Skip to main content
  • Correspondence
  • Open access
  • Published:

Soil microbiome characterization and its future directions with biosensing

Abstract

Soil microbiome characterization is typically achieved with next-generation sequencing (NGS) techniques. Metabarcoding is very common, and meta-omics is growing in popularity. These techniques have been instrumental in microbiology, but they have limitations. They require extensive time, funding, expertise, and computing power to be effective. Moreover, these techniques are restricted to controlled laboratory conditions; they are not applicable in field settings, nor can they rapidly generate data. This hinders using NGS as an environmental monitoring tool or an in-situ checking device. Biosensing technology can be applied to soil microbiome characterization to overcome these limitations and to complement NGS techniques. Biosensing has been used in biomedical applications for decades, and many successful commercial products are on the market. Given its previous success, biosensing has much to offer soil microbiome characterization. There is a great variety of biosensors and biosensing techniques, and a few in particular are better suited for soil field studies. Aptamers are more stable than enzymes or antibodies and are more ready for field-use biosensors. Given that any microbiome is complex, a multiplex sensor will be needed, and with large, complicated datasets, machine learning might benefit these analyses. If the signals from the biosensors are optical, a smartphone can be used as a portable optical reader and potential data-analyzing device. Biosensing is a rich field that couples engineering and biology, and applying its toolset to help advance soil microbiome characterization would be a boon to microbiology more broadly.

Introduction

Soil is the foundation of all terrestrial environments; it can harbor billions of microbes per gram [1] and plays critical roles in nutrient availability, biogeochemical cycling, and bioremediation processes. Despite its importance, the soil is perhaps the least understood part of the environment [2]. Characterizing its physical and chemical properties has been essential for agriculture for thousands of years. However, soil’s biological component has generally been overlooked until the past few centuries, and even then, scientists were usually limited to studying macrofauna like earthworms and small insects. The invention of optical microscopes powerful enough to resolve microorganisms enabled the visual study of microbes [3]. Only recently, with advances in sequencing and “meta-omics” technologies, have scientists been able to investigate soil microbes and their communities comprehensively. Today, there are many ways to determine various physical and chemical properties of soil and several metrics to evaluate soil quality as a whole. However, many soil quality metrics do not consider microbial community factors [4] despite their noted importance [4, 5]. The omission of community factors in these metrics is partly due to the difficulties of biological characterization. Determining what microorganisms are present is possible through next-generation sequencing (NGS), and though these methods have advantages over traditional culturing methods, NGS is not without its drawbacks. Moreover, knowing the microbes’ functions is often essential, requiring different data with separate analysis methods. It can be challenging to interpret the large amounts of data generated by NGS, particularly when gaps remain in the databases used for analysis. Because of these challenges, many papers have called for developing novel techniques to complement NGS methods [6,7,8,9,10,11], and biosensing may offer an excellent suite of tools to do just that.

Biosensing has been defined as “a device that uses specific biochemical reactions mediated by isolated enzymes, immunosystems, tissues, organelles, or whole cells to detect chemical compounds usually by electrical, thermal or optical signals” by the International Union of Pure and Applied Chemistry (IUPAC) [12]. In essence, biosensing detects a target compound or condition using some biological component. Such devices are often used in biomedical applications [13,14,15,16,17,18] and ecotoxicology [19,20,21]. Many biosensor studies use entire organisms to sense the target compound or condition [16, 19, 22,23,24,25,26,27,28], but other studies instead use only specific molecules without the surrounding cellular structure [11, 14, 15, 29, 30]. These molecules, or bioreceptors, are essential components in biosensing devices and are often the focal points of research.

Numerous research works and commercialization efforts have been made in the past couple of decades for biosensors, most notably toward reducing the device size, device cost, assay time, operation cost, and operation complexity while maintaining specificity and sensitivity comparable to more standard laboratory instruments. Such biosensing methods and biosensor devices may identify microbial species and their makeup in the soil, significantly reducing the cost and time associated with NGS methods. However, biosensors are inherently limited in specificity compared to NGS in identifying species. Substantial cross-binding can occur with antibody- or aptamer-based bacterial biosensing. In addition, biosensing may not be optimal for identifying a large number of species simultaneously. For example, detecting 100 microbial species could require 100 different bioreceptors pre-loaded on a biosensing platform, significantly augmenting the assay complexity and limiting its usefulness compared to current NGS technology.

This correspondence paper aims to summarize current methods and techniques for soil microbiome characterization and then to summarize and provide insights and recommendations for future opportunities with biosensing technologies. See Fig. 1 and Fig. 2 for visual overviews of these two technologies. These steps are described in detail in the following sections.

Fig. 1
figure 1

Overview of next-generation sequencing (NGS) process. Amplicon-based methods include the amplification step, while shotgun methods instead include DNA fragmentation. Figure created with BioRender.com

Fig. 2
figure 2

Overview of biosensing process as potentially applied to soil microbiome characterization. Figure created with BioRender.com

Soil microbiome characterization

In its entirety, the microbiome encompasses all the parameters that make up a community of microbes. This includes physical, chemical, and biological characteristics, but we will focus on the biological characteristics, like the microbial community dynamics and structure, for this correspondence paper.

Intrinsically, soil is a difficult medium to work with because of its physical properties. Soil is an opaque mixture, which significantly limits the use of optical microscopy techniques. Soil is also very spatially heterogeneous. Depth significantly affects parameters like moisture and the carbon–nitrogen ratio [31]. Also, roots create a unique environment rich in metabolites and signaling molecules; this environment is called the rhizosphere and is a critical region of plant–microbe interaction and symbiosis [23, 32,33,34]. There is spatial heterogeneity even at the microscale, such as an uneven distribution of nutrients, organisms, and microclimate conditions across millimeters or shorter distances [35]. This heterogeneity complicates soil analyses because average properties could be misleading for understanding microscale interactions [8]; however, measuring microscale properties is technically challenging. Lastly, soil properties can be time-dependent. Not only can soil parameters change across seasons, but the day-night fluctuations can be significant, along with the short-term disturbance or perturbation [36,37,38]. In addition, many of its components influence each other; no soil properties are genuinely independent [39]. There is also no “standard” or baseline for soil properties, complicating soil comparison studies [7]. Nonetheless, scientists can make loose comparisons within a soil type, which is characterized by physical parameters and geological history. Finally, most soil testing is destructive, so measuring any parameter over time for the same soil sample is generally impossible. This limits data to be snap-shots instead of continuous [40].

The obstacles listed above are mostly matrix effects, so the first option when characterizing microbes from the soil is to try to remove those effects as much as possible. Many studies have done this when working with soil or other complex matrices like feces [19, 24, 41,42,43,44,45]. Liquid extraction of soil is a common method to remove matrix effects. The sample is usually sieved through a 2-mm sieve to remove larger dirt particles and small rocks or leaves [46,47,48,49]. Then, the soil is incubated in a liquid medium, and the supernatant is extracted for further analysis. However, liquid extraction does not extract all of the microbes, and moreover, the precise method of transport to the lab and any pre-treatments can significantly affect the microbes detected by NGS [50, 51]. The sample could also be digested via chemical and mechanical means to create a homogeneous solution; there are many commercial kits available for this purpose that have been widely used in research for many years [4,5,6, 24, 25, 30, 33, 34, 36, 37, 44, 46, 47, 50, 52,53,54,55,56,57,58,59,60,61,62,63,64,65,66,67,68,69,70,71]. There are many different brands and products available, including FastDNA SPIN kits from MP Biomedicals, EZNA DNA kits from Omega Bio-tek, DNeasy Power kits from Qiagen, NuceloSpin kits from Macherey–Nagel, ZymoBIOMICS DNA kits from Zymbo Research, and INTEST.pro kits from BIOMES. Specific products are usually tailored to detect particular types of organisms from particular matrices or environments. Even with these kits, soil remains a difficult medium to work with. Until there are standardized procedures for soil analysis, various kits should be considered for different soil samples because each has its biases, pros, and cons [50, 72,73,74].

After dealing with matrix effects and generating a sample for further analysis, the microbes can begin to be characterized, and culturing in growth media is the traditional method for this. Different colony morphologies or absorbance characteristics can be detected for characterization. In addition, culturing allows for functional or biofilm assays in the selected presence or absence of various metabolites or conditions [4, 55, 64, 72, 75]. However, many, if not most, microbes resist culturing in lab settings [76], even though many different growth media and methods have been created and used over the years. For example, the media could be missing a critical nutrient that the microbe cannot make on its own, or the growth of one species could depend on the presence of another [25, 77, 78]. Different microbes also have specific temperature, pH, moisture, oxygen, and salt concentration requirements that can be unknown before culture [76].

Some recent advances in ‘culturomics’ for soil microbes include modifying the particle size fractions and culturing in microwell plates. Such advances mimic natural spatial conditions in which organisms are separated and attached to various micro-surfaces like soil particles [76]. There are also novel in situ devices that allow natural media to permeate a membrane or filter where the microbes have been inoculated [79, 80]. These and other advances in culture techniques have enabled more species to be grown in lab settings, and indeed, these techniques will remain a central part of microbiology. Even so, sequencing technologies can overcome many of these challenges and are vital to microbiology.

Brief overview of sequencing technologies

Today, there are several different ways to sequence DNA. But, at the beginning of sequencing efforts, generating even one short DNA read was very laborious. Then, “next-generation sequencing” (NGS) arrived, which has become pivotal to microbiome research. It enabled the mass parallelization of sequencing reactions, even though read lengths were limited to several hundred base pairs. With these technologies, many strands of DNA could be read at once, which greatly shortened the time required to read lots of DNA. Then there were the “third generation sequencing” technologies which would enable longer read lengths (hundreds of thousands of base pairs) [81, 82]. Longer read lengths are especially useful for sequencing entire genomes; with longer reads there are fewer sequences to stitch together to create the whole genome. Together, these sequencing technologies exposed scientists to the wealth of previously unculturable microbes since a raw environmental sample could be sequenced without culturing and without years of work to generate one sequence. While many NGS technologies are currently in use, a few representative technologies have been selected for a comparison of their key characteristics along with strengths and weaknesses in Table 1.

Table 1 Selected current NGS technologies with their strengths and weaknesses. Adapted from [81]

As sequencing technologies grew and matured, more organisms were sequenced, from bacteria and fungi to the less studied archaea and viruses. These genes and genomes were collected and deposited into massive databases, allowing molecular evolutionary testing and visualization. These databases also enabled sequence reads of unknown species to be matched and labeled; culturing was no longer a prerequisite to determine what species were present in a sample [83, 84]. In addition, with functional data from cultures, scientists could identify genes responsible for proteins that make up enzymes in metabolic pathways. Thus, just from the organisms’ genes, microbiologists could make guesses about an organism’s functional capabilities, its role in a community, and its responses to various types of stress [85]. The importance of these advances should not be understated; they make the field of microbiology what it is today.

“-Omics”

NGS is a cornerstone of microbiology, and there are several ways to use NGS. Perhaps the simplest application of NGS is to look at one gene from one organism. For example, there are several tests for oncogenes in humans [86, 87]. Though this is much more difficult for microorganisms, single-cell sequencing technologies do exist [88]. However, this approach only lets scientists learn about one part of one individual; it does not provide a full picture of one organism, nor is this approach suitable for community analysis.

To understand the individual more fully, scientists can look at all the genes in one individual, called genomics. Such an approach could be useful if one species is determined to be a keystone or indicator species for a microbiome. The genome can be very insightful for determining the potential function of an organism. However, the presence of genes alone does not guarantee that they function at all or as expected [89]. Analyzing all of the messenger RNA (mRNA) is a better way to determine gene activity, while characterizing the proteins or metabolites produced by an organism is a more concrete way to assess its functional ability [90]. These fields of study are called transcriptomics, proteomics, and metabolomics, respectively, and together with genomics, represent the core of the broader -omics field.

Ideally, each species study would include all of these -omics, but that is not feasible. There are general time and money limitations, but besides those, each -omics investigates different molecule types that work on different time scales and require different extraction and analysis methods [74]. In particular, proteomics and metabolomics commonly involve mass spectrometry [15, 37, 40, 77, 85, 91, 92], a highly technical and precise method requiring many non-standardized pre-processing steps. In addition, it is also difficult to compare mass spectrometry results across laboratories because their results depend on the previously tested libraries of molecules for that specific mass spectrometry machine and sample pre-processing method [93].

Metabarcoding

Since -omics is focused on the individual, a more suitable application of NGS for microbiome research is metabarcoding, which looks at the breadth of the community and how its members are related phylogenetically. Metabarcoding uses specific sequences to identify what species are present and how they are related. This is typically done using polymerase chain reaction (PCR) to amplify an identifier gene like the 16S ribosomal RNA (16S rRNA) or internal transcribed spacer (ITS) gene throughout the sample (Fig. 1). Thus, the other genes are ignored, and only taxonomic information is extracted for analysis. However, care must be taken when selecting not only the DNA extraction method [44] but also the PCR primers, which can affect what taxa are detected and in what relative abundances [58, 74]. Since primers can internally differentiate some groups better than others, they inherently have more or less affinity for specific species or taxa. For example, for looking at fungi, amplifying part or all of the ITS gene is more useful than the 16S rRNA gene, while for bacteria, the 16S rRNA gene works well, though there are several variable regions to choose from [44].

After PCR, the amplified DNA is sequenced via one of several different sequencing technologies, each with its strengths and weaknesses (Table 1). Examining these methods is outside the scope of this review (the main emphasis is biosensing methods), but some excellent reviews have been completed [81, 94]. The sequence data must be processed via bioinformatics pathways. Again, there are many different ways to do this, and analyzing them all is outside the scope of this review. Finally, a collection of cleaned and annotated reads is returned to the user, and the scientist can see which taxa are present and in what relative abundances. However, these taxa are often not identified at the species level [95, 96].

After metabarcoding and omics data are generated, they must be analyzed. The chosen bioinformatics pathway will affect the analysis. In the past, these pathways were not standardized, though there are now substantial efforts to enhance standardization across labs and organizations [97]. Still, one drawback of metabarcoding and omics remains: they often rely on data collected from culture. Current data analysis tools often depend on previously studied organisms [7]; however, modern bioinformatics tools enable less reliance on culture data, and in some cases, genomes can be constructed de novo without prior knowledge of the organisms [98]. Still, culturing remains a critical technique, and advances must be made in that field to address the more intractable taxa.

Meta-Omics

For soil microbiome characterization, metabarcoding only gives scientists insight on what taxa or species are present; it does not give direct insight on what they are doing or how. To find out, scientists will study the -omics of many microbes, which introduces “meta-omics,” in which community-wide data is taken and studied in aggregate. For example, metatranscriptomics looks at the mRNA of a community, metaproteomics looks at the proteins in a community, metabolomics looks at the metabolites in a community, and metagenomics looks at all the genomes in a community. Excellent reviews of these meta-omics have been published [99,100,101,102,103], and in this section, metagenomics will be used as an example of meta-omics use more broadly and its limitations.

Metagenomics is a zoomed-out view of genes and their potential functions in a microbiome. Microbiologists can look at the distribution of classes of genes throughout time or by taxa, or they can compare one metagenome from one community to another. However, meta-omics inherits many limitations of -omics, primarily related to feasibility. The sequence depth must be sufficiently deep to detect all of the genes of an entire community; a lot of DNA must be sequenced to get adequate coverage. Gathering this much DNA requires more precise pre-processing steps of the sample, and even so, detecting genes and genomes at low abundances is still challenging [104]. Meta-omics, like -omics, involves lots of data that require significant computing power to analyze, and this, rather than the ability to sequence, is now regarded as the limiting step for metagenomics studies [72], and several machine learning models have been developed for these analyses [60, 71, 85, 105,106,107]. In addition, meta-omics methods are not standardized, so comparing meta-omics data across labs is challenging because the pipelines are different, both for data generation and processing [72]. Lastly, as with -omics, even though culturing is not required to gather data, the analysis generally relies on previous culture data for labeling and interpretation; however, some de novo bioinformatics tools have been developed and tested to circumvent using any culture data [83, 84].

There are also more general obstacles to environmental sequence or molecule studies. Firstly, most of these technologies require the samples to be taken to a lab and processed in a clean environment; these methods are neither fast nor suitable for point-of-care or in-situ monitoring programs. Also, meta-omics methods are typically too expensive for every lab to have access to them. Moreover, since DNA is relatively stable and can be bound up in inorganic complexes in the soil, DNA from dead organisms can be found and mistakenly interpreted as if it were from living organisms [108]. However, treatments like propidium monoazide can remove relic DNA from a sample [109, 110]. Finally, standard NGS only provides relative abundance data; to determine absolute abundance, techniques like quantitative PCR (qPCR), digital PCR, or spike-ins with known microbial concentration are required [111].

Biosensing technology

Researchers have long understood the limitations of NGS for microbiome studies, and there have been calls for developing novel techniques to help overcome these limitations [6,7,8,9,10,11]. We argue that biosensing is a vital avenue to consider in this endeavor. While biosensing has had many definitions over the years, a biosensor can be defined as a device or method that uses a biologically derived recognition element, or bioreceptor, to help detect or quantify a target molecule or condition [12]. Biosensors are inherently interdisciplinary devices incorporating chemistry, physics, engineering, biology, and sometimes nanotechnology. This field, like NGS, can be very useful for investigating the microbial world; however, few examples of biosensors applied to the soil microbiome have been published at the time of this manuscript’s submission. Previously, biosensors have primarily been applied to the biomedical and ecotoxicology fields. However, biosensing has much to offer soil microbiology. Though many reviews are already available for detailed examinations of different biosensor examples, here we will discuss broad categories of biosensor types with potential uses in soil microbiology. Table 2 summarizes standard biosensors and novel technologies most suited for soil microbiome characterization. The following section will examine more direct applications to the soil microbiome.

Table 2 Biosensing applied to the soil microbiome

Biosensing, like sequencing, is not a new technology. Though the classification of biosensors can be a bit nebulous, they can be categorized by the biological recognition element, or bioreceptor, and the signal they produce. The bioreceptor chosen significantly impacts the biosensor function. Enzymes are often used to detect metabolites (e.g., glucose monitor), and antibodies were used to detect target cells or viruses (e.g., rapid COVID-19 test). These two classes of molecules can be very sensitive and selective in their binding. However, purifying enzymes and generating antibodies are costly and time-intensive processes. Moreover, enzymes and antibodies are unstable and must be kept in controlled conditions. Otherwise, they denature and become inactive. A newer group of bioreceptor molecules are aptamers (Fig. 2). These nucleic acid polymers can specifically bind to targets, just like antibodies. These molecules are more stable than antibodies and more inexpensive to produce. Moreover, they do not require animals or an immune response in their production. However, it is difficult for a single aptamer to bind a unique microbial species consistently, so recent work has generated aptamer libraries that collectively bind a species with high specificity and sensitivity [6, 29, 44, 68]. There are other types of bioreceptors that are much bigger and more complex. Some studies use entire organisms, and these organisms are often “model” organisms, like E. coli cells or even mice [24, 112,113,114], that have been well characterized over many years of research. Some studies will genetically modify these reporter organisms to produce specific responses to the target molecules or conditions [114].

While the choice of bioreceptor is a critical design parameter, biosensing is also classified based on the type of signal they produce, which can be broadly divided into electrochemical or optical (Fig. 2). Electrochemical biosensors have been used extensively in the past couple of decades, and these sensors report biological activity via an electric signal. The classic biosensor that has been the field’s flagship is the glucose monitor [14]. This electrochemical device measures the electron flow caused by enzymes interacting with glucose to determine how much glucose is present quantitatively. It is called an amperometric biosensor because it detects current. Another example is the potentiometric biosensor that measures voltage instead of current; it measures the difference in potential, or voltage, between two or more electrodes. One benefit of potentiometric sensing is that they generally do not consume the analytes; instead, these can be reused for multiple rounds of sensing. These are often used for the new class of wearable biosensors that can continuously monitor subsurface conditions [115, 116]. Voltammetric sensors report both current and voltage changes. Finally, electrical impedance spectroscopy (EIS) reports the impedance signal from an electrode, which is a complex number version of electrical resistance resulting from alternating current (AC). All of these have been applied to detect target analytes, and their success largely depends on the construction of the electrodes themselves. Generating the correct chemistry and understanding the electronics of the primary and secondary interactions is crucial to these biosensors.

The other large class of signals that biosensors produce is optical. Optical methods are varied, but one classic example is the lateral flow immunoassay (LFIA) [117]. For LFIA, a liquid sample is placed on a paper-based test strip, and the sample flows through the strip along with pre-deposited particles (gold nanoparticles or fluorescent polymeric particles) conjugated with bioreceptors (usually antibodies). The target molecule is sandwiched between the surface-bound bioreceptors and the bioreceptor particles at the test line, like in the enzyme-linked immunosorbent assay (ELISA). The presence of bioreceptor particles (and subsequently the target presence) can be confirmed visually via pink coloration (from gold nanoparticles) or fluorescence (from fluorescent polymeric particles). A commercial example of this is the urine-based pregnancy test. Though often designed to report a binary result, if the test and control bands are examined for fluorescent intensity, this method can also be used to generate quantitative data.

ELISA is a predecessor to LFIA, typically conducted in a lab setting, with better sensitivity and specificity. ELISA uses the sandwich method, with an antibody as a bioreceptor and an enzyme–substrate pair as a signaling molecule [118]. The latter can sometimes be replaced with a fluorescent dye conjugated with antibody-to-antibody. It is typically conducted on a microwell plate. While binding and washing are performed automatically via the capillary action through paper pores in LFIA, ELISA utilizes pipetting for adding and rinsing reagents. Automated pipettor systems are often used to automate and multiplex ELISA. Detection is conducted optically, typically utilizing a microplate reader.

Other optical biosensors include using whole, live cells that often contain engineered genetic circuits. These genetic circuits couple the genes of natural detection molecules with transgenic modules to produce a fluorescently active molecule like green fluorescence protein (GFP), luciferase, or violacein. Several studies detected quorum-sensing molecules or cell stress responses using these types of bioreceptors [23,24,25, 119,120,121]. These genetic circuits can even contain genetic logic such that the response is only generated when multiple conditions are met or a response is linearly dependent on a target concentration [122]. Genetic circuits have been made to detect and respond to various signals, and recent studies have shown the ability to produce other classes of molecules at target detection [123, 124].

In recent years, nanomaterials have become an important part of biosensing research. Because of their high surface-to-volume ratios, they can accommodate more bioreceptors on a sensor surface or generate stronger electrochemical (or unique optical) signals [125]. Various nanomaterials also allow for customization and functionalization of their structure and surfaces [126]. They can also enter organisms and interact with their internal systems [126]. Quantum dots and carbon dots fluoresce and have been used for detecting targets [10, 127], and nanodiamonds have been used to prevent biofouling of submerged oxygen sensors [128].

Biosensing design considerations for the soil microbiome

Meta-omics technologies are excellent tools for microbiology, but they have limitations that biosensing can help address. Biosensors can be applied to soil microbiome characterization to add another tool to the microbiologist’s toolbelt. Microbiome biosensing should strive for these characteristics: multiplexing [6, 18, 129, 130], stability of the biosensor itself in the field or non-ideal laboratory conditions, and ease of use and manufacture. These ideas will be discussed below, followed by more specific potential directions.

To understand the community of a microbiome, knowing and understanding one aspect by itself is not very useful in understanding community dynamics; thus, multiplexed analyses are critical. In other words, multiple targets or conditions must be analyzed collectively for a given sample. Multiplexing is known to be essential and has been implemented in many studies and products over the years (Fig. 2). There are commercially available functional assays that are multiplexed. For example, multi-well microplates determine what kinds of substrates organisms in a sample can digest and process [55]. Often, the signal is a visual intensity originating from each well on these plates. Imaging the entire plate and analyzing the color intensities from each well has been the general practice, typically using a microplate reader. For simplicity or field use, this microplate reader can easily be replaced with a smartphone camera [118]. Functional assays require a culturing step for the organisms to degrade the provided substrates successfully, and as stated previously, culturing can introduce bias. Multiplex terminal restriction fragment length polymorphism is another common method that uses multiplexing to detect a “signature,” “barcode,” or “fingerprint” of the community, and again the raw data for this method is a visual image [46, 55, 133] (Fig. 2). This method involves many steps that must be completed in a lab setting. One study looked at generating an assay of antibodies to provide a signature of a disease state [9]; this study used a machine learning algorithm to process their image data and determine data trends. Multiplexing has already been successfully applied to microbiome analysis, but these methods are thus far limited to laboratory analyses. They have not been developed for field use, and most have not been designed for rapid analysis. Because the soil microbiome can change quickly over time, detecting characteristics in situ is essential. Biosensors attempting to characterize the soil microbiome should take these existing multiplexing technologies and apply them to field detection platforms that can quickly collect data.

Several factors need to be considered to develop a field-ready biosensor. Firstly, the effects of the soil matrix must be considered. As mentioned previously, common practice is to remove the matrix effects via a liquid extraction method. This step alone could require extensive research and experimentation to determine the optimal way to extract meaningful analytes from the soil while minimizing bias and process effects. However, because liquid extraction is currently standard, that is where any research into that method ought to start. After matrix effects have been overcome, the bioreceptor must tolerate fluctuating temperatures, pH, and humidity/moisture levels. This likely excludes enzymes and antibodies from these sensors, but aptamers, modified electrodes, and genetic circuits in robust microbes could still be useful (Table 2). The bioreceptor should also be specific and sensitive enough to detect the target in complex samples at environmentally relevant concentrations. This requires rigorous testing and careful selection of bioreceptors and overall biosensing methods for the desired target in its natural or minimally modified state. In addition, the signal must be comparably detectable in various field conditions; otherwise, the biosensor can only be used in limited areas or situations. For optical signals, the environmental lighting should not significantly affect the data. Either electrochemical data should be collected instead, or the optical signals should be corrected for ambient lighting variations. Another possibility is to have a small, lighting-controlled space around the sensor to take that image or other optical data. Electrochemical sensing can also have challenges in field conditions. Biofouling is a common problem, as is electrode degradation. Nanodiamonds and carbon nanotubes have been shown to limit biofouling [128, 134]. In addition, the electrodes can be coated with various layers to improve performance [135], and electrode degradation can be minimized with material selection and manufacturing processes [136].

Potential biosensing applications to soil microbiome characterization

Multiplexing, field deployment, ease of use, and ease of manufacture are simple to discuss but can be tricky to implement. Still, researchers should consider those ideas when developing novel biosensing methods for characterizing soil microbiomes. Specific examples of biosensing applied to the soil microbiome are scarce, but several papers noted potential directions or future projects combining these fields. Examples are discussed below.

There are recognized challenges with optical methods, and electrochemical sensors have some advantages. Amperometric detection can be broadly applied; this method allows for low detection limits with reasonably simple devices, and current is easy to analyze relative to optical images. One issue with this approach is biofouling when the electrodes become impeded by organic matter build-up, though this can be overcome with nanomaterials, chemical layers, and material selection [128, 134,135,136]. Several studies have commented on the possibility of an aptamer-based electrochemical sensor [44, 68]. Perhaps an amperometric assay could be developed to sense a variety of metabolites all at once on a multi-well plate. These studies could follow the method [9] in which certain bioreceptors were bound to different areas of a larger plate. Thus, after applying the sample, each distinct plate region emits different signals to generate a sample signature. Detecting metabolites in this way would reduce the need for mass spectrometry, which is a technically challenging method to implement.

There are other ways to multiplex, like having different carbon dots with different colors for different targets [127]. Alternatively, studies have been conducted with microscopic beads coated with bioreceptors, and these have been used for multiplexed analysis when the beads have a variety of detection targets [91]. The LFIA method could be expanded to use various aptamers; perhaps there would be a way to create a multiplexed LFIA, e.g., multi-channel paper microfluidic chips [137]. If several channels radiated out from a central sample pad, it could be possible to seed different bioreceptor-coated beads at the beginning of each channel [138]. LFIA and paper microfluidic chips could be particularly insightful when the color intensity of the test band is quantified with an optical reader. Even if the data is purely qualitative, LFIA and paper microfluidic chips can provide results in minutes to enable field assessment of those targets.

A smartphone camera can be used to analyze optical signals rapidly and in the field from the LFIA and paper microfluidic chips; this is a viable method in previous works, particularly when used with a portable microscope (including a smartphone-based fluorescence microscope) [138,139,140,141,142]. The smartphone could be a vital component of a field-deployable biosensor because it is a mini-computer. It can take in high-quality visual data and run data analysis through a custom app. Or, if there is an internet connection, data could be remotely uploaded and processed on the cloud before researchers even get back to the lab. Figure 3 shows an example of analyzing bacterial species from environmental water samples, utilizing a set of peptides extracted from bacterial biofilms, a multi-channel paper microfluidic chip, a smartphone camera as an optical reader and data processing unit, and machine learning to analyze the data [115].

Fig. 3
figure 3

An example of analyzing bacterial species from environmental water samples. It utilizes a set of peptides extracted from bacterial biofilms, a multi-channel paper microfluidic chip, a smartphone camera as an optical reader and data processing unit, and machine learning to analyze the data. Reprinted with permission from [141].

Copyright 2021, Elsevier

Regardless of the data being collected, machine learning will likely be needed to analyze these larger, multiplexed datasets. Several papers have successfully implemented machine learning, and many compare two or more algorithms to see which is better for their dataset [141, 143,144,145,146]. Machine learning has also been used with NGS datasets [83], and many algorithms have been used with both NGS and biosensor data. A comparison of algorithms used is shown in Fig. 4.

Fig. 4
figure 4

Venn diagram showing a sample of machine learning algorithms applied to NGS and biosensor data. Most NGS algorithms found in the literature were supervised learning methods except for PCA. There were also several papers that detailed novel algorithms for NGS data. A great variety of algorithms are used with biosensor data, and many of them are also used with NGS data; there is significant overlap. ANN = artificial neural networks; kNN = k-nearest neighbors; SVM = support vector machine; XGBoost = extreme gradient boosting; LR = logistic regression; PCA = principal component analysis; LDA = linear discriminant analysis

The standard practices of removing the matrix and then destroying the sample during analysis are inadequate for studying the soil microbiome in its natural matrix. The field of wearable biosensors could address the issue of destructive sampling. These devices are meant to continuously measure metabolites on or in the human body via electrochemical sensing [115, 116], and this technology could be applied to continuous monitoring of targets in the soil. One paper analyzed gases released from soil to investigate the soil microbiome [40], and there seems to be great promise in gas analysis for microbiome characterization [147] (Fig. 5). Gas molecules and their concentrations can reveal respiration rates and other metabolic processes [147] while maintaining the soil matrix’s natural state. This minimizes processing effects while simplifying the biosensor and its method of use. Perhaps the genetic circuit technologies of fluorescent, whole-cell biosensors, which would have little direct application in opaque soil, could be reconfigured to emit alternate signals like gaseous metabolites that could pass through the soil matrix more easily than optical signals.

Fig. 5
figure 5

An example of a gas sensor for assessing the microbiome. While this work was designed to assess the intestinal flora, e.g., gut microbiome, it can be adapted to the soil microbiome. Reprinted from [147] under Creative Commons Attribution License

Our group recently applied biosensors to the soil microbiome. This study used induced natural fluorescence, a smartphone to capture optical intensity, and machine learning to categorize bacteria derived from the soil matrix. See Fig. 6 for a depiction of the process used. This process could be a model for future works applying biosensors to soil microbiome characterization.

Fig. 6
figure 6

An example of optical biosensor to soil microbiome characterization. A smartphone is modified with an acrylic film wheel (12 filters) and three LEDs to capture multiple autofluorescence images of various bacterial mixtures and soil samples. Learning databases are collected from various bacterial mixtures, and the field soil samples are used as a test set to predict the dominant bacterial species

Conclusion

In conclusion, the field of biosensing has a lot to offer soil microbiome characterization. NGS and meta-omics methods are central to microbiology, and they can generate data for in-depth analyses of communities and community structures via DNA and RNA analyses. However, these technologies typically require extensive time, money, expertise, and computing power to be effective. Moreover, these techniques are restricted mainly to controlled laboratory conditions; they are not applicable in field settings, nor can they rapidly generate data. Biosensing technology can be applied to soil microbiome characterization to overcome these limitations and to complement NGS and meta-omics techniques more broadly. Biosensing as a field has not been applied to the soil microbiome until recently, and some methods are better suited for this new application. Aptamers are more stable than enzymes or antibodies and are more appropriate for field-use biosensors. Given that any microbiome is a highly complex system, a multiplex sensor will be needed, and with large, complicated datasets, machine learning might benefit the analysis. If the signals from the biosensors are optical, a smartphone can be used as a portable optical reader and potential data-analyzing device; however, optical signals can be affected by environmental lighting conditions. In contrast, electrical signals might suffer less distortion in the field. Biosensing is a rich field that couples engineering and biology, and applying its toolset to help advance soil microbiome characterization would be a boon to microbiology more broadly.

Availability of data and materials

Not applicable.

References

  1. Whitman WB, Coleman DC, Wiebe WJ. Prokaryotes: The unseen majority. Proc Natl Acad Sci U S A. 1998;95:6578–83. https://doi.org/10.1073/pnas.95.12.6578.

    Article  Google Scholar 

  2. Mishra A, Singh L, Singh D. Unboxing the black box—one step forward to understand the soil microbiome: A systematic review. Microb Ecol. 2023;85:669–83. https://doi.org/10.1007/s00248-022-01962-5.

    Article  Google Scholar 

  3. Coyne MS. A cartoon history of soil microbiology. J Nat Res Life Sci Educ. 1996;25:30–6. https://doi.org/10.2134/jnrlse.1996.0030.

    Article  Google Scholar 

  4. Cha JY, Han S, Hong HJ, et al. Microbial and biochemical basis of a Fusarium wilt-suppressive soil. ISME J. 2016;10:119–29. https://doi.org/10.1038/ismej.2015.95.

    Article  Google Scholar 

  5. Durán P, Jorquera M, Viscardi S, Carrion VJ, Mora M de la L, Pozo MJ. Screening and characterization of potentially suppressive soils against Gaeumannomyces graminis under extensive wheat cropping by Chilean indigenous communities. Front Microbiol. 2017;8:1552. https://doi.org/10.3389/fmicb.2017.01552.

  6. Zhang Y, Xing H, Bolotnikov G, Krämer M, Gotzmann N, Knippschild U, Kissmann AK, Rosenau F. Enriched aptamer libraries in fluorescence-based assays for Rikenella microfusus-specific gut microbiome analyses. Microorganisms. 2023;11:2266. https://doi.org/10.3390/microorganisms11092266.

    Article  Google Scholar 

  7. Nesme J, Achouak W, Agathos SN, et al. Back to the future of soil metagenomics. Front Microbiol. 2016;7:73. https://doi.org/10.3389/fmicb.2016.0007.

    Article  Google Scholar 

  8. Jansson JK, Hofmockel KS. The soil microbiome — from metagenomics to metaphenomics. Curr Opin Microbiol. 2018;43:162–8. https://doi.org/10.1016/j.mib.2018.01.013.

    Article  Google Scholar 

  9. Maltz-Matyschsyk M, Melchiorre CK, Herbst KW, et al. Development of a biomarker signature using grating-coupled fluorescence plasmonic microarray for diagnosis of MIS-C. Front Bioeng Biotechnol. 2023;11:1066391. https://doi.org/10.3389/fbioe.2023.1066391.

    Article  Google Scholar 

  10. Radwan O, Chapleau ME, Chapleau RR, Ruiz ON. Discovery, development and implementation of biomarker-specific peptide BRE for biosensing hydrocarbon-degrading fungi. Int Biodeterior Biodegradation. 2023;178: 105568. https://doi.org/10.1016/j.ibiod.2023.105568.

    Article  Google Scholar 

  11. Genee HJ, Bali AP, Petersen SD, Siedler S, Bonde MT, Gronenberg LS, Kristensen M, Harrison SJ, Sommer MOA. Functional mining of transporters using synthetic selections. Nat Chem Biol. 2016;12:1015–22. https://doi.org/10.1038/nchembio.2189.

    Article  Google Scholar 

  12. IUPAC. Biosensor: The IUPAC Compendium of Chemical Terminology. 2014. https://doi.org/10.1351/goldbook.B00663.

  13. Chen LC, Wang E, Tai CS, Chiu YC, Li CW, Lin YR, Lee TH, Huang CW, Chen JC, Chen WL. Improving the reproducibility, accuracy, and stability of an electrochemical biosensor platform for point-of-care use. Biosens Bioelectron. 2020;155: 112111. https://doi.org/10.1016/j.bios.2020.112111.

    Article  Google Scholar 

  14. Lee I, Probst D, Klonoff D, Sode K. Continuous glucose monitoring systems - Current status and future perspectives of the flagship technologies in biosensor research -. Biosens Bioelectron. 2021;181: 113054. https://doi.org/10.1016/j.bios.2021.113054.

    Article  Google Scholar 

  15. Patnode ML, Beller ZW, Han ND, et al. Interspecies competition impacts targeted manipulation of human gut bacteria by fiber-derived glycans. Cell. 2019;179:59–73. https://doi.org/10.1016/j.cell.2019.08.011.

    Article  Google Scholar 

  16. Woo SG, Moon SJ, Kim SK, et al. A designed whole-cell biosensor for live diagnosis of gut inflammation through nitrate sensing. Biosens Bioelectron. 2020;168: 112523. https://doi.org/10.1016/j.bios.2020.112523.

    Article  Google Scholar 

  17. Yang G, Xiao Z, Tang C, Deng Y, Huang H, He Z. Recent advances in biosensor for detection of lung cancer biomarkers. Biosens Bioelectron. 2019;141: 111416. https://doi.org/10.1016/j.bios.2019.111416.

    Article  Google Scholar 

  18. Zhang S, Rong F, Guo C, Duan F, He L, Wang M, Zhang Z, Kang M, Du M. Metal–organic frameworks (MOFs) based electrochemical biosensors for early cancer diagnosis in vitro. Coord Chem Rev. 2021;439: 213948. https://doi.org/10.1016/j.ccr.2021.213948.

    Article  Google Scholar 

  19. Sochor J, Zitka O, Hynek D, et al. Bio-sensing of cadmium(II) ions using Staphylococcus aureus. Sensors. 2011;11:10638–63. https://doi.org/10.3390/s111110638.

    Article  Google Scholar 

  20. Terekhova VA. Biotesting of soil ecotoxicity in case of chemical contamination: Modern Approaches to integration for environmental assessment (a review). Eurasian Soil Sci. 2022;55:601–12. https://doi.org/10.1134/S106422932205009X.

    Article  Google Scholar 

  21. Zhang H, Li A, Wei Y, Miao Q, Xu W, Zhao B, Guo Y, Sheng Y, Yang Y. Development of a new methodology for multifaceted assessment, analysis, and characterization of soil contamination. J Hazard Mater. 2022;438: 129542. https://doi.org/10.1016/j.jhazmat.2022.129542.

    Article  Google Scholar 

  22. Baranova MN, Babikova PA, Kudzhaev AM, Mokrushina YA, Belozerova OA, Yunin MA, Kovalchuk S, Gabibov AG, Smirnov IV, Terekhov SS. Live biosensors for ultrahigh-throughput screening of antimicrobial activity against gram-negative bacteria. Antibiotics. 2021;10:1161. https://doi.org/10.3390/antibiotics10101161.

    Article  Google Scholar 

  23. Begum JF, Tamilarasi M, Pushpakanth P, Balachandar D. A simple method for direct isolation of N-acyl-L-homoserine lactone mediated biofilm-forming rhizobacteria from roots. J Microbiol Meth. 2019;156:34–9. https://doi.org/10.1016/j.mimet.2018.11.018.

    Article  Google Scholar 

  24. Lv D, Ma A, Tang X, Bai Z, Qi H, Zhuang G. Profile of the culturable microbiome capable of producing acyl-homoserine lactone in the tobacco phyllosphere. J Environ Sci. 2013;25:357–66. https://doi.org/10.1016/S1001-0742(12)60027-8.

    Article  Google Scholar 

  25. Reen FJ, Gutiérrez-Barranquero JA, McCarthy RR, Woods DF, Scarciglia S, Adams C, Fog Nielsen K, Gram L, O’Gara F. Quorum sensing signaling alters virulence potential and population dynamics in complex microbiome-host interactomes. Front Microbiol. 2019;10:2131. https://doi.org/10.3389/fmicb.2019.02131.

    Article  Google Scholar 

  26. Thilakarathna MS, Raizada MN. A biosensor-based assay (GlnluX-Agar) shows defoliation triggers rapid release of glutamine from nodules and young roots of forage legumes. Phytobiomes J. 2019;3:85–91. https://doi.org/10.1094/PBIOMES-03-19-0014-R.

    Article  Google Scholar 

  27. Vaaben TH, Vazquez-Uribe R, Sommer MOA. Characterization of eight bacterial biosensors for microbial diagnostic and therapeutic applications. ACS Synth Biol. 2022;11:4184–92. https://doi.org/10.1021/acssynbio.2c00491.

    Article  Google Scholar 

  28. Wang J, Zhang C, Childers WS. A biosensor for detection of indole metabolites. ACS Synth Biol. 2021;10:1605–14. https://doi.org/10.1021/acssynbio.1c00090.

    Article  Google Scholar 

  29. Raber HF, Kubiczek DH, Bodenberger N, et al. Flucell-selex aptamers as specific binding molecules for diagnostics of the health relevant gut bacterium akkermansia muciniphila. Int J Mol Sci. 2021;22:10425. https://doi.org/10.3390/ijms221910425.

    Article  Google Scholar 

  30. Sun H, Xu M, Wu S, Dong R, Angelidaki I, Zhang Y. Innovative air-cathode bioelectrochemical sensor for monitoring of total volatile fatty acids during anaerobic digestion. Chemosphere. 2021;273: 129660. https://doi.org/10.1016/j.chemosphere.2021.129660.

    Article  Google Scholar 

  31. Naumova NB, Belanov IP, Alikina TY, Kabilov MR. Undisturbed soil pedon under birch forest: Characterization of microbiome in genetic horizons. Soil Syst. 2021;5:14. https://doi.org/10.3390/soilsystems5010014.

    Article  Google Scholar 

  32. Maldonado JE, Gaete A, Mandakovic D, Aguado-Norese C, Aguilar M, Gutiérrez RA, González M. Partners to survive: Hoffmannseggia doellii root-associated microbiome at the Atacama Desert. New Phytol. 2022;234:2126–39. https://doi.org/10.1111/nph.18080.

    Article  Google Scholar 

  33. Azevedo-Silva D, Rasmussen JA, Carneiro M, Gilbert MTP, Azevedo H. Feasibility of applying shotgun metagenomic analyses to grapevine leaf, rhizosphere and soil microbiome characterisation. Aust J Grape Wine Res. 2021;27:519–26. https://doi.org/10.1111/ajgw.12508.

    Article  Google Scholar 

  34. Doherty JR, Crouch JA, Roberts JA. Elucidating the influence of resident seed and soil microbiota on the developing creeping bentgrass microbiome. Agrosyst Geosci Environ. 2020;3: e20038. https://doi.org/10.1002/agg2.20038.

    Article  Google Scholar 

  35. Cordero OX, Datta MS. Microbial interactions and community assembly at microscales. Curr Opin Microbiol. 2016;31:227–34. https://doi.org/10.1016/j.mib.2016.03.015.

    Article  Google Scholar 

  36. Nevins CJ, Nakatsu C, Armstrong S. Characterization of microbial community response to cover crop residue decomposition. Soil Biol Biochem. 2018;127:39–49. https://doi.org/10.1016/j.soilbio.2018.09.015.

    Article  Google Scholar 

  37. Xue P, Liu X, Zhao L, Zhang J, He Z. Integrating high-throughput sequencing and metabolomics to investigate the stereoselective responses of soil microorganisms to chiral fungicide cis-epoxiconazole. Chemosphere. 2022;300: 134198. https://doi.org/10.1016/j.chemosphere.2022.134198.

    Article  Google Scholar 

  38. Agam N, Berliner PR. Diurnal water content changes in the bare soil of a coastal desert. J Hydrometeorol. 2004;5:922–33. https://doi.org/10.1175/1525-7541(2004)005/3C0922:DWCCIT/3E2.0.CO;2.

    Article  Google Scholar 

  39. Pershina EV, Ivanova EA, Korvigo IO, Chirak EL, Sergaliev NH, Abakumov EV, Provorov NA, Andronov EE. Investigation of the core microbiome in main soil types from the East European plain. Sci Total Environ. 2018;631–632:1421–30. https://doi.org/10.1016/j.scitotenv.2018.03.136.

    Article  Google Scholar 

  40. Kim J, Goldstein AH, Chakraborty R, Jardine K, Weber R, Sorensen PO, Wang S, Faybishenko B, Misztal PK, Brodie EL. Measurement of volatile compounds for real-time analysis of soil microbial metabolic response to simulated snowmelt. Front Microbiol. 2021;12: 679671. https://doi.org/10.3389/fmicb.2021.679671.

    Article  Google Scholar 

  41. Zolti A, Green SJ, Sela N, Hadar Y, Minz D. The microbiome as a biosensor: Functional profiles elucidate hidden stress in hosts. Microbiome. 2020;8:71. https://doi.org/10.1186/s40168-020-00850-9.

    Article  Google Scholar 

  42. Popov IV, Mazanko MS, Kulaeva ED, et al. Gut microbiota of bats: pro-mutagenic properties and possible frontiers in preventing emerging disease. Sci Rep. 2021;11:21075. https://doi.org/10.1038/s41598-021-00604-z.

    Article  Google Scholar 

  43. Zúñiga A, Muñoz-Guamuro G, Boivineau L, Mayonove P, Conejero I, Pageaux GP, Altwegg R, Bonnet J. A rapid and standardized workflow for functional assessment of bacterial biosensors in fecal samples. Front Bioeng Biotechnol. 2022;10: 859600. https://doi.org/10.3389/fbioe.2022.859600.

    Article  Google Scholar 

  44. Xing H, Zhang Y, Krämer M, et al. A polyclonal aptamer library for the specific binding of the gut bacterium Roseburia intestinalis in mixtures with other gut microbiome bacteria and human stool samples. Int J Mol Sci. 2022;23:7744. https://doi.org/10.3390/ijms23147744.

    Article  Google Scholar 

  45. Moraskie M, Roshid MHO, O’Connor G, Artola Zavala T, Dikici E, Zingg JM, Deo S, Daunert S. Engineered biosensors for the quorum sensing molecule 3,5-dimethyl-pyrazine-2-ol (DPO) reveal its presence in humans, animals, and bacterial species beyond Vibrio cholerae. Biosens Bioelectron. 2023;237: 115494. https://doi.org/10.1016/j.bios.2023.115494.

    Article  Google Scholar 

  46. Gryta A, Frąc M. Methodological aspects of multiplex terminal restriction fragment length polymorphism-technique to describe the genetic diversity of soil bacteria, archaea and fungi. Sensors. 2020;20:3292. https://doi.org/10.3390/s20113292.

    Article  Google Scholar 

  47. Jurburg SD, Shek KL, McGuire K. Soil microbial composition varies in response to coffee agroecosystem management. FEMS Microbiol Ecol. 2020;96:fiaa164. https://doi.org/10.1093/femsec/fiaa164.

  48. Zhang Y, Yang S, Zeng Y, Chen Y, Liu H, Yan X, Pu S. A new quantitative insight: Interaction of polyethylene microplastics with soil - microbiome - crop. J Hazard Mater. 2023;460: 132302. https://doi.org/10.1016/j.jhazmat.2023.132302.

    Article  Google Scholar 

  49. Toledo S, Bondaruk VF, Yahdjian L, et al. Environmental factors regulate soil microbial attributes and their response to drought in rangeland ecosystems. Sci Total Environ. 2023;892: 164406. https://doi.org/10.1016/j.scitotenv.2023.164406.

    Article  Google Scholar 

  50. Finn DR, Schroeder J, Samad MS, Poeplau C, Tebbe CC. Importance of sample pre-treatments for the DNA-based characterization of microbiomes in cropland and forest soils. Soil Biol Biochem. 2023;184: 109077. https://doi.org/10.1016/j.soilbio.2023.109077.

    Article  Google Scholar 

  51. Fricker AM, Podlesny D, Fricke WF. What is new and relevant for sequencing-based microbiome research? A mini-review J Adv Res. 2019;19:105–12. https://doi.org/10.1016/j.jare.2019.03.006.

    Article  Google Scholar 

  52. Kui L, Xiang G, Wang Y, et al. Large-scale characterization of the soil microbiome in ancient tea plantations using high-throughput 16S rRNA and internal transcribed spacer amplicon sequencing. Front Microbiol. 2021;12: 745225. https://doi.org/10.3389/fmicb.2021.745225.

    Article  Google Scholar 

  53. Tuttle MS, Mostow E, Mukherjee P, Hu FZ, Melton-Kreft R, Ehrlich GD, Dowd SE, Ghannoum MA. Characterization of bacterial communities in venous insufficiency wounds by use of conventional culture and molecular diagnostic methods. J Clin Microbiol. 2011;49:3812–9. https://doi.org/10.1128/jcm.00847-11.

    Article  Google Scholar 

  54. Puig S, Ganigué R, Batlle-Vilanova P, Balaguer MD, Bañeras L, Colprim J. Tracking bio-hydrogen-mediated production of commodity chemicals from carbon dioxide and renewable electricity. Bioresour Technol. 2017;228:201–9. https://doi.org/10.1016/j.biortech.2016.12.035.

    Article  Google Scholar 

  55. Mącik M, Gryta A, Sas-Paszt L, Frąc M. The status of soil microbiome as affected by the application of phosphorus biofertilizer: Fertilizer enriched with beneficial bacterial strains. Int J Mol Sci. 2020;21:8003. https://doi.org/10.3390/ijms2121800.

    Article  Google Scholar 

  56. Vieira AR, Hiller NL, Powell E, Kim LHJ, Spirk T, Modesto A, Kreft R. Profiling microorganisms in whole saliva of children with and without dental caries. Clin Exp Dent Res. 2019;5:438–46. https://doi.org/10.1002/cre2.206.

    Article  Google Scholar 

  57. Meredith LK, Ogée J, Boye K, et al. Soil exchange rates of COS and CO18O differ with the diversity of microbial communities and their carbonic anhydrase enzymes. ISME J. 2019;13:290–300. https://doi.org/10.1038/s41396-018-0270-2.

    Article  Google Scholar 

  58. Benucci GMN, Bonito V, Bonito G. Fungal, bacterial, and archaeal diversity in soils beneath native and introduced plants in Fiji. South Pacific Microb Ecol. 2019;78:136–46. https://doi.org/10.1007/s00248-018-1266-1.

    Article  Google Scholar 

  59. Naylor D, Fansler S, Brislawn C, Nelson WC, Hofmockel KS, Jansson JK, Mcclure R. Deconstructing the soil microbiome into reduced-complexity functional modules. mBio. 2020;11:e01349–20. https://doi.org/10.1128/mBio.01349-20.

  60. Pang Z, Xu P, Yu D. Environmental adaptation of the root microbiome in two rice ecotypes. Microbiol Res. 2020;241: 126588. https://doi.org/10.1016/j.micres.2020.126588.

    Article  Google Scholar 

  61. Bao Y, Qi B, Huang W, Liu B, Li Y. The fungal community in non-rhizosphere soil of Panax ginseng are driven by different cultivation modes and increased cultivation periods. PeerJ. 2020;8: e9930. https://doi.org/10.7717/peerj.9930.

    Article  Google Scholar 

  62. Freitas L, Appolinario L, Calegario G, et al. Glacial-interglacial transitions in microbiomes recorded in deep-sea sediments from the western equatorial Atlantic. Sci Total Environ. 2020;746: 140904. https://doi.org/10.1016/j.scitotenv.2020.140904.

    Article  Google Scholar 

  63. Neira P, Henríquez-Castillo C, Ortiz J, Stolpe N, Dube F. Do different densities of tree cover affect pasture biomass and soil microbial communities? Agrofor Syst. 2021;95:1465–78. https://doi.org/10.1007/s10457-021-00653-3.

    Article  Google Scholar 

  64. Ruiz ON, Brown LM, Radwan O, Bowen LL, Gunasekera TS, Mueller SS, West ZJ, Striebich RC. Metagenomic characterization reveals complex association of soil hydrocarbon-degrading bacteria. Int Biodeterior Biodegradation. 2021;157: 105161. https://doi.org/10.1016/j.ibiod.2020.105161.

    Article  Google Scholar 

  65. Graziano S, Caldara M, Gullì M, Bevivino A, Maestri E, Marmiroli N. A metagenomic and gene expression analysis in wheat (T. durum) and maize (Z. mays) biofertilized with PGPM and biochar. Int J Mol Sci. 2022;23:10376. https://doi.org/10.3390/ijms231810376.

  66. Yang J, Lee J, Choi J, Ma L, Heaton EA, Howe A. Response of total (DNA) and metabolically active (RNA) microbial communities in miscanthus x giganteus cultivated soil to different nitrogen fertilization rates. Microbiol Spectr. 2022;10:e02116-e2121. https://doi.org/10.1128/spectrum.02116-21.

    Article  Google Scholar 

  67. Li X, Kong P, Daughtrey M, Kosta K, Schirmer S, Howle M, Likins M, Hong C. Characterization of the soil bacterial community from selected boxwood gardens across the United States. Microorganisms. 2022;10:1514. https://doi.org/10.3390/microorganisms1008151414.

    Article  Google Scholar 

  68. Xing H, Zhang Y, Krämer M, Kissmann AK, Henkel M, Weil T, Knippschild U, Rosenau F. A polyclonal selex aptamer library directly allows specific labelling of the human gut bacterium Blautia producta without isolating individual aptamers. Molecules. 2022;27:5693. https://doi.org/10.3390/molecules27175693.

    Article  Google Scholar 

  69. Mishra A, Singh D, Hathi Z, Purohit HJ, Jessy MD, Philip A, Uthup TK, Singh L. Soil microbiome dynamics associated with conversion of tropical forests to different rubber based land use management systems. Appl Soil Ecol. 2023;188: 104933. https://doi.org/10.1016/j.apsoil.2023.104933.

    Article  Google Scholar 

  70. Santorelli LA, Wilkinson T, Abdulmalik R, Rai Y, Creevey CJ, Huws S, Gutierrez-Merino J. Beehives possess their own distinct microbiomes. Environ Microbiomes. 2023;18:1. https://doi.org/10.1186/s40793-023-00460-6.

    Article  Google Scholar 

  71. Schaerer L, Ghannam R, Olson A, Van Camp A, Techtmann S. Persistence of location-specific microbial signatures on boats during voyages. Mar Pollut Bull. 2024;199: 115884. https://doi.org/10.1016/j.marpolbul.2023.115884.

    Article  Google Scholar 

  72. Thiele-Bruhn S, Schloter M, Wilke BM, Beaudette LA, Martin-Laurent F, Cheviron N, Mougin C, Römbke J. Identification of new microbial functional standards for soil quality assessment. Soil. 2020;6:17–34. https://doi.org/10.5194/soil-6-17-2020.

    Article  Google Scholar 

  73. Legeay J, Hijri M. A comprehensive insight of current and future challenges in large-scale soil microbiome analyses. Microb Ecol. 2023;86:75–85. https://doi.org/10.1007/s00248-022-02060-2.

    Article  Google Scholar 

  74. Knight R, Vrbanac A, Taylor BC, et al. Best practices for analysing microbiomes. Nat Rev Microbiol. 2018;16:410–22. https://doi.org/10.1038/s41579-018-0029-9.

    Article  Google Scholar 

  75. Manetsberger J, Caballero Gómez N, Benomar N, Christie G, Abriouel H. Characterization of the culturable sporobiota of spanish olive groves and its tolerance toward environmental challenges. Microbiol Spectr. 2023;11:e04013-e4022. https://doi.org/10.1128/spectrum.04013-22.

    Article  Google Scholar 

  76. Kapinusova G, Lopez Marin MA, Uhlik O. Reaching unreachables: Obstacles and successes of microbial cultivation and their reasons. Front Microbiol. 2023;14:1089630. https://doi.org/10.3389/fmicb.2023.1089630.

    Article  Google Scholar 

  77. Renella G, Ogunseitan O, Giagnoni L, Arenella M. Environmental proteomics: A long march in the pedosphere. Soil Biol Biochem. 2014;69:34–7. https://doi.org/10.1016/j.soilbio.2013.10.035.

    Article  Google Scholar 

  78. Nickel JC, Stephens A, Landis JR, Chen J, Mullins C, Van Bokhoven A, Lucia MS, Melton-Kreft R, Ehrlich GD. Search for microorganisms in men with urologic chronic pelvic pain syndrome: A culture-independent analysis in the MAPP research network. J Urol. 2015;194:127–35. https://doi.org/10.1016/j.juro.2015.01.037.

    Article  Google Scholar 

  79. Kaeberlein T, Lewis K, Epstein SS. Isolating “Uncultivable” Microorganisms in Pure culture in a simulated natural environment. Science. 2002;296:1127–9. https://doi.org/10.1126/science.1070633.

    Article  Google Scholar 

  80. Bollmann A, Lewis K, Epstein SS. Incubation of environmental samples in a diffusion chamber increases the diversity of recovered isolates. Appl Environ Microbiol. 2007;73:6386–90. https://doi.org/10.1128/AEM.01309-07.

    Article  Google Scholar 

  81. Hu T, Chitnis N, Monos D, Dinh A. Next-generation sequencing technologies: An overview. Hum Immunol. 2021;82:801–11. https://doi.org/10.1016/j.humimm.2021.02.012.

    Article  Google Scholar 

  82. Heather JM, Chain B. The sequence of sequencers: The history of sequencing DNA. Genomics. 2016;107:1–8. https://doi.org/10.1016/j.ygeno.2015.11.003.

    Article  Google Scholar 

  83. Liu H, Wu S, Li A, Ruan J. SMARTdenovo: a de novo assembler using long nosiy reads. GigaByte. 2021;2021:1–9. https://doi.org/10.46471/gigabyte.15.

  84. Lischer HEL, Shimizu KK. Reference-guided de novo assembly approach improves genome reconstruction for related species. BMC Bioinformatics. 2017;18:474. https://doi.org/10.1186/s12859-017-1911-6.

    Article  Google Scholar 

  85. Lee JY, Mitchell HD, Burnet MC, et al. Uncovering Hidden Members and Functions of the Soil Microbiome Using de Novo Metaproteomics. J Proteome Res. 2022;21:2023–35. https://doi.org/10.1021/acs.jproteome.2c00334.

    Article  Google Scholar 

  86. Burstein HJ, Demichele A, Somerfield MR, Henry NL. Testing for ESR1 Mutations to guide therapy for hormone receptor-positive, human epidermal growth factor receptor 2-negative metastatic breast cancer: ASCO Guideline Rapid Recommendation Update. J Clin Oncol. 2023;41:3423–5. https://doi.org/10.1200/JCO.23.00638.

    Article  Google Scholar 

  87. Pujol P, Barberis M, Beer P, et al. Clinical practice guidelines for BRCA1 and BRCA2 genetic testing. Eur J Cancer. 2021;146:30–47. https://doi.org/10.1016/j.ejca.2020.12.023.

    Article  Google Scholar 

  88. Kaster A-K, Sobol MS. Microbial single-cell omics: the crux of the matter. Appl Microbiol Biotechnol. 2020;104:8209–20. https://doi.org/10.1007/s00253-020-10844-0.

    Article  Google Scholar 

  89. Dey SS, Kester L, Spanjaard B, Bienko M, Van Oudenaarden A. Integrated genome and transcriptome sequencing of the same cell. Nat Biotechnol. 2015;33:285–9. https://doi.org/10.1038/nbt.3129.

    Article  Google Scholar 

  90. Su T, Hollas MAR, Fellers RT, Kelleher NL. Identification of Splice Variants and Isoforms in Transcriptomics and Proteomics. Annu Rev Biomed Data Sci. 2023;6:357–76. https://doi.org/10.1146/annurev-biodatasci-020722-044021.

    Article  Google Scholar 

  91. Wesener DA, Beller ZW, Peters SL, Rajabi A, Dimartino G, Giannone RJ, Hettich RL, Gordon JI. Microbiota functional activity biosensors for characterizing nutrient metabolism in vivo. eLife. 2021;10:e64478. https://doi.org/10.7554/eLife.64478.

  92. Sun B, Liu Z, Liu J, Zhao S, Wang L, Wang F. The utility of proteases in proteomics, from sequence profiling to structure and function analysis. Proteomics. 2023;23:2200132. https://doi.org/10.1002/pmic.202200132.

    Article  Google Scholar 

  93. Clark TN, Houriet J, Vidar WS, Kellogg JJ, Todd DA, Cech NB, Linington RG. Interlaboratory comparison of untargeted mass spectrometry data uncovers underlying causes for variability. J Nat Prod. 2021;84:824–35. https://doi.org/10.1021/acs.jnatprod.0c01376.

    Article  Google Scholar 

  94. Satam H, Joshi K, Mangrolia U, et al. Next-generation sequencing technology: Current trends and advancements. Biology. 2023;12:997. https://doi.org/10.3390/biology12070997.

    Article  Google Scholar 

  95. Delventhal K, Skillman V, Li X, Busby PE, Frost K. Characterizing variation in the bacterial and fungal tare soil microbiome of seed potato. Phytobiomes J. 2023;7:78–90. https://doi.org/10.1094/PBIOMES-11-22-0092-R.

    Article  Google Scholar 

  96. Osman JR, Fernandes GR, Kamilova E, DuBow MS. Genomic microbiome analyses of surface sand samples from the Kyzyl-Kum Desert (Uzbekistan): characterization and comparative study. Arch Microbiol. 2023;205:90. https://doi.org/10.1007/s00203-023-03432-z.

    Article  Google Scholar 

  97. Roy S, Coldren C, Karunamurthy A, et al. Standards and Guidelines for Validating Next-Generation Sequencing Bioinformatics Pipelines: A Joint Recommendation of the Association for Molecular Pathology and the College of American Pathologists. J Mol Diagn. 2018;20:4–27. https://doi.org/10.1016/j.jmoldx.2017.11.003.

    Article  Google Scholar 

  98. Svartström O, Alneberg J, Terrapon N, et al. Ninety-nine de novo assembled genomes from the moose (Alces alces) rumen microbiome provide new insights into microbial plant biomass degradation. ISME J. 2017;11:2538–51. https://doi.org/10.1038/ismej.2017.108.

    Article  Google Scholar 

  99. Zhang X, Li L, Butcher J, Stintzi A, Figeys D. Advancing functional and translational microbiome research using meta-omics approaches. Microbiome. 2019;7:154. https://doi.org/10.1186/s40168-019-0767-6.

    Article  Google Scholar 

  100. Bashiardes S, Zilberman-Schapira G, Elinav E. Use of metatranscriptomics in microbiome research. Bioinform Biol Insights. 2016;10:19–25. https://doi.org/10.4137/BBI.S34610.

    Article  Google Scholar 

  101. Shakya M, Lo CC, Chain PSG. Advances and challenges in metatranscriptomic analysis. Front Genet. 2019;10:904. https://doi.org/10.3389/fgene.2019.00904.

    Article  Google Scholar 

  102. Wang Y, Zhou Y, Xiao X, Zheng J, Zhou H. Metaproteomics: A strategy to study the taxonomy and functionality of the gut microbiota. J Proteomics. 2020;219: 103737. https://doi.org/10.1016/j.jprot.2020.103737.

    Article  Google Scholar 

  103. Utpott M, Rodrigues E, Rios A de O, Mercali GD, Flôres SH. Metabolomics: An analytical technique for food processing evaluation. Food Chem. 2022;366:130685. https://doi.org/10.1016/j.foodchem.2021.130685.

  104. Mandakovic D, Maldonado J, Pulgar R, Cabrera P, Gaete A, Urtuvia V, Seeger M, Cambiazo V, González M. Microbiome analysis and bacterial isolation from Lejía Lake soil in Atacama Desert. Extremophiles. 2018;22:665–73. https://doi.org/10.1007/s00792-018-1027-6.

    Article  Google Scholar 

  105. Cordier T, Lanzén A, Apothéloz-Perret-Gentil L, Stoeck T, Pawlowski J. Embracing environmental genomics and machine learning for routine biomonitoring. Trends Microbiol. 2019;27:387–97. https://doi.org/10.1016/j.tim.2018.10.012.

    Article  Google Scholar 

  106. Jiang X, Xu W, Park EK, Li G. Selecting protein families for environmental features based on manifold regularization. IEEE Trans Nanobiosci. 2014;13:104–8. https://doi.org/10.1109/TNB.2014.2316744.

    Article  Google Scholar 

  107. McElhinney JMWR, Catacutan MK, Mawart A, Hasan A, Dias J. Interfacing machine learning and microbial omics: a promising means to address environmental challenges. Front Microbiol. 2022;13: 851450. https://doi.org/10.3389/fmicb.2022.851450.

    Article  Google Scholar 

  108. Jansson JK, McClure R, Egbert RG. Soil microbiome engineering for sustainability in a changing environment. Nat Biotechnol. 2023;41:1716–28. https://doi.org/10.1038/s41587-023-01932-3.

    Article  Google Scholar 

  109. Fittipaldi M, Nocker A, Codony F. Progress in understanding preferential detection of live cells using viability dyes in combination with DNA amplification. J Microbiol Meth. 2012;91:276–89. https://doi.org/10.1016/j.mimet.2012.08.007.

    Article  Google Scholar 

  110. Nocker A, Cheung CY, Camper AK. Comparison of propidium monoazide with ethidium monoazide for differentiation of live vs. dead bacteria by selective removal of DNA from dead cells. J Microbiol Meth. 2006;67:310–320. https://doi.org/10.1016/j.mimet.2006.04.015.

  111. Wallace A. Absolute Quantification of Microbiota in Shotgun Sequencing Using Host Cells or Spike-Ins. bioRxiv 2023.08.23.554046. https://doi.org/10.1101/2023.08.23.554046.

  112. Perron GG, Whyte L, Turnbaugh PJ, Goordial J, Hanage WP, Dantas G, Desai MM. Functional characterization of bacteria isolated from ancient arctic soil exposes diverse resistance mechanisms to modern antibiotics. PLoS ONE. 2015;10: e0069533. https://doi.org/10.1371/journal.pone.0069533.

    Article  Google Scholar 

  113. Nagaishi T, Watabe T, Kotake K, et al. Immunoglobulin A-specific deficiency induces spontaneous inflammation specifically in the ileum. Gut. 2022;71:487–96. https://doi.org/10.1136/gutjnl-2020-322873.

    Article  Google Scholar 

  114. Talley S, Bonomo R, Gavini C, et al. Monitoring of inflammation using novel biosensor mouse model reveals tissue- and sex-specific responses to Western diet. Dis Model Mech. 2022;15:dmm049313. https://doi.org/10.1242/DMM.049313.

  115. Wang M, Yang Y, Min J, et al. A wearable electrochemical biosensor for the monitoring of metabolites and nutrients. Nat Biomed Eng. 2022;6:1225–35. https://doi.org/10.1038/s41551-022-00916-z.

    Article  Google Scholar 

  116. Zhang Y, Hu Y, Jiang N, Yetisen AK. Wearable artificial intelligence biosensor networks. Biosens Bioelectron. 2023;219: 114825. https://doi.org/10.1016/j.bios.2022.114825.

    Article  Google Scholar 

  117. O’Farrell B. Evolution in lateral flow–based immunoassay systems. In: Lateral Flow Immunoassay. Humana Press, 2009; pp 1–33. https://doi.org/10.1007/978-1-59745-240-3_1.

  118. Yoon J-Y. Introduction to biosensors: from electric circuits to immunosensors, Second Edition. Springer, 2016; Chapter 13. https://doi.org/10.1007/978-3-319-27413-3_13.

  119. Markus V, Share O, Shagan M, et al. Inhibitory effects of artificial sweeteners on bacterial quorum sensing. Int J Mol Sci. 2021;22:9863. https://doi.org/10.3390/ijms22189863.

    Article  Google Scholar 

  120. Sazykin I, Naumova E, Azhogina T, Klimova M, Karchava S, Khmelevtsova L, Chernyshenko E, Litsevich A, Khammami M, Sazykina M. Glyphosate effect on biofilms formation, mutagenesis and stress response of E. coli. J Hazard Mater. 2024;461:132574. https://doi.org/10.1016/j.jhazmat.2023.132574.

  121. O’Connor G, Quintero MA, Deo SK, Abreu MT, Daunert S. Bacterial quorum-sensing molecules in serum: A potential tool for Crohn’s disease management. Clin Transl Gastroenterol. 2022;13:e00547. https://doi.org/10.14309/ctg.0000000000000547.

  122. Nuñez S, Barra M, Garrido D. Developing a fluorescent inducible system for free fucose quantification in Escherichia coli. Biosensors. 2023;13:388. https://doi.org/10.3390/bios13030388.

    Article  Google Scholar 

  123. Serebrinsky-Duek K, Barra M, Danino T, Garrido D. Engineered bacteria for short-chain-fatty-acid-repressed expression of biotherapeutic molecules. Microbiol Spectr. 2023;11:e00049-e123. https://doi.org/10.1128/spectrum.00049-23.

    Article  Google Scholar 

  124. Guan C, Larson PJ, Fleming E, Tikhonov AP, Mootien S, Grossman TH, Golino C, Oh J. Engineering a “detect and destroy” skin probiotic to combat methicillin-resistant Staphylococcus aureus. PLoS ONE. 2022;17: e0276795. https://doi.org/10.1371/journal.pone.0276795.

    Article  Google Scholar 

  125. Purohit B, Vernekar PR, Shetti NP, Chandra P. Biosensor nanoengineering: Design, operation, and implementation for biomolecular analysis. Sens Int. 2020;1: 100040. https://doi.org/10.1016/j.sintl.2020.10004040.

    Article  Google Scholar 

  126. Kour R, Arya S, Young S-J, Gupta V, Bandhoria P, Khosla A. Review—Recent advances in carbon nanomaterials as electrochemical biosensors. J Electrochem Soc. 2020;167: 037555. https://doi.org/10.1149/1945-7111/ab6bc4.

    Article  Google Scholar 

  127. Ostadhossein F, Moitra P, Alafeef M, et al. Ensemble and single-particle level fluorescent fine-tuning of carbon dots via positional changes of amines toward “supervised” oral microbiome sensing. J Biomed Opt. 2023;28: 082807. https://doi.org/10.1117/1.jbo.28.8.082807.

    Article  Google Scholar 

  128. Melnikov PV, Alexandrovskaya AY, Naumova AO, Popova NM, Spitsyn B V., Zaitsev NK, Yashtulov NA. Modified nanodiamonds as a means of polymer surface functionalization. From fouling suppression to biosensor design. Nanomaterials. 2021;11:2980. https://doi.org/10.3390/nano11112980.

  129. Singh A, Sharma A, Ahmed A, Sundramoorthy AK, Furukawa H, Arya S, Khosla A. Recent advances in electrochemical biosensors: Applications, challenges, and future scope. Biosensors. 2021;11:336. https://doi.org/10.3390/bios11090336.

    Article  Google Scholar 

  130. Costanzo H, Gooch J, Frascione N. Nanomaterials for optical biosensors in forensic analysis. Talanta. 2023;253: 123945. https://doi.org/10.1016/j.talanta.2022.123945.

    Article  Google Scholar 

  131. Teng M, Hao M, Ding C, Wang L, Shen H, Yu S, Chen L, Yang F. Rapid detection of Saccharomyces cerevisiae with boronic acid-decorated multivariate metal-organic frameworks and aptamers. Analyst. 2023;148:4213–8. https://doi.org/10.1039/D3AN00835E.

    Article  Google Scholar 

  132. Pellegrino GM, Browne TS, Sharath K, Bari KA, Vancuren SJ, Allen-Vercoe E, Gloor GB, Edgell DR. Metabolically-targeted dCas9 expression in bacteria. Nucleic Acids Res. 2023;51:982–96. https://doi.org/10.1093/nar/gkac1248.

    Article  Google Scholar 

  133. Enespa CP. Tool and techniques study to plant microbiome current understanding and future needs: An overview. Commun Integr Biol. 2022;15:209–25. https://doi.org/10.1080/19420889.2022.2082736.

    Article  Google Scholar 

  134. Li G, Yuan B, Zhao L, Gao W, Xu C, Liu G. Fouling-resistant electrode for electrochemical sensing based on covalent-organic frameworks TpPA-1 dispersed cabon nanotubes. Talanta. 2024;267: 125162. https://doi.org/10.1016/j.talanta.2023.125162.

    Article  Google Scholar 

  135. Jarosińska E, Zambrowska Z, Witkowska NE. Methods of protection of electrochemical sensors against biofouling in cell culture applications. ACS Omega. 2024;9:4572–80. https://doi.org/10.1021/acsomega.3c07660.

    Article  Google Scholar 

  136. Cesewski E, Johnson BN. Electrochemical biosensors for pathogen detection. Biosens Bioelectron. 2020;159: 112214. https://doi.org/10.1016/j.bios.2020.112214.

    Article  Google Scholar 

  137. McCracken KE, Angus SV, Reynolds KA, Yoon JY. Multimodal imaging and lighting bias correction for improved μPAD-based water quality monitoring via smartphones. Sci Rep. 2016;6:27529. https://doi.org/10.1038/srep27529.

    Article  Google Scholar 

  138. Ulep TH, Zenhausern R, Gonzales A, Knoff DS, Lengerke Diaz PA, Castro JE, Yoon JY. Smartphone based on-chip fluorescence imaging and capillary flow velocity measurement for detecting ROR1+ cancer cells from buffy coat blood samples on dual-layer paper microfluidic chip. Biosens Bioelectron. 2020;153: 112042. https://doi.org/10.1016/j.bios.2020.112042.

    Article  Google Scholar 

  139. Breshears LE, Nguyen BT, Akarapipad P, Sosnowski K, Kaarj K, Quirk G, Uhrlaub JL, Nikolich-Žugich J, Worobey M, Yoon JY. Sensitive, smartphone-based SARS-CoV-2 detection from clinical saline gargle samples. PNAS Nexus. 2022;1:pgac028. https://doi.org/10.1093/pnasnexus/pgac028.

  140. Buchanan BC, Safavinia B, Wu L, Yoon JY. Smartphone-based autofluorescence imaging to detect bacterial species on laboratory surfaces. Analyst. 2022;147:2980–7. https://doi.org/10.1039/D2AN00358A.

    Article  Google Scholar 

  141. Kim S, Lee MH, Wiwasuku T, Day AS, Youngme S, Hwang DS, Yoon JY. Human sensor-inspired supervised machine learning of smartphone-based paper microfluidic analysis for bacterial species classification. Biosens Bioelectron. 2021;188: 113335. https://doi.org/10.1016/j.bios.2021.113335.

    Article  Google Scholar 

  142. Breshears LE, Mata-Robles S, Tang Y, Baker JC, Reynolds KA, Yoon JY. Rapid, sensitive detection of PFOA with smartphone-based flow rate analysis utilizing competitive molecular interactions during capillary action. J Hazard Mater. 2023;446: 130699. https://doi.org/10.1016/j.jhazmat.2022.130699.

    Article  Google Scholar 

  143. Liang Y, Lee MH, Zhou A, Khanthaphixay B, Hwang DS, Yoon JY. eXtreme gradient boosting-based classification of bacterial mixtures in water and milk using wireless microscopic imaging of quorum sensing peptide-conjugated particles. Biosens Bioelectron. 2023;227: 115144. https://doi.org/10.1016/j.bios.2023.115144.

    Article  Google Scholar 

  144. Kim S, Sosnowski K, Hwang DS, Yoon JY. Smartphone-based microalgae monitoring platform using machine learning. ACS ES&T Eng. 2023;4:186–95. https://doi.org/10.1021/acsestengg.3c0026.

    Article  Google Scholar 

  145. Wilhelm RC, van Es HM, Buckley DH. Predicting measures of soil health using the microbiome and supervised machine learning. Soil Biol Biochem. 2022;164: 108472. https://doi.org/10.1016/j.soilbio.2021.108472.

    Article  Google Scholar 

  146. Chung S, Loh A, Jennings CM, Sosnowski K, Ha SY, Yim UH, Yoon JY. Capillary flow velocity profile analysis on paper-based microfluidic chips for screening oil types using machine learning. J Hazard Mater. 2023;447: 130806. https://doi.org/10.1016/j.jhazmat.2023.130806.

    Article  Google Scholar 

  147. Toma K, Iwasaki K, Zhang G, Iitani K, Arakawa T, Iwasaki Y, Mitsubayashi K. Biochemical methanol gas sensor (MeOH bio-sniffer) for non-invasive assessment of intestinal flora from breath methanol. Sensors. 2021;21:4897. https://doi.org/10.3390/s21144897.

    Article  Google Scholar 

Download references

Funding

This work was supported by the University of Arizona’s One Health Initiative Pilot Grant and the multi-state research grant (NC-1194): Nanotechnology and Biosensors from the National Institute of Food and Agriculture (NIFA), United States Department of Agriculture (USDA).

Author information

Authors and Affiliations

Authors

Contributions

Lexi DeFord: Conceptualization, Methodology, Validation, Formal analysis, Investigation, Resources, Data curation, Writing – original draft, Visualization. Jeong-Yeol Yoon: Conceptualization, Methodology, Writing – review & editing, Visualization, Supervision, Project administration, Funding acquisition.

Corresponding author

Correspondence to Jeong-Yeol Yoon.

Ethics declarations

Ethics approval and consent to participate

Not applicable.

Consent for publication

Not applicable.

Competing interests

The authors declare no competing interests.

Additional information

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution-NonCommercial-NoDerivatives 4.0 International License, which permits any non-commercial use, sharing, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if you modified the licensed material. You do not have permission under this licence to share adapted material derived from this article or parts of it. The images or other third party material in this article are included in the article’s Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article’s Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by-nc-nd/4.0/.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

DeFord, L., Yoon, JY. Soil microbiome characterization and its future directions with biosensing. J Biol Eng 18, 50 (2024). https://doi.org/10.1186/s13036-024-00444-1

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s13036-024-00444-1

Keywords