolving in ways that could make them harmful to the host or less effective over time (Landry and Tabor, 2017; van Leeuwen et al., 2023). Ethical considerations include the need for informed consent from individuals receiving these treatments and the potential for unequal access to these advanced therapies, which could exacerbate health disparities (Bober et al., 2018; Kumar et al., 2022). Additionally, public perception and acceptance of genetically modified organisms (GMOs) play a crucial role in the successful implementation of SynCom-based probiotics. Addressing these concerns requires robust safety assessments, transparent communication, and regulatory oversight (Bober et al., 2018; Dou and Bennett, 2018).

 

6.3 Regulatory frameworks

The regulatory landscape for SynCom-based probiotics is still evolving and presents several challenges. Current regulatory frameworks may not be fully equipped to address the unique aspects of SynComs, such as their complexity and the potential for dynamic changes within the microbial community (Landry and Tabor, 2017; Kumar et al., 2022). Regulatory agencies need to develop guidelines that ensure the safety and efficacy of these products while fostering innovation. This includes establishing standardized methods for evaluating the stability, functionality, and safety of SynComs (Bober et al., 2018; Kumar et al., 2022). Additionally, there is a need for international harmonization of regulations to facilitate the global development and distribution of SynCom-based probiotics (Bober et al., 2018; Kumar et al., 2022). Collaboration between scientists, regulatory bodies, and industry stakeholders is essential to create a regulatory environment that supports the safe and effective use of SynComs in improving gut health.

 

7 Future Directions and Perspectives

7.1 Emerging trends and technologies in SynCom engineering for gut health

The field of synthetic microbial communities (SynComs) is rapidly evolving, with several emerging trends and technologies poised to enhance gut health. One significant trend is the integration of artificial intelligence (AI) with synthetic biology (SB) to modulate the therapeutic and nutritive potential of probiotics. AI techniques are being employed to analyze metagenomic data from healthy and diseased gut microbiomes, which can inform the design of more effective SynComs (Kumar et al., 2022). Additionally, advancements in gene editing tools, such as CRISPR-Cas systems, are enabling precise engineering of probiotics for diagnostic, therapeutic, and nutritive purposes (Kumar et al., 2022).

 

Another emerging trend is the development of engineered probiotics that can produce and deliver small molecule therapeutics within the gut. These engineered probiotics are designed to replicate the complexity and adaptability of native homeostatic mechanisms, incorporating features such as bistable switches, integrase memory arrays, and logic-gating mechanisms. This approach aims to create more sophisticated and responsive therapeutic probiotics capable of accurately diagnosing and responding to various disease states.

 

7.2 Potential integration of SynComs with personalized nutrition and medicine

The integration of SynComs with personalized nutrition and medicine holds significant promise for improving gut health. Personalized nutrition involves tailoring dietary recommendations based on an individual's unique microbiome composition, genetic makeup, and health status. By leveraging SynComs, it is possible to design microbial consortia that complement an individual's specific nutritional needs and health conditions (Bober et al., 2018). This approach can enhance the efficacy of dietary interventions and promote overall gut health.

 

In the realm of personalized medicine, SynComs can be engineered to serve as living diagnostics and therapeutics. These engineered probiotics can be designed to detect specific biomarkers associated with various diseases and deliver targeted treatments accordingly (Zhou et al., 2020). For instance, SynComs can be tailored to modulate the gut microbiota to prevent and treat disorders such as inflammatory bowel disease, obesity, and neurodegenerative diseases (Zhou et al., 2020). The ability to customize SynComs for individual patients represents a significant advancement in the field of precision medicine.

 

7.3 Long-term vision and potential breakthroughs in probiotic research

The long-term vision for probiotic research involves achieving a deeper understanding of the complex interactions between the gut microbiome and host health, and leveraging this knowledge to develop more effective and sustainable therapeutic strategies. One potential breakthrough is the use of SynComs as a reductionist approach to study multispecies and multikingdom interactions within the gut ecosystem. This approach can provide valuable insights into the mechanisms governing microbe-host-immune interactions and inform the design of more effective SynComs (van Leeuwen et al., 2023).

 

Another potential breakthrough is the development of "smart probiotics" that can both diagnose and treat diseases. These engineered microbes would be equipped with sensors, genetic circuits, and output genes necessary for diagnostic and therapeutic functions (Landry and Tabor, 2017). The ability to deploy such bacteria in vivo, with considerations for sensor detection thresholds, circuit computation speed, and evolutionary stability, represents a significant advancement in the field (Landry and Tabor, 2017).

 

Overall, the future of SynCom engineering for gut health is promising, with emerging trends and technologies paving the way for more personalized, effective, and sustainable probiotic therapies. Continued research and innovation in this field will likely lead to significant breakthroughs that improve human health and well-being.

 

8 Concluding Remarks

Engineered probiotics can be designed to deliver therapeutics and respond to environmental cues within the gut, showing promise in treating diseases like inflammatory bowel disease and metabolic disorders. However, current synthetic probiotics often lack the complexity and adaptability of native gut microbiota, necessitating advanced synthetic biology tools to enhance their functionality. Furthermore, the survival and effectiveness of engineered probiotics are significantly influenced by dietary factors, such as the intake of vitamin B1, which affects the gut microbial community's composition and functionality. The integration of AI and synthetic biology has accelerated the development of probiotics with diagnostic and therapeutic capabilities, facilitating personalized medicine approaches.

 

For researchers, the findings call for the development of more complex and adaptable synthetic probiotics that can mimic the native gut microbiota. Emphasis should be placed on interdisciplinary collaboration to advance synthetic biology tools and methodologies. Healthcare providers should be aware of the new avenues engineered probiotics offer for non-invasive treatment options for various gastrointestinal and metabolic disorders, staying informed about advancements in probiotic therapies to provide cutting-edge treatments. For policymakers, there is a need to update regulatory frameworks to address the safety and efficacy of synthetic probiotics, supporting research and development in this field while ensuring public health safety.

 

To advance the field of synthetic microbial communities and their applications in gut health, further research is needed in several areas. Understanding the complex interactions within synthetic microbial communities to design more effective probiotics is essential. Leveraging AI and computational models to predict probiotic behavior and optimize their design for better health outcomes is another critical area of focus. Conducting rigorous clinical trials to evaluate the safety, efficacy, and long-term effects of synthetic probiotics in diverse populations is necessary. Additionally, addressing the ethical implications and establishing robust regulatory guidelines to ensure the safe use of engineered probiotics are crucial steps.

 

In conclusion, engineering synthetic microbial communities holds great promise for enhancing probiotic functionality and improving gut health. Continued interdisciplinary research and collaboration are essential to overcome current challenges and realize the full potential of these innovative therapies.

 

Acknowledgments

The author would like to thank the anonymous peer reviewers for their insightful and constructive feedback, which has enriched and improved the content of this study.

 

Conflict of Interest Disclosure

The author affirms that this research was conducted without any commercial or financial relationships that could be construed as a potential conflict of interest.

 

References

Arcidiacono S., Spangler J., Litteral V., Doherty L., Stamps B., Walper S., Goodson M., and Soares J., 2023, In Vitro fermentation evaluation of engineered sense and respond probiotics in polymicrobial communities, ACS Biomaterials Science & Engizneering, 9(9): 5176-5185.

https://doi.org/10.1021/acsbiomaterials.3c00630

 

Baldi S., Mundula T., Nannini G., and Amedei A., 2021, Microbiota shaping - the effects of probiotics, prebiotics, and fecal microbiota transplant on cognitive functions: a systematic review, World Journal of Gastroenterology, 27(39): 6715-6732.

https://doi.org/10.3748/wjg.v27.i39.6715

 

Bamunuarachchige T., Wickramasinghe H., Dissanayaka D., and Wickramarathna N., 2011, Genetic engineering of probiotic microorganisms, In: Liong M.T. ed., Probiotics biology, genetics and health aspects, Springer, Heidelberg, pp.109-138.

https://doi.org/10.1007/978-3-642-20838-6_5

 

Berkhout M., Zoetendal E., Plugge C., and Belzer C., 2022, Use of synthetic communities to study microbial ecology of the gut, Microbiome Research Reports, 1(1): 4.

https://doi.org/10.20517/mrr.2021.11

 

Bober J.R., Beisel C.L., and Nair N.U., 2018, Synthetic biology approaches to engineer probiotics and members of the human microbiota for biomedical applications, Annual Review of Biomedical Engineering, 20: 277-300.

https://doi.org/10.1146/annurev-bioeng-062117-121019

 

Clark R., Connors B., Stevenson D., Hromada S., Hamilton J., Amador-Noguez D., and Venturelli O., 2020, Design of synthetic human gut microbiome assembly and function, bioRxiv, 1-30.

https://doi.org/10.1101/2020.08.19.241315

 

Clark R., Connors B., Stevenson D., Hromada S., Hamilton J., Amador-Noguez D., and Venturelli O., 2021, Design of synthetic human gut microbiome assembly and butyrate production, Nature Communications, 12(1): 3254.

https://doi.org/10.1038/s41467-021-22938-y

 

Crovesy L., El-Bacha T., and Rosado E.L., 2021, Modulation of the gut microbiota by probiotics and symbiotics is associated with changes in serum metabolite profile related to a decrease in inflammation and overall benefits to metabolic health: a double-blind randomized controlled clinical trial in women with obesity, Food & Function, 12(5): 2161-2170.

https://doi.org/10.1039/d0fo02748k

 

De Souza R., Armanhi J., and Arruda P., 2020, From microbiome to traits: designing synthetic microbial communities for improved crop resiliency, Frontiers in Plant Science, 11: 553605.

https://doi.org/10.3389/fpls.2020.01179

 

Diener C., Gibbons S., and Reséndis-Antonio O., 2020, MICOM: metagenome-scale modeling to infer metabolic interactions in the gut microbiota, MSystems, 5(1): 10.1128.

https://doi.org/10.1128/mSystems.00606-19

 

Dou J., and Bennett M., 2018, Synthetic biology and the gut microbiome, Biotechnology Journal, 13(5): 1700159.

https://doi.org/10.1002/biot.201700159

 

Durmusoglu D., Al’Abri I., Williams T., Collins L., Martínez J., and Crook N., 2022, Improving therapeutic protein secretion in the probiotic yeast Saccharomyces boulardii using a multifactorial engineering approach, Microbial Cell Factories, 22(1): 109.

https://doi.org/10.1186/s12934-023-02117-y

 

Hemarajata P., and Versalovic J., 2013, Effects of probiotics on gut microbiota: mechanisms of intestinal immunomodulation and neuromodulation, Therapeutic Advances in Gastroenterology, 6(1): 39-51.

https://doi.org/10.1177/1756283X12459294

 

Huang Y., Lin X., Yu S., Chen R., and Chen W., 2022, Intestinal engineered probiotics as living therapeutics: chassis selection, colonization enhancement, gene circuit design, and biocontainment, ACS Synthetic Biology, 11(10): 3134-3153.

https://doi.org/10.1021/acssynbio.2c00314

 

Hwang I., Koh E., Wong A., March J., Bentley W., Lee Y., and Chang M., 2017, Engineered probiotic Escherichia coli can eliminate and prevent Pseudomonas aeruginosa gut infection in animal models, Nature Communications, 8(1): 15028.

https://doi.org/10.1038/ncomms15028

 

Jain S., Chatterjee A., Panwar S., Yadav A., Majumdar R., and Kumar A., 2021, Metabolic engineering approaches for improvement of probiotics functionality, Advances in Probiotics for Sustainable Food and Medicine, 225-240.

https://doi.org/10.1007/978-981-15-6795-7_10

 

Jansma J., Chatziioannou A., Castricum K., Hemert S., and Aidy S., 2023, Metabolic network construction reveals probiotic-specific alterations in the metabolic activity of a synthetic small intestinal community, MSystems, 8(5): e00332-23.

https://doi.org/10.1128/msystems.00332-23

 

Kaur H., and Ali S., 2022, Probiotics and gut microbiota: mechanistic insights into gut immune homeostasis through TLR pathway regulation, Food & Function, 13(14): 7423-7447.

https://doi.org/10.1039/d2fo00911k

 

Khan R., Roy N., Ali H., and Naeem M., 2022, Fecal microbiota transplants for inflammatory bowel disease treatment: synthetic- and engineered communities-based microbiota transplants are the future, Gastroenterology Research and Practice, (1): 9999925.

https://doi.org/10.1155/2022/9999925

 

Kho Z., and Lal S., 2018, The human gut microbiome - a potential controller of wellness and disease, Frontiers in Microbiology, 9: 356589.

https://doi.org/10.3389/fmicb.2018.01835

 

Kumar P., Sinha R., and Shukla P., 2022, Artificial intelligence and synthetic biology approaches for human gut microbiome, Critical Reviews in Food Science and Nutrition, 62(8): 2103-2121.

https://doi.org/10.1080/10408398.2020.1850415

 

Landry B.P., and Tabor J.J., 2017, Engineering diagnostic and therapeutic gut bacteria, Microbiology Spectrum, 5(5): 1-22.

https://doi.org/10.1128/microbiolspec.BAD-0020-2017

 

Li H., Zhou D., Gan R., Huang S., Zhao C., Shang A., Xu X., and Li H., 2021, Effects and mechanisms of probiotics, prebiotics, synbiotics, and postbiotics on metabolic diseases targeting gut microbiota: a narrative review, Nutrients, 13(9): 3211.

https://doi.org/10.3390/nu13093211

 

Liu Y., Qin Y., and Bai Y., 2019, Reductionist synthetic community approaches in root microbiome research, Current Opinion in Microbiology, 49: 97-102.

https://doi.org/10.1016/j.mib.2019.10.010

 

Marín O., González B., and Poupin M., 2021, From microbial dynamics to functionality in the rhizosphere: a systematic review of the opportunities with synthetic microbial communities, Frontiers in Plant Science, 12: 650609.

https://doi.org/10.3389/fpls.2021.650609

 

Martins S., Pasche J., Silva H., Selten G., Savastano N., Abreu L., Bais H., Garrett K., Kraisitudomsook N., Pieterse C., and Cernava T., 2023, The use of synthetic microbial communities (syncoms) to improve plant health, Phytopathology, 113(8): 1369-1379.

https://doi.org/10.1094/PHYTO-01-23-0016-IA

 

Naydich A., Nangle S., Bues J., Trivedi D., Nissar N., Inniss M., Neiderhuber M., Way J., Silver P., and Riglar D., 2019, Synthetic gene circuits enable systems-level biosensor discovery at the host-microbe interface, bioRxiv, 557934.

https://doi.org/10.1101/557934

 

Pirbaglou M., Katz J., Souza R., Stearns J., Motamed M., and Ritvo P., 2016, Probiotic supplementation can positively affect anxiety and depressive symptoms: a systematic review of randomized controlled trials, Nutrition Research, 36(9): 889-898.

https://doi.org/10.1016/j.nutres.2016.06.009

 

Satapathy P., Khan K., Devi A., Patil A., Govindaraju A., Gopal S., Prasad M., More V., Kakarla R., Raghu A., Hudeda S., More S., and Zameer F., 2019, Synthetic gutomics: deciphering the microbial code for futuristic diagnosis and personalized medicine, Methods in Microbiology, 46: 197-225.

https://doi.org/10.1016/BS.MIM.2019.02.001

 

Schrodt C., Mahavni A., Mcnamara G., Tallman M., Bruger B., Schwarz L., and Bhattacharyya A., 2023, The gut microbiome and depression: a review, Nutritional Neuroscience, 26(10): 953-959.

https://doi.org/10.1080/1028415X.2022.2111745

 

Stefanaki C., Peppa M., Mastorakos G., and Chrousos G., 2017, Examining the gut bacteriome, virome, and mycobiome in glucose metabolism disorders: Are we on the right track, Metabolism, 73: 52-66.

https://doi.org/10.1016/j.metabol.2017.04.014

 

Sun X., Xie J., Zheng D., Xia R., Wang W., Xun W., Huang Q., Zhang R., Kovács Á., Xu Z., and Shen Q., 2023, Metabolic interactions affect the biomass of synthetic bacterial biofilm communities, MSystems, 8(6): e01045-23.

https://doi.org/10.1128/msystems.01045-23

 

Tonucci L., Santos K., Ferreira C., Ribeiro S., Oliveira L., and Martino H., 2017, Gut microbiota and probiotics: focus on diabetes mellitus, Critical Reviews in Food Science and Nutrition, 57(11): 2296-2309.

https://doi.org/10.1080/10408398.2014.934438

 

van Leeuwen P., Brul S., Zhang J., and Wortel M., 2023, Synthetic microbial communities (SynComs) of the human gut: design, assembly, and applications, FEMS Microbiology Reviews, 47(2): fuad012.

https://doi.org/10.1093/femsre/fuad012

 

Venturelli O., Carr A., Fisher G., Hsu R., Lau R., Bowen B., Northen T., and Arkin A., 2018, Deciphering microbial interactions in synthetic human gut microbiome communities, Molecular Systems Biology, 14(6): e8157.

https://doi.org/10.15252/msb.20178157

 

Vogt S., and Finlay B., 2017, Gut microbiota-mediated protection against diarrheal infections, Journal of Travel Medicine, 24(1): S39-S43.

https://doi.org/10.1093/jtm/taw086

 

Vrancken G., Gregory A., Huys G., Faust K., and Raes J., 2019, Synthetic ecology of the human gut microbiota, Nature Reviews Microbiology, 17(12): 754-763.

https://doi.org/10.1038/s41579-019-0264-8

 

Wang C., Li Y., Li M., Zhang K., Ma W., Zheng L., Xu H., Cui B., Liu R., Yang Y., Zhong Y., and Liao H., 2021, Functional assembly of root-associated microbial consortia improves nutrient efficiency and yield in soybean, Journal of Integrative Plant Biology, 63(6): 1021-1035.

https://doi.org/10.1111/jipb.13073

 

Wang Z., Hu X., Solanki M., and Pang F., 2023, A synthetic microbial community of plant core microbiome can be a potential biocontrol tool, Journal of Agricultural and Food Chemistry, 71(13): 5030-5041.

https://doi.org/10.1021/acs.jafc.2c08017

 

Yadav M., and Shukla P., 2020, Efficient engineered probiotics using synthetic biology approaches: a review, Biotechnology and Applied Biochemistry, 67(1): 22-29.

https://doi.org/10.1002/bab.1822

 

Yadav R., Kumar V., Baweja M., and Shukla P., 2018, Gene editing and genetic engineering approaches for advanced probiotics: a review, Critical Reviews in Food Science and Nutrition, 58(10): 1735-1746.

https://doi.org/10.1080/10408398.2016.1274877

 

Yin C., Hagerty C., and Paulitz T., 2022, Synthetic microbial consortia derived from rhizosphere soil protect wheat against a soilborne fungal pathogen, Frontiers in Microbiology, 13: 908981.

https://doi.org/10.3389/fmicb.2022.908981

 

Zhou Z., Chen X., Sheng H., Shen X., Sun X., Yan Y., Wang J., and Yuan Q., 2020, Engineering probiotics as living diagnostics and therapeutics for improving human health, Microbial Cell Factories, 19: 1-12.

https://doi.org/10.1186/s12934-020-01318-z

 

International Journal of Clinical Case Reports
• Volume 14
View Options
. PDF
. FPDF(win)
. FPDF(mac)
. HTML
. Online fPDF
Associated material
. Readers' comments
Other articles by authors
. Jingqiang Wang
Related articles
. Synthetic microbial communities
. Engineered probiotics
. Gut health
. Synthetic biology
. Personalized medicine
Tools
. Post a comment