Next Article in Journal
Hydrological Modeling of Karst Watershed Containing Subterranean River Using a Modified SWAT Model: A Case Study of the Daotian River Basin, Southwest China
Next Article in Special Issue
Monitoring of SARS-CoV-2 Variants by Wastewater-Based Surveillance as a Sustainable and Pragmatic Approach—A Case Study of Jaipur (India)
Previous Article in Journal
Historical Water Supply System of the City of Brno—Social-Environmental Consequences
Previous Article in Special Issue
Imprints of Lockdown and Treatment Processes on the Wastewater Surveillance of SARS-CoV-2: A Curious Case of Fourteen Plants in Northern India
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Molecular Methods for Pathogenic Bacteria Detection and Recent Advances in Wastewater Analysis

1
School of Civil, Mining and Environmental Engineering, University of Wollongong, Wollongong 2522, Australia
2
Department of Microbiology and Immunology at the University of Melbourne, Melbourne 3010, Australia
3
Queensland Alliance for Environmental Health Sciences (QAEHS), University of Queensland, Woolloongabba 4102, Australia
4
Illawarra Health and Medical Research Institute (IHMRI), University of Wollongong, Wollongong 2522, Australia
*
Author to whom correspondence should be addressed.
Water 2021, 13(24), 3551; https://doi.org/10.3390/w13243551
Submission received: 15 November 2021 / Revised: 2 December 2021 / Accepted: 9 December 2021 / Published: 12 December 2021
(This article belongs to the Special Issue Pathogen Detection and Identification in Wastewater)

Abstract

:
With increasing concerns about public health and the development of molecular techniques, new detection tools and the combination of existing approaches have increased the abilities of pathogenic bacteria monitoring by exploring new biomarkers, increasing the sensitivity and accuracy of detection, quantification, and analyzing various genes such as functional genes and antimicrobial resistance genes (ARG). Molecular methods are gradually emerging as the most popular detection approach for pathogens, in addition to the conventional culture-based plate enumeration methods. The analysis of pathogens in wastewater and the back-estimation of infections in the community, also known as wastewater-based epidemiology (WBE), is an emerging methodology and has a great potential to supplement current surveillance systems for the monitoring of infectious diseases and the early warning of outbreaks. However, as a complex matrix, wastewater largely challenges the analytical performance of molecular methods. This review synthesized the literature of typical pathogenic bacteria in wastewater, types of biomarkers, molecular methods for bacterial analysis, and their recent advances in wastewater analysis. The advantages and limitation of these molecular methods were evaluated, and their prospects in WBE were discussed to provide insight for future development.

1. Introduction

Diseases induced by human pathogens are a major threat to public health worldwide. According to an estimation by the World Health Organization (WHO), 600 million people (almost 1 in 10 people) fall ill from eating contaminated food, and among them 420,000 people die every year, leading to the loss of 33 million healthy lives [1]. Unsafe food causes an annual loss of USD 110 billion in productivity and medical expenses in low- and middle-income countries. In addition, the ever-increasing waterborne diseases become a global burden, causing a financial loss of almost USD 12 billion and over 2.2 million deaths each year. Water and foodborne diseases overburden health care systems and affect global economies, thus hindering socio-economic development.
Among various waterborne and foodborne pathogens, pathogenic bacteria are the largest and most common group. Food and waterborne pathogenic bacteria, including Escherichia coli (E. coli), Campylobacter spp., (C. jejuni and C. coli), Legionella spp., Salmonella spp., and Shigella spp. are responsible for most infection cases, sometimes with severe and fatal outcomes. In recent years, although water and foodborne disease outbreaks have been declining with the increasing efforts in improving public health, the burden of infectious water and foodborne diseases is still a pressing global issue [2,3]. Furthermore, the growing antimicrobial resistance (AMR) of pathogens threatens the effective prevention and treatment of an ever-increasing range of infections, reminding us of the urgency and importance of strengthening our capacity to monitor and prevent the increasing risk of these human pathogens.
The traditional detection method of pathogenic bacteria mainly relies on microbial culturing. Culture-based methods are low-cost, easy to operate, and highly standardized and thus are broadly used for the regulatory purposes of pathogenic bacteria monitoring such as enumerating FIB in bathing water [4]. However, the main limitations of these methods are the lack of differentiation between the target and other non-target endogenous microorganisms of the same samples, false negative/positive results, time and labor-consuming procedures, and the inability to detect viable but nonculturable (VBNC) cells [5]. Moreover, in the application of quantitative research, culture-based methods often underestimate the number of bacteria. This affects the quantification accuracy of targets and underestimates the prevalence of pathogens in the human community. In addition, VBNC can become viable and may cause disease and increase the public health risk [4]. Therefore, as a rapid analyzing tool with high accuracy and specificity, molecular methods have quickly become the mainstream detection technique of pathogenic bacteria.
According to the biological markers being used, molecular methods can be divided in to two major groups, i.e., the nucleic acid targeting method and protein/antigen targeting method [6]. The nucleic acid targeting method includes fluorescence amplification-based methods, such as the polymerase chain reaction (PCR), quantitative or real-time PCR (qPCR), digital PCR (dPCR), deoxyribonucleic acid (DNA) microarray, fluorescence in situ hybridization (FISH), and molecular beacon, and sequencing-based methods such as pyrosequencing, Illumina sequencing, and nanopore sequencing. The protein and antigen targeting method includes a traditional antibody–antigen interaction method similar to immunological methods (lateral flow tests (LFTs)) and enzyme-linked immunosorbent assays (ELISA) [7]. Moreover, by combining the basic molecular detection approaches with a metal and paper platform, biosensor-based and paper-based devices have become a rapid, cheap, and portable on-site method for pathogenic bacteria detection [8,9,10].
Wastewater-based epidemiology (WBE) is a method to obtain qualitative and quantitative data on the chemical use/exposure and infectious cases of residents within a given wastewater catchment area based on the analysis of chemical compounds, pathogens, and certain biomarkers in raw sewage [11,12]. Sewage collected from a wastewater treatment plant (WWTP) or sewers can be regarded as the pooled urine and stool sample within a community and can be used to evaluate the health status of the whole community [11]. Studies based on wastewater have showed that sewage can reveal not only illicit drug use, diet, and lifestyles, but also disease outbreaks within a community [11,12,13]. In addition, different from clinical testing, wastewater analysis can include pathogens shed by asymptomatic and presymptomatic individuals. This makes it a powerful tool for the early warning and timely intervention of infectious disease [14,15,16]. Wastewater can be monitored for pathogenic and benign microbes through a variety of technologies. Traditional techniques usually include filtering, staining, and examining samples under a microscope. More sensitive and specific methods such as PCR-based methods and DNA sequencing have also been employed in the analysis of human pathogens in wastewater. However, the concentration of pathogenic bacteria is usually lower than indicator microorganisms and thus requires a highly sensitive detection method. The complex wastewater matrix often causes false-negative results because of the presence of various inhibitors. In addition, sample processing methods vary for different downstream analytical methodologies. Therefore, it is still challenging to utilize molecular detection methods in the accurate and quantitative detection of pathogenic bacteria in raw wastewater. This review focuses on the molecular analytical techniques for pathogenic bacteria in wastewater and summarizes recent advances of these approaches in wastewater-based epidemiology. The prospects of these approaches in wastewater analysis are discussed to provide insights for further application.

2. Pathogenic Bacteria in Wastewater

Human pathogens, causing serious infections and even death, are one of the leading threats to global public health. Various human pathogens can be grouped as bacteria (e.g., enterohemorrhagic E. coli (EHEC), Campylobacter spp., and Salmonella spp., etc.), viruses (e.g., influenza viruses, hepatitis virus, rotaviruses, and Norwalk viruses, etc.), protozoa (e.g., Giardia lamblia and Cryptosporidium parvum, etc.), and parasites (e.g., ascaris, Ancylostoma, etc.) [17]. Currently, there are approximately 538 species of pathogenic bacteria infecting human beings. This number is much higher than the overall number of other pathogens such as viruses (around 208 types), parasitic protozoa (around 57 species), and some fungi and helminths [17]. The bacteria listed in Table 1 are bacterial human pathogens detected in raw wastewaters, which are reported as the most common species that can cause global health concern (e.g., gastroenteritis or pneumonia). Most pathogens in wastewater are shed by human patients, although some might originate from other sources such as animals.

2.1. Enteric Pathogenic Bacteria

2.1.1. E. coli O157:H7

The strain of E. coli that causes diarrhea is called diarrheagenic E. coli. Among several pathotypes of diarrheagenic E. coli, enterohemorrhagic E. coli (EHEC) is different because of its ability to produce Shiga toxin. Both of E. coli O157:H7 and other non-O157 STEC (Shiga toxin-producing E. coli) such as O26:H11, O111:H8, and O118:H16 can release Shiga toxins, but only O157:H7 genotypes can induce disease in humans. Others commonly reside in cattle without causing diseases [28]. E. coli O157:H7 is the predominant and most virulent serotype in the pathogenic subgroup of E. coli. It could cause not only diarrhea and hemorrhagic colitis, but also hemolytic uremic syndrome, which is a serious long-term complication, mainly affecting children, that leads to kidney failure and death. Virulence factors of E. coli O157:H7 include the type III secretion system, Shiga-like toxin 1 and 2, acid tolerance response system, hemolysin, and extracellular serine protease [29,30]. E. coli is widely used as the fecal contamination indicator, and the O157:H7 genotype is often employed as a model for pathogenic bacteria study in wastewater [18]. The density of E. coli O157:H7 in raw wastewater was found as 10–106 CFU/100 mL [18].

2.1.2. Campylobacter spp.

Campylobacter spp. is one of four major causes of diarrhea, and it is also regarded as the most common cause of human gastroenteritis worldwide. There are 13 pathogenic Campylobacter species known to be associated with human infections such as C. jejuni, C. coli, C. lari, C. concisus, C. rectus, C. hyointestinalis, C. insulaenigrae, C. sputorum, C. helveticus, C. fetus, C. mucosalis, C. upsaliensis, and C. ureolyticus. Among 17 species and six subspecies of Campylobacter, C. jejuni and C. coli are the most related to infections, accounting for 80–85% and 10–15% of total infections, respectively [31]. C. jejuni and C. coli are also the main species widely detected and isolated from wastewater [19,32,33]. Pathogenic Campylobacter is responsible for 400–500 million infections annually [34]. In Europe, nearly 230 thousand cases have been reported every year since 2015 [35]. Presumably, the infection dose of campylobacteriosis is very low, with 360 colony-forming units (CFU) being adequate to cause the illness. Campylobacter spp. possess different virulence factors (VFs) related to motility, adhesion, invasion, toxin-activity, immune evasion, and iron-uptake [36]. VFs, such as the cadF gene and iam locus, are involved in different invasion steps [37,38]. Other VFs, such as the tripartite toxin encoded in the cdtA, cdtB, and cdtC genes [39], block the CDC2 kinase, inducing progressive cellular distension, which causes cell death [36]. Therefore, those genes are widely used in the diagnosis of Campylobacter spp. since thermotolerant Campylobacter spp. is difficult to culture [19,40,41].

2.1.3. Salmonella spp.

Salmonellosis is one of the most important zoonotic diseases caused by Salmonella spp. and transmits to humans through raw food products. A few serotypes, such as S. typhimurium and S. enteritidis, can cause human infection with poultry as the main host. Two foods that are most commonly associated with Salmonella infection are eggs and poultry meat [42]. According to the Centers for Disease Control and Prevention, from 2006 to 2017, Salmonella was responsible for about 53.4% of all foodborne disease outbreaks in the USA, and approximately 32.7% of these outbreaks were related to produce consumption [43]. Additionally, S. Typhi and S. Paratyphi are the main causes of typhoid fever and paratyphoid fever, respectively. Both are human-specific, Gram-negative, and human-restricted bacterial pathogens. Transmission can occur from person to person by eating contaminated food or water or by contact with an acute or chronic infected person [44]. Although a few culturing methods have been developed to isolate and culture those two Salmonella species in various samples, the cultivation is still a difficult task since they are fastidious microorganisms. The molecular approach is considered better for the detection and quantification of Salmonella spp. than the culture-based approach. Many protocols have been developed to target different genes or gene regions specific to Salmonella spp. The most popular gene targets are invA and flagellin genes (fliC-d for Salmonella Typhi, fliC-a for Salmonella Paratyphi A). Some tests are multiplexed to improve sensitivity and specificity [45,46,47,48,49]. Protein markers such as membrane vesicle protein PagC have also been employed in the detection of pathogenic Salmonella as a novel biomarker [50].

2.1.4. Shigella spp.

Shigella is a Gram-negative bacterium, a facultative anaerobe of the Enterobacteriaceae family. It is thought to be responsible for shigellosis or bacillary dysentery [51]. Shigella spp. includes four serotypes: S. dysenteriae, S. flexneri, S. boydii, and S. sonnei. Shigellosis is an invasive disease of the colon that is mainly caused by S. sonnei and S. flexneri. The main mode of transmission is fecal–oral infection, with an infection dose as low as 10 bacterial cells. The continuous transmission in humans must be passed from one person to another, as the bacterium does not survive long outside the body during the plankton-like phase [52]. About 165 million cases of Shigella disease are recorded worldwide each year, resulting in 1 million deaths, particularly in developing countries. It is reported that the phenotypic and genotypic characteristics of Shigella species are too close to be distinguished from diarrheagenic E. coli. This close genetic relationship between Shigella and E. coli leads to the widespread presence of virulence genes, making it difficult or even impossible to distinguish the virulence types of Shigella from E. coli. In recent years, in order to achieve rapid and reliable identification of the four Shigella species, a series of PCR-based methods has been developed by targeting various genes in plasmid DNA, including invasion plasmid antigen H (ipaH) [53], ial [54], virA [55], the she pathogenicity island (spi) [56] and tuf [57].

2.1.5. Clostridioides difficile

Clostridioides difficile (homotypic synonym Clostridium difficile, also known as C. difficile, or C. diff) is a kind of Gram-positive spore-forming bacterium [58]. Pathogenic C. difficile strains can induce diarrhea and life-threatening pseudomembranous colitis, often requiring antibiotic treatment. These strains are mainly identified by their ability to produce the enterotoxin A (TcdA) and/or the cytotoxin B (TcdB) [59]. C. difficile-associated diarrhea (CDAD) is a very common nosocomial infection related with high morbidity and mortality, which imposes a huge financial burden to healthcare facilities [60]. In the last two decades, toxigenic C. difficile became one of the most important causes of hospital infections, with many infections leading to diarrhea and potentially fatal pseudomembranous colitis [61]. It has been recognized that community-acquired C. difficile infections are increasing among people with no apparent contact with healthcare facilities and without any known risk factors for C. difficile infection (CDI) [62]. Since C. difficile is a spore-forming bacterium, it is considered as an environmentally resistant pathogenic bacterium with the ability to prolong survival under environmental conditions. Therefore, C. difficile in the feces of both symptomatic and asymptomatic CDI patients can enter hospitals and domestic wastewater, which can be the possible approaches for CDI transmission within a community [63]. Moreover, AMR of C. difficile raises a major threat to the global health care system, not only because of the treatment of CDI, but also because it can be a reservoir of AMR genes to spread them to other pathogens [64]. These facts raise concerns of C. difficile infection and transmission and entail its surveillance based on WBE. Several studies have investigated the prevalence of C. difficile in wastewater by cultivation and PCR methods [59,65,66].

2.2. Non-Enteric Pathogenic Bacteria

Many of the pathogenic bacteria detected in wastewater are enteric in origin. However, a few of the pathogenic bacteria, which cause non-enteric diseases such as Legionella spp. and Mycobacterium spp., have also been detected in wastewater [67,68,69].

2.2.1. Legionella spp.

Pneumonia caused by Legionella spp. is a life-threatening pulmonary infection that is mostly caused by Legionella pneumophila [70]. In addition, another 19 species have also been confirmed as human pathogens based on results isolated from clinical specimens [71]. Infections could be spread not only in communities, but also in hospitals. Moreover, Legionnaires’ disease (LD) is clinically and radiologically indistinguishable from community-acquired pneumonia (CAP) caused by other bacterial pathogens [72]. For the treatment, Legionella spp. are unaffected by β-lactam antibiotics since they are obligatory intracellular bacteria. The treatment of infections thus requires a high dose of quinolones or macrolides [73]. Early diagnosis of LD is essential for the monitoring of outbreak and treatment in hospitals [74,75]. Legionella has been found in wastewater with concentrations up to 108 CFU/L. A recent study found that exposure to aerosols dispersed from WWTPs caused LD in residents living near WWTPs during 2013–2018 in the Netherlands [26].

2.2.2. Mycobacterium spp.

The Mycobacterium genus includes more than 170 species [76], of which at least two, Mycobacterium tuberculosis and Mycobacterium leprosy, are regarded as obligate human pathogens. Most others are opportunistic organisms that cause disease both in humans and animals when conditions are favorable. Generally, Mycobacteria are classified into two main groups, the genetically related M. tuberculosis complex (MTC) organisms and nontuberculous mycobacteria (NTM). The NTM are also known as environmental mycobacteria due to their widespread presence in soil and water [77]. Tuberculosis (TB) is a disease caused by infection with M. tuberculosis, which caused 1.4 million deaths in 2019. TB became one of the top ten causes of death and is the leading cause from a single infectious agent (ahead of HIV/AIDS). In addition, multidrug-resistant TB (MDR-TB) is also a public health crisis and a health security threat. In 2019, 206,030 patients with multidrug-resistant or rifampicin tuberculosis (MDR/RR-TB) were detected and reported globally, an increase of 10% from 186,883 in 2018 [78]. However, nontuberculous mycobacteria (NTM) have never been quantified in wastewaters before Radomski’s study because of the inefficient analytical approaches [27]. More wastewater studies should be conducted for its environmental surveillance.

3. Molecular Methods for Pathogenic Bacteria Detection

3.1. Biomarkers of Pathogenic Bacteria

Biomarkers including nucleic acids, proteins, antigens, adenosine triphosphate (ATP), and metabolic products [10] are employed in the analysis of microorganisms. To differentiate microorganisms within one sample, nucleic acids (DNA/RNA), proteins, and antigens are usually selected as biomarkers because of their special physical and chemical characteristics within different pathogens. The detection of DNA/RNA is based on the specific hybridization and amplification of targets, thus enabling good specificity and accuracy. In case of pathogenic bacteria in wastewater, the most important biomarker is the pathogenic DNA or RNA residues from these bacteria. The biomarkers include the genus/species-specific genes, functional genes, and antimicrobial resistance genes [11]. Moreover, in the analysis of antimicrobial resistance, gene transfer is another significant point. Various mobile genetic elements, including plasmids, transposons, bacteriophages, integrons, and combinations of them, are notable nucleic acid targets for investigating the prevalence and spread of resistance genes in bacteria [79].
ATP assay, enzymatic activity tests, and metabolic products are mostly used to assess the activity of living cells. Due to the linearity between the total number of ATP and the total colony-forming units, the metabolically active cells could be directly quantified using the amount of ATP [80,81]. Ions and some organic acids are the metabolic products of microorganisms. These metabolites could be detected by electrochemical methods, thus were utilized to reflect the metabolic states of microorganisms [82].
Microbial surfaces contain a variety of proteins that are expressed by specific DNA/RNA in different pathogens. By screening these proteins using antibodies and nucleic acids, new biomarkers can be discovered, and pathogens can be specifically detected. Antigens are another kind of molecules on the cell surface of pathogens. They can be specifically bound to antibodies and induce immune responses of the host. Each type of pathogen carries one or more unique antigens on their surface, even within strains. It thus enables the specific identification of pathogens using antibodies [83]. Moreover, by analyzing the specific antigens of each strain, the subtypes of the strain can be determined.
Aptamers are single-stranded DNA or RNA oligonucleotides with high affinities and specificities that can bind a variety of targets, from single molecules to whole cells [84]. They can form diverse, complex secondary structures such as multi-branched loops and G-quadruplexes, which can specifically target the surface proteins of microorganisms or cells. In environmental monitoring, aptamers are superior to antibodies due to their chemical stability, easy chemical modification, relative ease of synthesis, and biocompatibility. With the systematic evolution of ligands by exponential enrichment (SELEX) method, many aptamers have been successfully employed to detect various pathogens in environmental samples [8,85,86].

3.2. Molecular Methods

For a long time, the culture and colony counting-based method has been the dominant method in the detection of pathogens (i.e., the ‘gold standard’). It can assess live microbes or viable cells in samples. However, these methods may produce false-positive or false-negative results when evaluating highly aggregated microbial cells. Furthermore, not all microbial cultures can be grown under laboratory conditions. For example, a study of Campylobacter indicated that the culture-based method failed to correctly detect Campylobacter in 30% of positive patient stool samples compared to non-cultural methods, including PCR and enzyme immunoassay (EIA) [87]. Moreover, the culture methods are time and resource intensive, which are gradually replaced by more rapid and specific molecular methods. Therefore, in order to meet the requirements for reliable analysis of pathogenic bacteria, including high specificity, high sensitivity, good reproducibility, automation, and cost effectivity, molecular methods have gradually emerged to replace the dominant position of culture methods. In recent decades, various rapid, sensitive, and specific molecular methods have been developed. These molecular methods are discussed below and listed in Table 2.

3.2.1. Nucleic Acid Targeting Methods

Nucleic acid targeting methods are designed to detect the specific DNA/RNA of pathogens. It is achieved by the hybridization between target nucleic acid sequences and synthetic oligonucleotides. Thus, the species-specific gene of pathogens and virulence genes can be detected through nucleic acid targeting methods. They are usually fast, efficient, and do not require the culture of the pathogens. These methods include polymerase chain reaction (PCR)-based methods such as conventional PCR, real-time/quantitative PCR (qPCR), droplet digital PCR (ddPCR), multiplex PCR (mPCR), and other methods such as microarrays, loop-mediated isothermal amplification (LAMP), sequencing, and fluorescence in situ hybridization (FISH).

PCR-Based Method

PCR is the most common molecular-based technique for the detection and quantification of pathogens. PCR enables the detection of a single pathogenic bacteria by targeting specific DNA sequences [57]. Through this method, a small sample of a DNA sequence could be rapidly amplified into a large amount. This advantage enables the detection and quantification of a low amount of the target DNA sequence. It is thus widely used in the diagnosis of human pathogens. It significantly increases the sensitivity of detection of microorganisms at low concentrations in environmental samples [103,104]. PCR has already been utilized to the detection of a series of pathogenic bacteria such as E. coli and spores of C. perfringens [104,105].
Conventional PCR needs gel electrophoresis to detect the formation of PCR products. Real-time polymerase chain reaction, also called quantitative PCR, is the real-time detection of the PCR process during the amplification of the target DNA sequence. qPCR determines the PCR amplification by measuring specific dual-labeled probes or fluorescent signals emitted by inserting dyes. The fluorescence intensity reflects the amount of the template DNA. There is a linear relationship between the cycle threshold (Ct or Cq) value and the initial concentration of the template gene during the exponential period of PCR amplification. Thus, the concentration of target sequences could be calculated from a well-established standard curve to achieve an absolute quantification. Real-time quantitative PCR is widely used in the real-time detective and quantitative analysis of target DNA sequences with higher specificity and sensitivity than conventional PCR [106].
Two main fluorescence systems have been developed for qPCR, i.e., the SYBR green method and the TaqMan probes method. SYBR green is a fluorescent pigment that can bind double-stranded DNA (dsDNA). This non-sequence-specific pigment enhances the fluorescence signal when it binds to DNA double helix minor grooves, thus enabling the quantification of the targeting sequence. In contrast, TaqMan probe does not require the addition of fluorescent pigment. The template-specific TaqMan probe further improves the specificity of qPCR by increasing primer specificity. For each amplification of a specific target, one molecule of fluorescent dye is released. The instrument detects the fluorescence produced by specific amplification, which is not impacted by non-specific amplification. This ensures the high specificity of the qPCR detection. There are many reporter–quencher sets with different wavelengths, which can be labeled with the TaqMan probe. This enables the TaqMan method to be able to detect multiple PCR reactions in the same tube, leading to reduced cost and improved efficiency and accuracy. It can also avoid the influence of different fluorescent dyes on the PCR reaction.
The mPCR is a faster detection methodology than simplex PCR, which can detect multiple gene targets simultaneously. Fan et al. (2008) reported one mPCR assay to achieve the simultaneous detection of various human pathogens in a single tube, with the detection sensitivities between 10 to 102 CFU/100 mL in seawater. To differentiate the pathogenic and commensal E. coli in clinical and water samples, an mPCR assay was developed to detect the occurrence of 11 virulence genes in E. coli [107]. Recently, the presence of enteropathogens in sewage was investigated by using the commercially available FilmArray® mPCR system [88]. Compared with simplex PCR, mPCR provides faster detection by simultaneously amplifying multiple gene targets. It can also differentiate closely related pathogenic bacteria.
Digital PCR is a biotechnology improvement on conventional PCR and can be used to directly amplify and quantify DNA, cDNA, or RNA. Droplet digital polymerase chain reaction (ddPCR) is a kind of dPCR technique that is emerging as a powerful analytical tool for absolute quantification. Similar to qPCR, ddPCR also utilizes Taq polymerase to amplify a target DNA sequence in a standard qPCR assay. The differences are that ddPCR separates the whole qPCR reaction into thousands of individual reactions before amplification, and ddPCR collects data at the reaction end point. These differences provide ddPCR many advantages, such as the direct and independent quantification of target DNA without standard curves and more precise and reproducible data than conventional qPCR, especially when PCR inhibition is present [108,109,110]. In comparison to qPCR, ddPCR shows better performance in detecting low concentrations of target genes in environmental samples. Moreover, it has the potential to reduce the effect of qPCR inhibitors, although its application to complex environmental samples needs further optimization [111]. However, qPCR is more reliable in detecting higher concentrations (2 × 105 or 2 × 104 gene copies/PCR), since ddPCR displays higher variability and less precision in these concentration ranges [112]. A recent study shows that, in addition to being faster, the ddPCR method exhibited higher sensitivity with a limitation of 10−5 ng/μL for genomic DNA templates and 10−1 CFU/mL for Shigella bacteria culture, when compared to PCR and qPCR [113].
Nowadays, with the increasing availability of sequencing data, it is theoretically possible to design qPCR assays for every microorganism [106]. The qPCR method has many benefits over other techniques. Firstly, the quantitative data produced by qPCR method could reach an accurate dynamic range of 7–8 log orders of magnitude without requiring post-amplification manipulation. Secondly, although the sensitivity of qPCR is varied towards different samples and might be inhibited by inhibitors, it has been reported to have higher sensitivity than many other molecular methods [114]. Theoretically, it is high enough to detect a single copy of a transcript. Studies have shown that qPCR is reliable and sufficient for the quantitative detection of various pathogens such as E. coli O157:H7 [115] and Campylobacter spp. [116] The qPCR method has been applied to detect and monitor the occurrence and concentrations of pathogens in drinking water sources [117], water and wastewater treatment plants [118,119], and recreational beaches [120]. The use of qPCR in water analysis enables quick microbial risk assessment, which may lead to immediate remedial actions.
However, for the detection and quantification of waterborne bacteria with low abundance, high requirements are demanded for qPCR, such as high accuracy, low limit of detection (LoD) and quantification (LoQ), and the ability to distinguish dead and viable cells. Studies have reported that it is possible to detect viable cells by detecting messenger RNA (mRNA), since it only exists in viable organisms [7,121]. However, not all mRNAs are present in all life phases of an organism, thus the target mRNA should be carefully chosen for viable organism detection. In addition, rRNA-based RT-qPCR assays were also confirmed to have a better association with the active bacterial populations in surface water samples than rDNA-based assays [122,123,124]. Furthermore, multiplex real-time PCR has been reported to be a valuable technique for the identification of viruses [125], bacteria [126], and parasites [127]. However, due to the limitation of instruments and the fluorescence groups, only four targets could be detected at the same time in TaqMan methods. This limitation prohibits its application in profiling microbiome communities in complex samples.

DNA Microarrays

DNA microarrays, also known as DNA arrays, are commonly known as gene chips. It is a special piece of glass or silicon chip with a DNA microarray, which places thousands or tens of thousands of nucleic acid probes on an area of several square centimeters [128]. DNA, complementary DNA (cDNA), and RNA in the sample are detected by fluorescence or electric signal after being combined with the probes. DNA microarrays enable the hybridization-based detection of multiple targets in a single experiment, which makes it suitable for the analysis of massive targets. Using a high-throughput DNA microarray assay, a study investigated the prevalence of 941 pathogenic bacteria in groundwater and differentiated their sources of origin [129]. In general, DNA microarray allows for the simultaneous detection of multiple pathogenic bacteria. It is thus a fast and reliable diagnostic method in analyzing large numbers of clinical/environmental samples. However, the complicated probe design work, the reliability of the microarray data, and the clinical applicability of the early results have been criticized [130]. The criticism and intensified competition from other technologies, such as next-generation sequencing (NGS), have hampered the growth of microarray-based testing in the molecular diagnostics market [131].

Loop-Mediated Isothermal Amplification (LAMP)

LAMP is an isothermal nucleic acid amplification technique. It has been utilized for the alternative detection of certain diseases because of its low cost. At present, LAMP has been applied to the identification and quantification of pathogenic bacteria with significant advantages in sensitivity, specificity, and rapidity [95,132]. Since LAMP requires four primers specifically designed for six different regions of the target, any incomplete matching of the primers will theoretically lead to the phenomenon of non-reaction and non-specific amplification. In addition, the LAMP method was confirmed to be 10–100 times more sensitive than PCR detection [133], with a detection limit of 10 copies or less in the template for one reaction. Furthermore, it can directly detect pathogenic microorganisms in diseased tissue, thus avoiding the tedious cultivation and nucleic acid extraction step [134]. Finally, and most importantly, the result of the reaction can be judged with naked eyes by demonstrating the absence of the target gene with the production of white precipitate of magnesium pyrophosphate. It is more difficult to design specific primers for LAMP than PCR (because LAMP requires 4–6 primers and PCR requires only two). A software tool named PrimerExplorer is available to help the primer design for LAMP (http://primerexplorer.jp/e/). Therefore, as a rapid detection method without the need of any equipment, LAMP shows great potential in the rapid diagnosis of human pathogens in various samples.

Fluorescent in Situ Hybridization (FISH)

FISH is a cytogenetic technique used to detect and locate nucleic acids in cells or sample matrices. Fluorescently labeled nucleic acid probes hybridize only with highly similar nucleic acids and can be used to locate genes on chromosomes or to label ribosomal RNA in different taxonomic bacteria or archaea in molecular ecology. FISH could be employed in the enumeration of particular microbial populations [135]. Compared to PCR, FISH is more suitable for complex matrices because of its lesser sensitivity to inhibitory substances. However, a major limitation of FISH is the small number of phylogenetically distinct targets that can be detected at the same time. A recent study developed a multi-FISH method that uses eight fluorophores, which is highly suitable for investigating the structure and function of microbial communities in different samples [96]. Furthermore, FISH has been used to discover emerging human pathogens in water, wastewater, and sludge, to produce quantitative descriptions of the microbial community in wastewater and activated sludge [136,137] and to investigate survival and infection mechanisms at the cellular level. However, this method is still partly based on cell culture.

Sequencing

Sequencing is the process of determining the sequence of nucleotides in a section of DNA. It includes any method or technique used to determine the order of the four bases: adenine, guanine, cytosine, and thymine (or uracil for RNA). In 1977, DNA sequencing technology was firstly developed by Frederick Sanger based on the chain-termination method (also known as Sanger sequencing). In the early stage, DNA sequencing was employed for small genomes such as viruses and organelles. Complete sequencing of a bacterium genome was not feasible because of the economic and technical limitations. Later, with the emergence of the shotgun method developed by Sanger et al., whole genome sequencing of bacteria was achieved. The shotgun method is considered the gold standard, and whole genome sequencing of many bacteria has been carried out using this method over years [138].
Next-generation sequencing (NGS), also known as high-throughput sequencing, is the overall term used to describe several different modern sequencing pathways. These technologies allow DNA and RNA to be sequenced faster and at lower cost than Sanger sequencing, which was previously used, thus revolutionizing genomics and molecular biology research [139]. NGS technologies include Illumina (Solexa) sequencing, Roche 454 sequencing, and proton/PGM sequencing. The NGS technologies achieve high throughput and reduced cost by using massively parallel analysis, which allows 300 Gb of DNA to be read in a single run on a single chip. The four main advantages of NGS over classical Sanger sequencing are: (i) NGS needs significantly less DNA, as it can obtain a sequence from a single strand; (ii) NGS is significantly quicker than Sanger sequencing by combining the two separate processes of Sanger sequencing, i.e., chemical reaction and signal detection, in some versions of NGS; (iii) NGS is more cost-effective due to reduced time, manpower, and reagents; (iv) repeats in NGS caused by many short overlapping reads lead to a more accurate and reliable sequence, even though individual reads are less accurate. These advantages enable a great potential of NGS in the application of environmental research. NGS is capable of producing large numbers of reads at exceptionally high coverages throughout the genome with dramatically reduced cost through the massively paralleled approach. However, NGS requires the amplification of DNA molecules, which introduces random errors in the DNA synthesis. The amplified DNA strands would become progressively out-of-sync, which means the signal quality deteriorates as the read length grows. Therefore, long DNA molecules must be broken up into smaller pieces to maintain the quality of the reading, leading to a critical limitation of second-generation sequencing [140].
To solve the limitation, third-generation sequencing (TGS) technologies were developed to produce substantially longer reads than NGS by the direct sequencing of single DNA molecules. Nanopore sequencing (Oxford Nanopore Technologies, Oxford, UK) is a representative TGS approach for the sequencing of biopolymers, specifically polynucleotides in the form of DNA/RNA. Through nanopore sequencing, individual molecules of a DNA/RNA can be sequenced without PCR amplification or chemical labeling of the sample. Nanopore sequencing has a great potential in providing relatively low-cost genotyping, high mobility for testing, and the ability to rapidly process samples and display results in real time [141]. Applications of this method in the rapid identification of viral pathogens [142], plant genome sequencing [143], monitoring of antibiotic resistance [144], and haplotyping [145] has been reported. One major limitation of nanopore sequencing is its high raw read error rate, which remains between 5% and 15% despite recent improvements in nanopore chemistry and computational tools [138]. However, according to the latest updates from Nanopore technologies (accessed on 15 October 2021), an accuracy of 98.3% could be achieved through the production software MinKNOW 4.3 (“Super-accuracy” basecalling model) and Guppy 5 (https://nanoporetech.com/accuracy). In addition, the quality of the sequencing result is affected by library quality and the presence of sequencing inhibitors. Although more efforts are needed to improve the quality of Nanopore sequencing results, studies have confirmed that it has better bacterial identification performance in complex samples than traditional Illumina platforms [146]. Winand et al. compared the bacterial identification performance of second (Illumina) and third-generation sequencing technologies (Nanopore sequencing, Oxford Nanopore Technologies, Oxford, UK) by targeting the 16S rRNA gene. The results revealed that both techniques provide reliable identification of bacterial genera but may mislead the identification of bacterial species and constitute viable alternatives to Sanger sequencing for rapid analysis of mixed samples without any culture steps [98].

3.2.2. Immunology-Based Methods

Immunological methods are based on the specific interaction between antibodies and antigens. These methods include enzyme-linked immunosorbent assays (ELISA), immunofluorescence assays (IFA), and serum neutralization tests (SNTs) [6]. For immunology-based methods, specific fluorochrome labeled antibodies are used to capture targeted antigens, which serves as the enumeration of fluorescently labeled cells by detecting the fluorescence signal using microscopy or flow cytometry. However, the biomarkers of these methods should be carefully chosen to achieve specific detection at different classification levels including genus, species, and serotypes. The detection of S. typhimurium on an immunochromatographic strip was reported by Park et al. (2010). This study achieved the quantitative detection of S. typhimurium in the range of 9.2 × 103 to 9.2 × 106 CFU/mL in river samples within 20 min [100].
Although these methods can specifically detect targeted bacteria and their toxins and can be multiplexed for multiple samples, they are still limited by false-negative results and cross-reactions with similar antigens. False-negative results are a serious problem and often happen to classical methods. They can be induced by various inhibitory compounds and matrices of different types of samples, which vary largely and thus might cause different effects on the analytical performance of different detection methods. In addition, cross-reaction is another big problem for immunology-based methods. In one study, a monoclonal antibody was used for specifically detecting E. coli O157:H7 lipopolysaccharide (LPS1) [147]. The cross-reactivity with other bacteria happened due to the presence of a constituent sugar of LPS. One recent method comparison study for C. difficile surveillance in Switzerland showed that, compared to the PCR method, enzyme immunoassay led to more false-negative results of human stool samples [148]. Immunological methods usually require pre-enrichment to expose surface antigens, which leads to extended detection time. Moreover, due to their lower sensitivity than other molecular methods, immunology-based methods were less employed in the direct detection of pathogenic bacteria in wastewater samples [9].

3.2.3. Biosensor-Based Methods

A biosensor is an analytical platform composed of two elements: a bio-receptor and a transducer. Bio-receptors are responsible for recognizing the targets such as enzymes, proteins, nucleic acids, and cell receptors. After recognition, the transducer converts the biological interactions into electrical signals that can be measured (e.g., optical, electrochemical, or magnetic). Biosensors provide a rapid, real-time, on-site, and multiple detection of bacteria. Optical biosensors are selective, sensitive, and can be used for real-time monitoring of toxins, drugs, and pathogens in wastewater [149]. For example, by applying a fluorescently labeled specific aptamer, Yildirim et al. developed a portable optical biosensor for the indirect sensing of an E. coli O157:H7 strain in wastewater [150]. Surface-enhanced Raman scattering (SERS) pathogen biosensors, with noble metal nanoparticles (e.g., silver and gold) as an impressive substrate, have become an attractive research field. The colorimetric changes induced by the hybridization between single-stranded DNA probes modified by gold nanoparticles and their complementary DNA can avoid the requirement of expensive and complex fluorescent labeling [151]. Gold nanoparticles are widely used in biosensor instruments, especially for dark water samples [152]. Another notable biosensor for E. coli O157:H7 used carboxyl functionalized graphene quantum dots (cf-GQDs) to label a specific antibody [101]. This sensor can specifically recognize E. coli O157:H7 from different sources, such as water and food, with the minimum detection limit of 100 CFU/mL. However, the sensitivity to changes in pH, mass, and temperature are some of the challenges that must be addressed in using biosensors for bacterial pathogens in wastewater [153].

3.2.4. Paper-Based Device

A paper-based device is a small analytical tool that is printed by a wax printer and has different functional areas. It can integrate all the processes required for nucleic acid detection (enrichment, extraction, amplification, and visual detection) into a cheap paper material [154]. The whole detection process can be completed by folding paper-based devices in different ways and in different sections, which overcomes the limitation of PCR tests. Paper-based device can achieve multichannel, sensitive detection, comparable to PCR detection, and provide a high-quality, rapid, and accurate diagnosis of pathogens [155]. Moreover, paper-based devices are easy to stack, store, and transport because they are thin, lightweight, and of different thicknesses [156]. Similar to biosensors, paper-based devices can also be used to target a variety of biomarkers, including nucleic acids, proteins, antigens, and chemicals [9,102,155,157]. By integrating various molecular detection methods, paper-based devices have emerged as a powerful platform for the fast diagnosis of pathogens and the determination of infection transmission [9,13,102]. However, the shelf life of paper-based device limits its further applications. Some paper-based devices contain reagents with a short shelf life, such as enzymes, and thus need to be stored in a refrigerator or freezer to maintain the activity of the reagents [158,159]. Studies about the shelf life of enzymes on paper-based devices have yielded some promising results and proven techniques, although further research is still needed. Furthermore, the analytical performance of paper-based devices is also limited by features of paper, including the paper fibers and pattern. Moreover, the coffee ring effect of paper-based devices can cause non-uniform distributions of detection reagents and samples, thus affecting the detection accuracy [158]. To overcome these limitations, future efforts should be put into developing more uniform papers and modifying the size and shape of the test zone.

4. Recent Advances of Molecular Methods for Pathogenic Bacteria in Wastewater

Current estimates of the burden of infectious diseases are often based on severe cases requiring hospitalization, which fails to cover asymptomatic patients. The emerging wastewater-based epidemiology (WBE) is based on the analysis of biomarkers in raw wastewater, which is then used to back-estimate the status of public health. Wastewater is a complex mixture of chemicals and microorganisms in water. It contains chemical and biological information directly discharged from our bodies. From a surveillance point of view, urban wastewater is an attractive resource, since it provides sampling material within a large and mostly healthy population.
The WBE approach was first outlined as a potential tool to evaluate the use of illicit drugs and misused therapeutic drugs within a community [160]. To date, WBE has become an important tool for estimating illicit and licit drug consumption by detecting and quantifying unchanged drugs and their human-specific metabolites in wastewater [12]. WBE studies also showed that wastewater can reveal not only illicit drug use and diet, but also infectious disease risk within a community [11,161,162]. Many studies have validated the feasibility of various molecular methods in wastewater. Sensitive and specific methods such as PCR, real-time PCR, and DNA sequencing have been employed in the analysis of wastewater to achieve the fast detection and accurate quantification of human pathogens [90,163,164] or the evaluation of community structure and antimicrobial resistance level [165,166,167].

4.1. Sample Processing and DNA/RNA Extraction Methods

Wastewater components, including fats, proteins, humic acids, and fulvic acids, can lead to problems in the downstream analysis (molecular detection). Wastewater sample processing is a key step for the detection of pathogenic bacteria by separating, concentrating, extracting, and purifying biomarkers for further analysis. The availability of different commercial DNA/RNA extraction kits showed variable efficiency when extracting samples such as wastewater and sediment. Table 3 lists several comparison studies of different sample storage, pre-treatment, and DNA/RNA extraction methods for various downstream analyses.
Mumy and Findlay developed an external DNA recovery standard for sediments by comparing the performance of three commercial kits (UltraClean™Soil DNA, FastDNA®SPIN®, and Soil Master™DNA Extraction) [168]. The results indicated that the FastDNA®SPIN® kit has the highest recovery rate and makes it possible to collect additional DNA by cleaning beaded sediments. Gyawali et al. investigated six rapid DNA extraction methods for recovering Ancylostoma caninum ova DNA from wastewater and reported that the filtration method recovered higher DNA concentrations in both treated and raw wastewater than centrifugation, hollow fiber ultrafiltration (HFUF), and flotation [169]. A comparative study about the relative effectiveness of 10 different bacterial DNA extraction methods for wastewater samples showed that only a few could achieve satisfactory results when applied to bacterial pathogens [170]. The method of combined bead beating separation and ammonium acetate purification was suggested as the most suitable approach for bacterial DNA extraction from wastewater prior to specific microbial detection using microarray hybridization technology. Li et al. compared the ARG sequencing analysis results of three DNA extraction methods [171]. It was found that ARGs captured by the FastDNA SPIN Kit for Soil had the highest DNA yield, purity, and diversity. Moreover, no discernable effects were found on ARG profiles with fixation in ethanol, deep-freezing, and overseas transportation of samples compared with fresh samples. Another comparative study indicated that the DNA Mini kit and PowerSoil kit produce the most consistent sequencing results in water and wastewater [172]. Collectively speaking, the performance of DNA/RNA extraction methods of wastewater varies, and it is essential to develop standard DNA/RNA extraction methods for different downstream analysis methods to achieve high recovery and quality of the nucleic acids of various pathogens in wastewater.

4.2. Detection and Quantification of Pathogenic Bacteria

The low concentration of targeted pathogenic bacteria in wastewater samples brings difficulties to their detection, which entails high sensitivity and repeatability. As a complex matrix, wastewater contains various inhibitors for a number of molecular methods, which is a significant challenge for their application in wastewater analysis. In addition, from a disease surveillance perspective, wastewater samples should be analyzed quickly enough to provide an early warning. Several molecular approaches have showed great potential in rapid analysis. Table 4 critically compares various detection methods used for wastewater samples in the last five years.
So far, the PCR-based method is the most popular molecular approach for specific pathogenic bacteria detection and quantification (relative and absolute quantification) in wastewater because of its high sensitivity, specificity, and the low cost compared to the sequencing method. Plenty of primer-probe sets targeting various human pathogenic bacteria have been developed. Those primer-probe sets showed high specificity, sensitivity, and efficiency for pathogenic bacteria detection at genus and species levels. A series of PCR-based methods with or without bacteria isolation and cultivation procedures has been reported for the fast detection and quantification of pathogenic bacteria in wastewater samples [173,174,175]. Recently developed primer-probe sets for the bacterial pathogen detection of various PCR-based methods are listed in Table 5. Some of them have been confirmed to have good performance in wastewater samples and thus could be potentially used in wastewater analysis, although further feasibility studies should be conducted. Among the PCR-based methods, ddPCR has been increasingly reported as having better analysis performance than traditional PCR-based methods in wastewater [186]. However, some studies have still claimed that the sensitivity of qPCR is superior to ddPCR in some circumstances (e.g., high targeted gene concentration), and their performance might vary with different assays [187].
The LAMP method showed great potential in the application to wastewater samples because of its advantages such as inhibitor resistance, short reaction time (<1 h), and no need for advanced thermal cycling instruments [192]. By targeting the lmo0753 gene, Nathaniel et al. developed a loop-mediated isothermal amplification assay for the detection of L. monocytogenes in wastewater. The LoD was 65 fg/µL of DNA and 38 CFU/mL, which was 10 times more sensitive than conventional PCR with primers targeting the HlyA gene. However, in the application to wastewater, a pre-culture procedure at 37 °C for 48 h was required [179]. An SA23 probe targeting Salmonella specifically by FISH has been developed by Santiago et al. (2008). The SA23 probe was shown to be capable of rapid and specific identification and visualization of Salmonella cells directly in the sample. By combining with the pre-enrichment, it could achieve the detection of 1 CFU/mL in seeded meat products [137]. This study also demonstrated the resistance of FISH to inhibitory substances in wastewater and the ability to differentiate viable but non-culturable (VBNC) cells. The advantage of the FISH method is that it is not inactivated by inhibitors and does not depend on the type of sample, even when dealing with large numbers of samples. Sequencing is also a powerful analysis tool for not only detection and relative quantification but also absolute quantification of bacteria in environmental samples, similar to the PCR-based method [193]. By spiking the samples with internal microorganism markers, the absolute bacterial number of targeting microbiomes could be calculated through the abundance of the internal markers with a known number. Different types of spiking markers have been used previously, including indigenous microorganism, synthetic, and heterogeneous markers [193,194,195]. The reference markers and spiking strategy should be optimized because only validated markers can be used to achieve reliable results [196].
In the application of biosensor-based methods, surface-enhanced Raman scattering/spectroscopy (SERS) has a high sensitivity, although its stability is unsatisfactory, and that limits its application in wastewater analysis [149]. Furthermore, colorimetric and fluorescent sensors generally have poorer LoDs than electrochemical, and they are easily disrupted by colored or turbid samples. Thus, it seems that the electrochemical aptasensor is more reliable in wastewater matrices [197]. In addition, the biosensor-based method showed good stability in wastewater samples. For example, Riquelme et al., developed a stable oligonucleotide-functionalized gold nanosensor for mecA ARG monitoring in 2017 [180]. This mecA-specific nanosensor can keep stable under environmental conditions and at high ionic strength, and it can demonstrate high selectivity even in the presence of target interference. This study supports the environmental suitability of a new, low-cost, field-deployable, and large-scale ARG analysis tool.
Most detection and quantification methods for pathogenic bacteria in wastewater involve sampling, which is followed by transportation to a central analytical laboratory for further analysis. In comparison, paper-based devices could achieve multiplexed, sensitive assays that rival PCR-based methods and provide high-quality, fast precision on-site diagnostics for pathogens. Although wastewater is a complex substrate, paper-based devices have shown the potential to detect pathogens in wastewater. Due to the outbreaks of COVID-19, paper-based devices are being quickly developed and employed in SARS-CoV-2 detection in sewage. In another previous study, it was successfully employed in the detection of various genes and microbiomes. A fully disposable and integrated paper-based sample-in-answer-out device was developed for nucleic acid testing, which can sensitively detect S. typhimurium with a detection limit of as low as 102 CFU/mL in wastewater [181]. The presence of β-lactamase-mediated resistance was also detected using paper-based analytical devices (PADs). It was shown that, compared to traditional methods including culture methods, antibiotic susceptibility testing, and PCR gene analysis, their method can still reduce the laboratory processing time by 14–20 h, although a laboratory is still required to concentrate the wastewater samples [157].

4.3. Profiling Potential Pathogens

Sequencing, as a powerful analysis tool, has been widely used for profiling bacterial diversity and potential pathogens in wastewater. The DNA sequencing-based method can perform large-scale parallel analysis of PCR products and environmental nucleic acids. This provides a new dimension for the analysis of pathogenic bacteria in wastewater. The application methodology of NGS technologies in wastewater study can be divided into four subcategories: whole genome sequencing (WGS), metagenomic sequencing, metatranscriptomic sequencing, and sequencing of an amplified targeted gene (e.g., 16S rRNA and 18S rRNA genes) [198]. WGS is a powerful approach for microorganism identification in wastewater, while it relies on bacteria isolation and culture, extraction of long DNA, and the development of long read sequencing platforms. Future development on these aspects could advance and simplify its application in wastewater analysis [199].
To date, sequencing assays based on the amplified gene regions take the dominant place in wastewater analysis. Microbial communities of waterborne pathogens were often studied by targeting high-variation region sequences of small subunit (SSU) rRNA genes (e.g., V1, V3, V4, V6) and large subunit (LSU) rRNA genes [182]. By using the full-length 16S rRNA gene sequence, Numberger et al. characterized and compared bacterial communities of the influent and effluent of a WWTP in Berlin, Germany [184]. The study found that during sewage treatment, the relative abundance of most pathogenic bacteria was effectively reduced, while Legionella and Leptospirosis showed an increase in relative proportion from inflow to effluent. This indicated that WWTPs may enrich and release certain potential pathogens into the environment, although they are effective in removing enteric bacteria. Oluseyi et al. studied the presence of pathogenic bacteria in three WWTPs in South Africa. Their study also confirmed the presence of bacterial pathogens in treated effluent, which may pose a potential contamination risk by transmission through soil, agriculture, water, or sediments [185]. Using Illumina MiSeq sequencing, Xue et al. (2019) investigated the spatial and temporal variability of bacterial structure and the presence of a human-associated Bacteroidale (HF183) marker in two WWTPs. Their findings illustrated how changes in bacterial communities can serve as a reliable means of monitoring the quality and performance of wastewater treatment plants for public and environmental health purposes [200].
Metagenomic study is an emerging methodology based on the sequencing data of genetic material recovered directly from environmental samples. This method has developed rapidly in the detection of microbial communities and their functional capabilities in wastewater. Currently, the application of metagenomics in wastewater is commonly employed for the identification of ARGs and genes associated with pathogens [183,201]. It is also increasingly utilized to support the assembly of whole or partial genomes from short-read sequencing data acquired from uncultured microbial communities [202]. Metatranscriptomic sequencing has similar principles with metagenomic sequencing, but it targets RNA rather than DNA, which is essential for identifying RNA viruses. Sequencing the transcribed mRNA could provide the information about which microbes are functionally active, as mRNA degradation rate varies among different species, thus challenging the preservation and analysis of mRNA [203]. In addition, the excess of ribosomal RNA (rRNA) in transcriptomes also interferes with the identification of mRNA in environmental samples. All these challenges limit its application in wastewater systems [198]. Comparison studies have also been conducted to access the performance of various methods to identify pathogens and associated virulence genes. For example, Yergeau et al. compared pre- and post-treatment biosolids from two WWTPs by using enumeration methods combined with molecular techniques including quantitative PCR, 16S rRNA and cpn60 gene amplicon sequencing, and shotgun metagenomic sequencing [204]. Their study showed that shotgun metagenomics indicted the widest range of pathogen DNA and was the only method that can obtain functional gene information in treated biosolids among all approaches.

4.4. Antimicrobial Resistance Analysis

With the growing concern about antimicrobial resistance, the WHO established the Global Antimicrobial Resistance Surveillance System (GLASS) in 2015 for sharing information on a global level to strengthen data on national and international actions and to aid decision making [205]. WWTPs treating wastewater from different sources provide a suitable circumstance for the emergence and spread of antibiotic resistance genes (ARGs) and antibiotic resistant bacteria (ARB) [79,206]. Analysis of ARGs in influent wastewater can provide a broader perspective for the study of ARGs in the population. Various ARGs have been investigated and reported in wastewater based on qPCR technology [207,208]. A quantitative analysis of ARGs and horizontal gene transfer (HGT) potential was conducted over four seasons at a WWTP using a high-throughput qPCR array [166]. In this study, 285 primer sets targeting ARGs and nine transposase genes related to HGT were successfully used with wastewater samples. A microfluidic quantitative polymerase chain reaction (MF-qPCR) method was developed and optimized for simultaneously quantifying 16S rRNA genes, ARGs, heavy metal resistance genes, and an integrase gene that encodes three different types of integrons. This MF-qPCR method has better detection limits than shotgun metagenomics, which has also been used to detect large amounts of ARGs in other studies [92,209].
Sequencing-based methods also contributed to the analysis of ARGs and ARB (e.g., prevalence, relative abundance, and persistence) in wastewater. Using metagenomic analysis, a pipeline covered the analysis of gene transfer potential and the potential, pathway, and phylogenetic origin of ARGs was developed for identifying antibiotic resistance determinants in wastewater samples [99,182,183]. Meanwhile, discrepancies in ARG quantification by using different sequencing approaches have been reported by several previous studies where some ARGs were only identified by Nanopore sequencing and some others were only detected via Illumina sequencing [210]. Sequencing platform biases on the ARG quantification outputs were due to different ARG identification or prediction algorithms. The Illumina algorithm is based on the similarity search using BLAST, whereas Nanopore sequencing is based on workflows designed for the alignment of long high-error-rate sequences [211].

4.5. Prospect of Molecular Methods for Pathogenic Bacteria in Wastewater Analysis

Based on the above recent advances of various molecular methods, their advantages and limitations in wastewater analysis are summarized in Table 6. All molecular methods are able to achieve the detection of pathogenic bacteria in wastewater with an LoD range from 1 to 100 CFU or gene copies per 100 mL. However, the LoD varies according to different targeted pathogenic bacteria and sample pre-treatment procedures. Overall, the sensitivity of the current molecular methods is adequate for WBE purposes.
Nucleic acid targeting methods have the potential to become the most suitable molecular method for wastewater analysis because one extracted nucleic acid sample could be analyzed by various methods through different downstream workflows to achieve comprehensive analysis, including target gene detection and quantification (relative/absolute), microbiome community profiling, and ARG/functional gene analysis. Moreover, nucleic acid targeting methods could realize the direct identification and quantification of specific genes rather than gene expression products and thus could reduce the mistakes induced by some sample pre-treatment procedures, including enrichment and cultivation. The methods need to be improved to overcome limitations including sensitivity to inhibitors in wastewater, DNA/RNA loss caused by sample processing, and high cost. In addition, more specific gene biomarkers including species-specific genes, host-specific genes, and reliable cell quantification genes (one DNA/RNA fragments per cell) should be carefully selected. Standardization for sample storage, pre-processing, inner-extraction control, recovery methods, and certain analytical methods will highly expand their prospects in WBE study.
Immunology-based methods are powerful methods for detecting and capturing gene expression products such as cell surface proteins and bacterial toxins. For wastewater analysis, they were widely used in specific bacteria detection, isolation, and enrichment. They are more suitable for specific pathogen and microbial antigen detection and isolation from wastewater. They can also combine with other molecular methods to achieve deep analysis of a target pathogen’s genome. By combining with a biosensor platform or paper-based device, they are expected to achieve fast, on-site, real-time, and low-cost identification and enumeration of pathogens in wastewater.
Biosensor-based methods have showed great potential in pathogenic bacteria analysis in wastewater. However, in the real wastewater analysis, they are not cost-effective compared to other molecular methods and cannot process large numbers of environmental samples, such as wastewater, from a long-term surveillance point-of-view. Paper-based devices are a good platform to be combined with many other molecular detection methods to achieve fast analysis of pathogenic bacteria in wastewater. Their application prospect towards wastewater has been well confirmed as a fast, on-site, cost-effective, portable, and disposal device. However, their sensitivity and specificity should be further improved towards detecting various targets in wastewater.
According to different research objectives, appropriate molecular methods can be selected and combined to achieve satisfactory analytical performance. The sensitivity and specificity of different molecular methods should be evaluated on various wastewater samples to improve their analytical performance. Infectious disease surveillance based on WBE could be divided into three main phases including pathogen monitoring in raw sewage, disinfection evaluation and ARG variation in WWTPs, and risk evaluation for further spreading of effluent in the environment. Among those three phases, nucleic acid targeting methods are more suitable for the pathogen monitoring of raw sewage since this phase requires the fast analysis of deep and comprehensive genomic information of pathogenic microbiomes delivered from communities. Especially for the sequencing-based method, with the decreasing cost and by combining with different workflows, it is expected to realize all kinds of analysis, including identification, relative and absolute quantification, bacterial community profiling, and ARG and functional gene analysis. This sets it apart from other nucleic acid targeting methods. For disinfection and ARG evaluation in WWTPs, except for nucleic acid targeting methods, immunology-based methods and biosensor-based methods are powerful analysis tools for the assessment of the activity and infectivity of pathogenic bacteria. For the risk evaluation of effluent and the environment, since the concentration of pathogenic bacteria in effluent and in the surrounding environment is usually low, and this evaluation does not require accurate detection and quantification results, the paper-based device was considered as the best choice since it can achieve fast, cost-effective, and on-site screening of the concentration level of target genes and pathogens. In addition, immunology-based methods could also be used in this phase to evaluate the infectivity and activity of pathogenic bacteria for risk assessment. However, the gold standard approach of sampling, transport and storage, pre-treatment, and the enrichment of biomarkers in wastewater for various downstream molecular detections would dominate in WBE applications. The efficient recovery of high-quality biomarkers from wastewater samples should thus be the priority of future method development.

5. Conclusions

With the ever-increasing concerns over infectious diseases caused by pathogenic bacteria and their antimicrobial resistance, it is widely recognized that effective surveillance is key to the rapid intervention and control of outbreaks of infectious diseases. Wastewater-based epidemiology has become a popular tool due to its great potential as a population prevalence surveillance system and an early warning tool for disease outbreaks. With the development of molecular techniques for the detection of pathogenic bacteria and associated biomarkers, WBE applications are expanding to cover a wide range of pathogens using different advanced molecular methods tailored for wastewater analysis.
PCR-based methods have high sensitivity, and they are broadly used for the rapid analysis of pathogenic bacteria in wastewater, following DNA/RNA extraction procedures. Methods such as DNA microarray and sequencing-based methods are suitable for the in-depth study of bacterial communities and the presence of pathogenic bacteria and antimicrobial resistance due to their capability of large-scale parallel analysis of the whole microbiome. Alternative nucleic acid targeting methods such as FISH and LAMP are relatively sensitive, specific, and cost-efficient. However, nucleic acid targeting methods are not able to provide information about the activity and infectivity of various pathogens in wastewater. In addition, the need for sample pretreatment and research into multiplexing microorganism detection in a single sample are still challenges. Biosensors are easy to operate and do not need trained personnel for the detection of pathogenic bacteria in wastewater. Moreover, paper-based devices have recently emerged and have been widely used in pathogenic bacteria detection in wastewater because of their rapidness and cost effectiveness.
Molecular methods have found their diverse applications in WBE. Nucleic acid-based methods enable the direct and comprehensive analysis of the DNA/RNA of wastewater samples, including target gene (e.g., species-specific genes, ARGs, and functional genes) detection and quantification (both direct and relative quantification), profiling of the whole microbiome in the sample, genome sequencing, and analyzing. Thus, nucleic acid-based methods have the broadest prospect in wastewater analysis. Biosensor-based methods and paper-based devices exhibited great potential in fast and on-site detection of chemicals and microbiomes, which is suitable for the early warning of infectious disease outbreaks. Although, it seems that the application of immunology-based methods in wastewater is limited by a lot of disadvantages, it is the only method that targets the gene expression products of microbiomes. A lot of biosensors and paper-based devices are developed based on immunology-based methods. It is essential to improve immunology-based methods to suit WBE applications.
Although molecular methods have shown great potential in the analysis of pathogenic bacteria in wastewater, there are still several key challenges for their application in WBE. The low concentration of pathogenic bacteria in wastewater and inhibition of the complex wastewater matrix are additional concerns for these methods in comparison to the analysis of other types of samples. The low DNA/RNA recovery efficiency of pathogenic bacteria from wastewater needs to be improved and reported quantitatively with the results. The accuracy and reliability of WBE would be significantly enhanced with well-established molecular detection methods.

Author Contributions

Conceptualization, S.Z. and G.J.; writing—original draft preparation, S.Z.; writing—review, S.Z.; X.L.; J.W.; L.C.; J.O.; and F.H.; supervision, funding acquisition, and writing—review and editing, G.J. All authors have read and agreed to the published version of the manuscript.

Funding

This research was supported by the ARC Discovery project (DP190100385). Shuxin Zhang receives the support from a University of Wollongong PhD scholarship.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Hoffmann, V.; Moser, C.; Saak, A. Food safety in low and middle-income countries: The evidence through an economic lens. World Dev. 2019, 123, 104611. [Google Scholar] [CrossRef]
  2. O’Brien, S.J. Foodborne Diseases: Prevalence of Foodborne Diseases in Europe. Encycl. Food Saf. 2014, 13, 302–311. [Google Scholar] [CrossRef]
  3. Benedict, K.M.; Reses, H.; Vigar, M.; Roth, D.M.; Roberts, V.A.; Mattioli, M.; Cooley, L.A.; Hilborn, E.D.; Wade, T.J.; Fullerton, K.E.; et al. Surveillance for Waterborne Disease Outbreaks Associated with Drinking Water—United States, 2013–2014. MMWR Morb. Mortal. Wkly. Rep. 2017, 66, 1216–1221. [Google Scholar] [CrossRef]
  4. Tiwari, A.; Oliver, D.M.; Bivins, A.; Sherchan, S.P.; Pitkänen, T. Bathing Water Quality Monitoring Practices in Europe and the United States. Int. J. Environ. Res. Public Health 2021, 18, 5513. [Google Scholar] [CrossRef] [PubMed]
  5. Sohier, D.; Pavan, S.; Riou, A.; Combrisson, J.; Postollec, F. Evolution of microbiological analytical methods for dairy industry needs. Front. Microbiol. 2014, 5, 16. [Google Scholar] [CrossRef] [Green Version]
  6. Deshmukh, R.A.; Joshi, K.; Bhand, S.; Roy, U. Recent developments in detection and enumeration of waterborne bacteria: A retrospective minireview. Microbiologyopen 2016, 5, 901–922. [Google Scholar] [CrossRef] [Green Version]
  7. Gilbride, K. Molecular methods for the detection of waterborne pathogens. In Waterborne Pathogens; Elsevier: Amsterdam, The Netherlands, 2014; pp. 231–290. [Google Scholar]
  8. Kudłak, B.; Wieczerzak, M. Aptamer based tools for environmental and therapeutic monitoring: A review of developments, applications, future perspectives. Crit. Rev. Environ. Sci. Technol. 2020, 50, 816–867. [Google Scholar] [CrossRef]
  9. Pang, B.; Zhao, C.; Li, L.; Song, X.; Xu, K.; Wang, J.; Liu, Y.; Fu, K.; Bao, H.; Song, D.; et al. Development of a low-cost paper-based ELISA method for rapid Escherichia coli O157:H7 detection. Anal. Biochem. 2018, 542, 58–62. [Google Scholar] [CrossRef] [PubMed]
  10. Jayan, H.; Pu, H.; Sun, D.-W. Recent development in rapid detection techniques for microorganism activities in food matrices using bio-recognition: A review. Trends Food Sci. Technol. 2020, 95, 233–246. [Google Scholar] [CrossRef]
  11. Sims, N.; Kasprzyk-Hordern, B. Future Perspectives of Wastewater-Based Epidemiology: Monitoring Infectious Disease Spread and Resistance to the Community Level. Environ. Int. 2020, 139, 105689. [Google Scholar] [CrossRef] [PubMed]
  12. Feng, L.; Zhang, W.; Li, X. Monitoring of regional drug abuse through wastewater-based epidemiology—A critical review. Sci. China Earth Sci. 2018, 61, 239–255. [Google Scholar] [CrossRef]
  13. Hui, Q.; Pan, Y.; Yang, Z. Paper-based devices for rapid diagnostics and testing sewage for early warning of COVID-19 outbreak. Case Stud. Chem. Environ. Eng. 2020, 2, 100064. [Google Scholar] [CrossRef]
  14. Choi, P.M.; Tscharke, B.J.; Donner, E.; O’Brien, J.W.; Grant, S.C.; Kaserzon, S.L.; Mackie, R.; O’Malley, E.; Crosbie, N.D.; Thomas, K.V. Wastewater-based epidemiology biomarkers: Past, present and future. TrAC Trends Anal. Chem. 2018, 105, 453–469. [Google Scholar] [CrossRef]
  15. Ahmed, W.; Bibby, K.; D’Aoust, P.M.; Delatolla, R.; Gerba, C.P.; Haas, C.N.; Hamilton, K.A.; Hewitt, J.; Julian, T.R.; Kaya, D.; et al. Differentiating between the possibility and probability of SARS-CoV-2 transmission associated with wastewater: Empirical evidence is needed to substantiate risk. FEMS Microbes 2021, 2, xtab007. [Google Scholar] [CrossRef]
  16. Tiwari, A.; Lipponen, A.; Hokajärvi, A.-M.; Luomala, O.; Sarekoski, A.; Rytkönen, A.; Österlund, P.; Al-Hello, H.; Juutinen, A.; Miettinen, I.T.; et al. Detection and quantification of SARS-CoV-2 RNA in wastewater influent in relation to reported COVID-19 incidence in Finland. medRxiv 2021, 770, 145274. [Google Scholar] [CrossRef]
  17. Ramirez-Castillo, F.Y.; Loera-Muro, A.; Jacques, M.; Garneau, P.; Avelar-Gonzalez, F.J.; Harel, J.; Guerrero-Barrera, A.L. Waterborne pathogens: Detection methods and challenges. Pathogens 2015, 4, 307–334. [Google Scholar] [CrossRef] [PubMed]
  18. Fijalkowski, K.L.; Kacprzak, M.J.; Rorat, A. Occurrence changes of Escherichia coli (including O157:H7 serotype) in wastewater and sewage sludge by quantitation method of (EMA) real time—PCR. Desalination Water Treat. 2014, 52, 3965–3972. [Google Scholar] [CrossRef]
  19. Banting, G.S.; Braithwaite, S.; Scott, C.; Kim, J.; Jeon, B.; Ashbolt, N.; Ruecker, N.; Tymensen, L.; Charest, J.; Pintar, K. Evaluation of various Campylobacter-specific quantitative PCR (qPCR) assays for detection and enumeration of Campylobacteraceae in irrigation water and wastewater via a miniaturized most-probable-number–qPCR assay. Appl. Environ. Microbiol. 2016, 82, 4743–4756. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  20. Teklehaimanot, G.Z.; Genthe, B.; Kamika, I.; Momba, M.N.B. Prevalence of enteropathogenic bacteria in treated effluents and receiving water bodies and their potential health risks. Sci. Total Environ. 2015, 518–519, 441–449. [Google Scholar] [CrossRef] [PubMed]
  21. Teklehaimanot, G.Z.; Coetzee, M.A.A.; Momba, M.N.B. Faecal pollution loads in the wastewater effluents and receiving water bodies: A potential threat to the health of Sedibeng and Soshanguve communities, South Africa. Environ. Sci. Pollut. Res. 2014, 21, 9589–9603. [Google Scholar] [CrossRef]
  22. Koivunen, J.; Siitonen, A.; Heinonen-Tanski, H. Elimination of enteric bacteria in biological–chemical wastewater treatment and tertiary filtration units. Water Res. 2003, 37, 690–698. [Google Scholar] [CrossRef]
  23. Sandhu, B.K.; McBride, S.M. Clostridioides difficile. Trends Microbiol. 2018, 26, 1049–1050. [Google Scholar] [CrossRef]
  24. Xu, C.; Salsali, H.; Weese, S.; Warriner, K. Inactivation of Clostridium difficile in sewage sludge by anaerobic thermophilic digestion. Can. J. Microbiol. 2015, 62, 16–23. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  25. Caicedo, C.; Rosenwinkel, K.H.; Exner, M.; Verstraete, W.; Suchenwirth, R.; Hartemann, P.; Nogueira, R. Legionella occurrence in municipal and industrial wastewater treatment plants and risks of reclaimed wastewater reuse: Review. Water Res. 2019, 149, 21–34. [Google Scholar] [CrossRef] [PubMed]
  26. Vermeulen, L.C.; Brandsema, P.S.; van de Kassteele, J.; Bom, B.C.J.; Sterk, H.A.M.; Sauter, F.J.; van den Berg, H.H.J.L.; de Roda Husman, A.M. Atmospheric dispersion and transmission of Legionella from wastewater treatment plants: A 6-year case-control study. Int. J. Hyg. Environ. Health 2021, 237, 113811. [Google Scholar] [CrossRef] [PubMed]
  27. Radomski, N.; Betelli, L.; Moilleron, R.; Haenn, S.; Moulin, L.; Cambau, E.; Rocher, V.; Gonçalves, A.; Lucas, F.S. Mycobacterium Behavior in Wastewater Treatment Plant, A Bacterial Model Distinct From Escherichia coli and Enterococci. Environ. Sci. Technol. 2011, 45, 5380–5386. [Google Scholar] [CrossRef]
  28. Manning, S.D.; Babcock, H. Escherichia Coli Infections; Chelsea House Publishers: New York, NY, USA, 2010. [Google Scholar]
  29. Levin, R.E. Rapid Detection and Characterization of Foodborne Pathogens by Molecular Techniques; CRC Press: Boca Raton, FL, USA, 2009. [Google Scholar]
  30. Singleton, P.; Sainsbury, D. Dictionary of Microbiology and Molecular Biology; John Wiley & Sons: Hoboken, NJ, USA, 2006. [Google Scholar]
  31. Leblanc-Maridor, M.; Beaudeau, F.; Seegers, H.; Denis, M.; Belloc, C. Rapid identification and quantification of Campylobacter coli and Campylobacter jejuni by real-time PCR in pure cultures and in complex samples. BMC Microbiol. 2011, 11, 113. [Google Scholar] [CrossRef] [Green Version]
  32. Rinsoz, T.; Hilfiker, S.; Oppliger, A. Quantification of Thermotolerant Campylobacter in Swiss Water Treatment Plants, by Real-Time Quantitative Polymerase Chain Reaction. Water Environ. Res. 2009, 81, 929–933. [Google Scholar] [CrossRef] [Green Version]
  33. Moreno, Y.; Botella, S.; Alonso, J.L.; Ferrús, M.A.; Hernández, M.; Hernández, J. Specific detection of Arcobacter and Campylobacter strains in water and sewage by PCR and fluorescent in situ hybridization. Appl. Environ. Microbiol. 2003, 69, 1181–1186. [Google Scholar] [CrossRef] [Green Version]
  34. Igwaran, A.; Okoh, A.I. Human campylobacteriosis: A public health concern of global importance. Heliyon 2019, 5, e02814. [Google Scholar] [CrossRef]
  35. Hansson, I.; Sandberg, M.; Habib, I.; Lowman, R.; Engvall, E.O. Knowledge gaps in control of Campylobacter for prevention of campylobacteriosis. Transbound. Emerg. Dis. 2018, 65, 30–48. [Google Scholar] [CrossRef] [Green Version]
  36. Bolton, D.J. Campylobacter virulence and survival factors. Food Microbiol. 2015, 48, 99–108. [Google Scholar] [CrossRef]
  37. Eucker, T.P.; Konkel, M.E. The cooperative action of bacterial fibronectin-binding proteins and secreted proteins promote maximal Campylobacter jejuni invasion of host cells by stimulating membrane ruffling. Cell. Microbiol. 2012, 14, 226–238. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  38. Carvalho Alexandro, C.T.; Ruiz-Palacios Guillermo, M.; Ramos-Cervantes, P.; Cervantes, L.-E.; Jiang, X.; Pickering Larry, K. Molecular Characterization of Invasive and Noninvasive Campylobacter jejuni and Campylobacter coli Isolates. J. Clin. Microbiol. 2001, 39, 1353–1359. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  39. Mendez-Arancibia, E.; Vargas, M.; Soto, S.; Ruiz, J.; Kahigwa, E.; Schellenberg, D.; Urassa, H.; Gascón, J.; Vila, J. Prevalence of different virulence factors and biofilm production in enteroaggregative Escherichia coli isolates causing diarrhea in children in Ifakara (Tanzania). Am. J. Trop Med. Hyg. 2008, 78, 985–989. [Google Scholar] [CrossRef] [PubMed]
  40. De Boer, R.F.; Ott, A.; Güren, P.; van Zanten, E.; van Belkum, A.; Kooistra-Smid, A.M. Detection of Campylobacter species and Arcobacter butzleri in stool samples by use of real-time multiplex PCR. J. Clin. Microbiol. 2013, 51, 253–259. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  41. Henry, R.; Schang, C.; Chandrasena, G.I.; Deletic, A.; Edmunds, M.; Jovanovic, D.; Kolotelo, P.; Schmidt, J.; Williamson, R.; McCarthy, D. Environmental monitoring of waterborne Campylobacter: Evaluation of the Australian standard and a hybrid extraction-free MPN-PCR method. Front. Microbiol. 2015, 6, 74. [Google Scholar] [CrossRef] [PubMed]
  42. Dar, M.A.; Ahmad, S.M.; Bhat, S.A.; Ahmed, R.; Urwat, U.; Mumtaz, P.T.; Bhat, S.A.; Dar, T.A.; Shah, R.A.; Ganai, N.A. Salmonella typhimurium in poultry: A review. Worlds Poult. Sci. J. 2017, 73, 345–354. [Google Scholar] [CrossRef]
  43. Liu, H.; Whitehouse, C.A.; Li, B. Presence and Persistence of Salmonella in Water: The Impact on Microbial Quality of Water and Food Safety. Front. Public Health 2018, 6, 159. [Google Scholar] [CrossRef]
  44. House, D.; Bishop, A.; Parry, C.; Dougan, G.; Wain, J. Typhoid fever: Pathogenesis and disease. Curr. Opin. Infect. Dis. 2001, 14, 573–578. [Google Scholar] [CrossRef]
  45. Cheng, C.-M.; Doran, T.; Lin, W.; Chen, K.-S.; Williams-Hill, D.; Pamboukian, R. Interlaboratory validation for a real-time PCR Salmonella detection method using the ABI 7500 FAST real-time PCR system. J. Food Prot. 2015, 78, 1119–1124. [Google Scholar] [CrossRef] [PubMed]
  46. González-Escalona, N.; Brown, E.W.; Zhang, G. Development and evaluation of a multiplex real-time PCR (qPCR) assay targeting ttrRSBCA locus and invA gene for accurate detection of Salmonella spp. in fresh produce and eggs. Food Res. Int. 2012, 48, 202–208. [Google Scholar] [CrossRef]
  47. Postollec, F.; Falentin, H.; Pavan, S.; Combrisson, J.; Sohier, D. Recent advances in quantitative PCR (qPCR) applications in food microbiology. Food Microbiol. 2011, 28, 848–861. [Google Scholar] [CrossRef] [PubMed]
  48. Chaudhry, R.; Chandel, D.; Verma, N.; Singh, N.; Singh, P.; Dey, A. Rapid diagnosis of typhoid fever by an in-house flagellin PCR. J. Med. Microbiol. 2010, 59, 1391–1393. [Google Scholar] [CrossRef]
  49. Andrews, J.R.; Ryan, E.T. Diagnostics for invasive Salmonella infections: Current challenges and future directions. Vaccine 2015, 33, C8–C15. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  50. Xu, J.; Suita, K.; Okuno, K.; Takaya, A.; Yamamoto, T.; Isogai, E. Membrane vesicle protein PagC as a novel biomarker for detecting pathogenic Salmonella in the viable but not culturable state. J. Vet. Med. Sci 2018, 80, 133–137. [Google Scholar] [CrossRef] [Green Version]
  51. The, H.C.; Thanh, D.P.; Holt, K.E.; Thomson, N.R.; Baker, S. The genomic signatures of Shigella evolution, adaptation and geographical spread. Nat. Rev. Microbiol. 2016, 14, 235–250. [Google Scholar] [CrossRef] [PubMed]
  52. Murugesan, P.; Revathi, K.; Elayaraja, S.; Vijayalakshmi, S.; Balasubramanian, T. Distribution of enteric bacteria in the sediments of Parangipettai and Cuddalore coast of India. J. Environ. Biol. 2012, 33, 705–711. [Google Scholar] [PubMed]
  53. Binet, R.; Deer, D.M.; Uhlfelder, S.J. Rapid detection of Shigella and enteroinvasive Escherichia coli in produce enrichments by a conventional multiplex PCR assay. Food Microbiol. 2014, 40, 48–54. [Google Scholar] [CrossRef] [Green Version]
  54. Thong, K.L.; Hoe, S.L.; Puthucheary, S.D.; Yasin, R. Detection of virulence genes in Malaysian Shigella species by multiplex PCR assay. BMC Infect. Dis. 2005, 5, 8. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  55. Vantarakis, A.; Komninou, G.; Venieri, D.; Papapetropoulou, M. Development of a multiplex PCR detection of Salmonella spp. and Shigella spp. in mussels. Lett. Appl. Microbiol. 2000, 31, 105–109. [Google Scholar] [CrossRef] [PubMed]
  56. Farfán, M.J.; Garay, T.A.; Prado, C.A.; Filliol, I.; Ulloa, M.T.; Toro, C.S. A new multiplex PCR for differential identification of Shigella flexneri and Shigella sonnei and detection of Shigella virulence determinants. Epidemiol. Infect. 2010, 138, 525–533. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  57. Maheux, A.F.; Bissonnette, L.; Boissinot, M.; Bernier, J.-L.T.; Huppé, V.; Picard, F.J.; Bérubé, È.; Bergeron, M.G. Rapid concentration and molecular enrichment approach for sensitive detection of Escherichia coli and Shigella species in potable water samples. Appl. Environ. Microbiol. 2011, 77, 6199–6207. [Google Scholar] [CrossRef] [Green Version]
  58. Romano, V.; Pasquale, V.; Lemee, L.; El Meouche, I.; Pestel-Caron, M.; Capuano, F.; Buono, P.; Dumontet, S. Clostridioides difficile in the environment, food, animals and humans in southern Italy: Occurrence and genetic relatedness. Comp. Immunol. Microbiol. Infect. Dis. 2018, 59, 41–46. [Google Scholar] [CrossRef] [PubMed]
  59. Numberger, D.; Riedel, T.; McEwen, G.; Nübel, U.; Frentrup, M.; Schober, I.; Bunk, B.; Spröer, C.; Overmann, J.; Grossart, H.-P.; et al. Genomic analysis of three Clostridioides difficile isolates from urban water sources. Anaerobe 2019, 56, 22–26. [Google Scholar] [CrossRef]
  60. Peniche, A.G.; Savidge, T.C.; Dann, S.M. Recent insights into Clostridium difficile pathogenesis. Curr. Opin. Infect. Dis. 2013, 26, 447–453. [Google Scholar] [CrossRef] [PubMed]
  61. Dubberke, E.R.; Haslam, D.B.; Lanzas, C.; Bobo, L.D.; Burnham, C.A.D.; Gröhn, Y.T.; Tarr, P.I. The ecology and pathobiology of Clostridium difficile infections: An interdisciplinary challenge. Zoonoses Public Health 2011, 58, 4–20. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  62. Freeman, J.; Bauer, M.P.; Baines, S.D.; Corver, J.; Fawley, W.N.; Goorhuis, B.; Kuijper, E.J.; Wilcox, M.H. The changing epidemiology of Clostridium difficile infections. Clin. Microbiol. Rev. 2010, 23, 529–549. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  63. Hensgens, M.P.M.; Keessen, E.C.; Squire, M.M.; Riley, T.V.; Koene, M.G.J.; de Boer, E.; Lipman, L.J.A.; Kuijper, E.J. Clostridium difficile infection in the community: A zoonotic disease? Clin. Microbiol. Infect. 2012, 18, 635–645. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  64. O’Grady, K.; Knight, D.R.; Riley, T.V. Antimicrobial resistance in Clostridioides difficile. Eur. J. Clin. Microbiol. Infect. Dis. 2021, 40, 2459–2478. [Google Scholar] [CrossRef] [PubMed]
  65. Nikaeen, M.; Aghili Dehnavi, H.; Hssanzadeh, A.; Jalali, M. Occurrence of Clostridium difficile in two types of wastewater treatment plants. J. Formos. Med. Assoc. 2015, 114, 663–665. [Google Scholar] [CrossRef] [Green Version]
  66. Blasi, F.; Lovito, C.; Albini, E.; Bano, L.; Dalmonte, G.; Drigo, I.; Maresca, C.; Massacci, F.R.; Orsini, S.; Primavilla, S.; et al. Clostridioides difficile in Calves in Central Italy: Prevalence, Molecular Typing, Antimicrobial Susceptibility and Association with Antibiotic Administration. Animals 2021, 11, 515. [Google Scholar] [CrossRef]
  67. Caicedo, C.; Beutel, S.; Scheper, T.; Rosenwinkel, K.H.; Nogueira, R. Occurrence of Legionella in wastewater treatment plants linked to wastewater characteristics. Environ. Sci. Pollut. Res. 2016, 23, 16873–16881. [Google Scholar] [CrossRef] [PubMed]
  68. Guo, F.; Zhang, T.; Li, B.; Wang, Z.; Ju, F.; Liang, Y.-T. Mycobacterial species and their contribution to cholesterol degradation in wastewater treatment plants. Sci. Rep. 2019, 9, 836. [Google Scholar] [CrossRef] [Green Version]
  69. Bierque, E.; Thibeaux, R.; Girault, D.; Soupé-Gilbert, M.-E.; Goarant, C. A systematic review of Leptospira in water and soil environments. PLoS ONE 2020, 15, e0227055. [Google Scholar] [CrossRef] [PubMed]
  70. Leslie, E.; Hinds, J.; Hai, F.I. Causes, Factors, and Control Measures of Opportunistic Premise Plumbing Pathogens—A Critical Review. Appl. Sci. 2021, 11, 4474. [Google Scholar] [CrossRef]
  71. Muder, R.R.; Victor, L.Y. Infection due to Legionella species other than L. pneumophila. Clin. Infect. Dis. 2002, 35, 990–998. [Google Scholar] [CrossRef] [Green Version]
  72. Chauhan, D.; Shames, S.R. Pathogenicity and Virulence of Legionella: Intracellular replication and host response. Virulence 2021, 12, 1122–1144. [Google Scholar] [CrossRef]
  73. Sabrià, M.; Pedro-Botet, M.L.; Gómez, J.; Roig, J.; Vilaseca, B.; Sopena, N.; Baños, V. Fluoroquinolones vs macrolides in the treatment of Legionnaires disease. Chest 2005, 128, 1401–1405. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  74. Postma, D.F.; Van Werkhoven, C.H.; Van Elden, L.J.; Thijsen, S.F.; Hoepelman, A.I.; Kluytmans, J.A.; Boersma, W.G.; Compaijen, C.J.; Van Der Wall, E.; Prins, J.M. Antibiotic treatment strategies for community-acquired pneumonia in adults. N. Engl. J. Med. 2015, 372, 1312–1323. [Google Scholar] [CrossRef] [Green Version]
  75. Garin, N.; Genné, D.; Carballo, S.; Chuard, C.; Eich, G.; Hugli, O.; Lamy, O.; Nendaz, M.; Petignat, P.-A.; Perneger, T. β-Lactam monotherapy vs β-lactam–macrolide combination treatment in moderately severe community-acquired pneumonia: A randomized noninferiority trial. JAMA Intern. Med. 2014, 174, 1894–1901. [Google Scholar] [CrossRef] [Green Version]
  76. Johnson, M.M.; Odell, J.A. Nontuberculous mycobacterial pulmonary infections. J. Thorac. Dis 2014, 6, 210–220. [Google Scholar] [CrossRef] [PubMed]
  77. Pontiroli, A.; Khera, T.T.; Oakley, B.B.; Mason, S.; Dowd, S.E.; Travis, E.R.; Erenso, G.; Aseffa, A.; Courtenay, O.; Wellington, E.M.H. Prospecting Environmental Mycobacteria: Combined Molecular Approaches Reveal Unprecedented Diversity. PLoS ONE 2013, 8, e68648. [Google Scholar] [CrossRef] [Green Version]
  78. Chakaya, J.; Khan, M.; Ntoumi, F.; Aklillu, E.; Fatima, R.; Mwaba, P.; Kapata, N.; Mfinanga, S.; Hasnain, S.E.; Katoto, P.; et al. Global Tuberculosis Report 2020—Reflections on the Global TB burden, treatment and prevention efforts. Int. J. Infect. Dis. 2021, 6, 1201–9712. [Google Scholar] [CrossRef] [PubMed]
  79. Rizzo, L.; Manaia, C.; Merlin, C.; Schwartz, T.; Dagot, C.; Ploy, M.C.; Michael, I.; Fatta-Kassinos, D. Urban wastewater treatment plants as hotspots for antibiotic resistant bacteria and genes spread into the environment: A review. Sci. Total Environ. 2013, 447, 345–360. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  80. Hyun, B.; Cha, H.-G.; Lee, N.; Yum, S.; Baek, S.H.; Shin, K. Development of an ATP assay for rapid onboard testing to detect living microorganisms in ballast water. J. Sea Res. 2018, 133, 73–80. [Google Scholar] [CrossRef]
  81. Zhang, Y.; Liu, W.-T. The application of molecular tools to study the drinking water microbiome—Current understanding and future needs. Crit. Rev. Environ. Sci. Technol. 2019, 49, 1188–1235. [Google Scholar] [CrossRef] [Green Version]
  82. Liu, Q.; Wu, J.e.; Lim, Z.Y.; Aggarwal, A.; Yang, H.; Wang, S. Evaluation of the metabolic response of Escherichia coli to electrolysed water by 1H NMR spectroscopy. LWT-Food Sci. Technol. 2017, 79, 428–436. [Google Scholar] [CrossRef]
  83. Slavov, S.; Alusta, P.; Buzatu, D.A.; Wilkes, J.G. Feature selection from mass spectra of bacteria for serotyping Salmonella. J. Anal. Appl. Pyrolysis 2017, 124, 393–402. [Google Scholar] [CrossRef]
  84. Zhang, Y.; Lai, B.S.; Juhas, M. Recent Advances in Aptamer Discovery and Applications. Molecules 2019, 24, 941. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  85. Zou, Y.; Duan, N.; Wu, S.; Shen, M.; Wang, Z. Selection, Identification, and Binding Mechanism Studies of an ssDNA Aptamer Targeted to Different Stages of E. coli O157:H7. J. Agric. Food Chem. 2018, 66, 5677–5682. [Google Scholar] [CrossRef] [PubMed]
  86. Yu, X.; Chen, F.; Wang, R.; Li, Y. Whole-bacterium SELEX of DNA aptamers for rapid detection of E.coli O157:H7 using a QCM sensor. J. Biotechnol. 2018, 266, 39–49. [Google Scholar] [CrossRef] [PubMed]
  87. Buss, J.E.; Cresse, M.; Doyle, S.; Buchan, B.W.; Craft, D.W.; Young, S. Campylobacter culture fails to correctly detect Campylobacter in 30% of positive patient stool specimens compared to non-cultural methods. Eur. J. Clin. Microbiol. Infect. Dis. 2019, 38, 1087–1093. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  88. Ormen, O.; Aalberg, K.; Madslien, E.H. Multiplex polymerase chain reaction detection of enteropathogens in sewage in Norway. Acta Vet. Scand. 2019, 61, 11. [Google Scholar] [CrossRef] [PubMed]
  89. Yanagimoto, K.; Yamagami, T.; Uematsu, K.; Haramoto, E. Characterization of Salmonella Isolates from Wastewater Treatment Plant Influents to Estimate Unreported Cases and Infection Sources of Salmonellosis. Pathogens 2020, 9, 52. [Google Scholar] [CrossRef] [Green Version]
  90. Kasturi, K.N.; Drgon, T. Real-Time PCR Method for Detection of Salmonella spp. in Environmental Samples. Appl. Environ. Microbiol. 2017, 83, e00644-17. [Google Scholar] [CrossRef] [Green Version]
  91. Lekunberri, I.; Balcázar, J.L.; Borrego, C.M. Detection and quantification of the plasmid-mediated mcr-1 gene conferring colistin resistance in wastewater. Int. J. Antimicrob. Agents 2017, 50, 734–736. [Google Scholar] [CrossRef] [PubMed]
  92. Sandberg, K.D.; Ishii, S.; LaPara, T.M. A Microfluidic Quantitative Polymerase Chain Reaction Method for the Simultaneous Analysis of Dozens of Antibiotic Resistance and Heavy Metal Resistance Genes. Environ. Sci. Technol. Lett. 2018, 5, 20–25. [Google Scholar] [CrossRef]
  93. Pan, H.; Dong, K.; Rao, L.; Zhao, L.; Wu, X.; Wang, Y.; Liao, X. Quantitative detection of viable but nonculturable state Escherichia coli O157:H7 by ddPCR combined with propidium monoazide. Food Control. 2020, 112, 107140. [Google Scholar] [CrossRef]
  94. Li, Y. Establishment and Application of a Visual DNA Microarray for the Detection of Food-borne Pathogens. Anal. Sci. 2016, 32, 215–218. [Google Scholar] [CrossRef] [Green Version]
  95. Lee, J.-E.; Mun, H.; Kim, S.-R.; Kim, M.-G.; Chang, J.-Y.; Shim, W.-B. A colorimetric Loop-mediated isothermal amplification (LAMP) assay based on HRP-mimicking molecular beacon for the rapid detection of Vibrio parahaemolyticus. Biosens. Bioelectron. 2020, 151, 111968. [Google Scholar] [CrossRef] [PubMed]
  96. Salimi, G.; Mousavi, Z.E.; Kiani, H. Efficiency of fluorescence in situ hybridization (FISH) method for the rapid detection of Salmonella in minced lamb meat: Method analysis and optimization. J. Microbiol. Methods 2020, 175, 105989. [Google Scholar] [CrossRef] [PubMed]
  97. Lyu, J.; Zhang, J.; Ren, X. Detection and identification of bacterial pathogens directly from sputum samples by pyrosequencing. J. Med. Microbiol. 2019, 68, 368–373. [Google Scholar] [CrossRef] [PubMed]
  98. Winand, R.; Bogaerts, B.; Hoffman, S.; Lefevre, L.; Delvoye, M.; Van Braekel, J.; Fu, Q.; Roosens, N.H.; De Keersmaecker, S.C.; Vanneste, K. Targeting the 16s rRNA gene for bacterial identification in complex mixed samples: Comparative evaluation of second (illumina) and third (oxford nanopore technologies) generation sequencing technologies. Int. J. Mol. Sci. 2020, 21, 298. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  99. Su, J.Q.; An, X.L.; Li, B.; Chen, Q.L.; Gillings, M.R.; Chen, H.; Zhang, T.; Zhu, Y.G. Metagenomics of urban sewage identifies an extensively shared antibiotic resistome in China. Microbiome 2017, 5, 84. [Google Scholar] [CrossRef]
  100. Park, S.; Kim, Y.T.; Kim, Y.-K. Optical enzyme-linked immunosorbent assay on a strip for detection of Salmonella typhimurium. BioChip J. 2010, 4, 110–116. [Google Scholar] [CrossRef]
  101. Yang, X.; Feng, L.; Qin, X. Preparation of the Cf-GQDs-Escherichia coli O157: H7 bioprobe and its application in optical imaging and sensing of Escherichia coli O157: H7. Food Anal. Methods 2018, 11, 2280–2286. [Google Scholar] [CrossRef]
  102. Sun, Y.; Chang, Y.; Zhang, Q.; Liu, M. An Origami Paper-Based Device Printed with DNAzyme-Containing DNA Superstructures for Escherichia coli Detection. Micromachines 2019, 10, 531. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  103. Khan, I.U.; Gannon, V.; Kent, R.; Koning, W.; Lapen, D.R.; Miller, J.; Neumann, N.; Phillips, R.; Robertson, W.; Topp, E. Development of a rapid quantitative PCR assay for direct detection and quantification of culturable and non-culturable Escherichia coli from agriculture watersheds. J. Microbiol. Methods 2007, 69, 480–488. [Google Scholar] [CrossRef]
  104. Maheux, A.F.; Bérubé, È.; Boudreau, D.K.; Villéger, R.; Cantin, P.; Boissinot, M.; Bissonnette, L.; Bergeron, M.G. Abilities of the mCP agar method and CRENAME alpha toxin-specific real-time PCR assay to detect Clostridium perfringens spores in drinking water. Appl. Environ. Microbiol. 2013, 79, 7654–7661. [Google Scholar] [CrossRef] [Green Version]
  105. Ram, S.; Vajpayee, P.; Shanker, R. Rapid culture-independent quantitative detection of enterotoxigenic Escherichia coli in surface waters by real-time PCR with molecular beacon. Environ. Sci. Technol. 2008, 42, 4577–4582. [Google Scholar] [CrossRef]
  106. Kralik, P.; Ricchi, M. A Basic Guide to Real Time PCR in Microbial Diagnostics: Definitions, Parameters, and Everything. Front. Microbiol 2017, 8, 108. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  107. Omar, K.; Barnard, T. Detection of diarrhoeagenic Escherichia coli in clinical and environmental water sources in South Africa using single-step 11-gene m-PCR. World J. Microbiol. Biotechnol. 2014, 30, 2663–2671. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  108. Racki, N.; Dreo, T.; Gutierrez-Aguirre, I.; Blejec, A.; Ravnikar, M. Reverse transcriptase droplet digital PCR shows high resilience to PCR inhibitors from plant, soil and water samples. Plant. Methods 2014, 10, 42. [Google Scholar] [CrossRef] [PubMed]
  109. Taylor, S.C.; Carbonneau, J.; Shelton, D.N.; Boivin, G. Optimization of Droplet Digital PCR from RNA and DNA extracts with direct comparison to RT-qPCR: Clinical implications for quantification of Oseltamivir-resistant subpopulations. J. Virol. Methods 2015, 224, 58–66. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  110. Devonshire, A.S.; Sanders, R.; Whale, A.S.; Nixon, G.J.; Cowen, S.; Ellison, S.L.; Parkes, H.; Pine, P.S.; Salit, M.; McDaniel, J.; et al. An international comparability study on quantification of mRNA gene expression ratios: CCQM-P103.1. Biomol. Detect. Quantif. 2016, 8, 15–28. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  111. Kokkoris, V.; Vukicevich, E.; Richards, A.; Thomsen, C.; Hart, M.M. Challenges Using Droplet Digital PCR for Environmental Samples. Appl. Microbiol. 2021, 1, 74–88. [Google Scholar] [CrossRef]
  112. Singh, G.; Sithebe, A.; Enitan, A.M.; Kumari, S.; Bux, F.; Stenstrom, T.A. Comparison of droplet digital PCR and quantitative PCR for the detection of Salmonella and its application for river sediments. J. Water Health 2017, 15, 505–508. [Google Scholar] [CrossRef]
  113. Yang, J.; Zhang, N.; Lv, J.; Zhu, P.; Pan, X.; Hu, J.; Wu, W.; Li, S.; Li, H. Comparing the performance of conventional PCR, RTQ-PCR, and droplet digital PCR assays in detection of Shigella. Mol. Cell. Probes 2020, 51, 101531. [Google Scholar] [CrossRef] [PubMed]
  114. Gosiewski, T.; Flis, A.; Sroka, A.; Kędzierska, A.; Pietrzyk, A.; Kędzierska, J.; Drwiła, R.; Bulanda, M. Comparison of nested, multiplex, qPCR.; FISH.; SeptiFast and blood culture methods in detection and identification of bacteria and fungi in blood of patients with sepsis. BMC Microbiol. 2014, 14, 313. [Google Scholar] [CrossRef] [Green Version]
  115. Ibekwe, A.M.; Watt, P.M.; Grieve, C.M.; Sharma, V.K.; Lyons, S.R. Multiplex fluorogenic real-time PCR for detection and quantification of Escherichia coli O157:H7 in dairy wastewater wetlands. Appl Env. Microbiol 2002, 68, 4853–4862. [Google Scholar] [CrossRef] [Green Version]
  116. LaGier, M.J.; Joseph, L.A.; Passaretti, T.V.; Musser, K.A.; Cirino, N.M. A real-time multiplexed PCR assay for rapid detection and differentiation of Campylobacter jejuni and Campylobacter coli. Mol. Cell. Probes 2004, 18, 275–282. [Google Scholar] [CrossRef]
  117. Reischer, G.H.; Kasper, D.C.; Steinborn, R.; Farnleitner, A.H.; Mach, R.L. A quantitative real-time PCR assay for the highly sensitive and specific detection of human faecal influence in spring water from a large alpine catchment area. Lett. Appl. Microbiol. 2007, 44, 351–356. [Google Scholar] [CrossRef]
  118. Nayak, A.; Rose, J. Detection of Helicobacter pylori in sewage and water using a new quantitative PCR method with SYBR® green. J. Appl. Microbiol. 2007, 103, 1931–1941. [Google Scholar] [CrossRef] [PubMed]
  119. Wery, N.; Lhoutellier, C.; Ducray, F.; Delgenes, J.-P.; Godon, J.-J. Behaviour of pathogenic and indicator bacteria during urban wastewater treatment and sludge composting, as revealed by quantitative PCR. Water Res. 2008, 42, 53–62. [Google Scholar] [CrossRef] [PubMed]
  120. Haugland, R.A.; Siefring, S.C.; Wymer, L.J.; Brenner, K.P.; Dufour, A.P. Comparison of Enterococcus measurements in freshwater at two recreational beaches by quantitative polymerase chain reaction and membrane filter culture analysis. Water Res. 2005, 39, 559–568. [Google Scholar] [CrossRef] [PubMed]
  121. Baeumner, A.J.; Humiston, M.C.; Montagna, R.A.; Durst, R.A. Detection of viable oocysts of Cryptosporidium parvum following nucleic acid sequence based amplification. Anal. Chem. 2001, 73, 1176–1180. [Google Scholar] [CrossRef] [PubMed]
  122. Pitkänen, T.; Ryu, H.; Elk, M.; Hokajärvi, A.-M.; Siponen, S.; Vepsäläinen, A.; Räsänen, P.; Santo Domingo, J.W. Detection of Fecal Bacteria and Source Tracking Identifiers in Environmental Waters Using rRNA-Based RT-qPCR and rDNA-Based qPCR Assays. Environ. Sci. Technol. 2013, 47, 13611–13620. [Google Scholar] [CrossRef] [PubMed]
  123. Tiwari, A.; Niemelä, S.I.; Vepsäläinen, A.; Rapala, J.; Kalso, S.; Pitkänen, T. Comparison of Colilert-18 with miniaturised most probable number method for monitoring of Escherichia coli in bathing water. J. Water Health 2015, 14, 121–131. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  124. Inkinen, J.; Siponen, S.; Jayaprakash, B.; Tiwari, A.; Hokajärvi, A.-M.; Pursiainen, A.; Ikonen, J.; Kauppinen, A.; Miettinen, I.T.; Paananen, J.; et al. Diverse and active archaea communities occur in non-disinfected drinking water systems-Less activity revealed in disinfected and hot water systems. Water Res. X 2021, 12, 100101. [Google Scholar] [CrossRef]
  125. Fumian, T.M.; Leite, J.P.G.; Castello, A.A.; Gaggero, A.; Caillou, M.S.L.d.; Miagostovich, M.P. Detection of rotavirus A in sewage samples using multiplex qPCR and an evaluation of the ultracentrifugation and adsorption-elution methods for virus concentration. J. Virol. Methods 2010, 170, 42–46. [Google Scholar] [CrossRef]
  126. Al-Tebrineh, J.; Pearson, L.A.; Yasar, S.A.; Neilan, B.A. A multiplex qPCR targeting hepato- and neurotoxigenic cyanobacteria of global significance. Harmful Algae 2012, 15, 19–25. [Google Scholar] [CrossRef]
  127. Kamau, E.; Alemayehu, S.; Feghali, K.C.; Saunders, D.; Ockenhouse, C.F. Multiplex qPCR for detection and absolute quantification of malaria. PLoS ONE 2013, 8, e71539. [Google Scholar] [CrossRef] [Green Version]
  128. Bumgarner, R. Overview of DNA microarrays: Types, applications, and their future. Curr. Protoc. Mol. Biol. 2013, 101, 22-1. [Google Scholar] [CrossRef] [Green Version]
  129. Inoue, D.; Hinoura, T.; Suzuki, N.; Pang, J.; Malla, R.; Shrestha, S.; Chapagain, S.K.; Matsuzawa, H.; Nakamura, T.; Tanaka, Y. High-throughput DNA microarray detection of pathogenic bacteria in shallow well groundwater in the Kathmandu Valley, Nepal. Curr. Microbiol. 2015, 70, 43–50. [Google Scholar] [CrossRef] [PubMed]
  130. Lemuth, K.; Rupp, S. Microarrays as Research Tools and Diagnostic Devices. In RNA and DNA Diagnostics; Springer: Cham, Switzerland, 2015; pp. 259–280. [Google Scholar] [CrossRef]
  131. Cristescu, M.E.; Hebert, P.D.N. Uses and Misuses of Environmental DNA in Biodiversity Science and Conservation. Annu. Rev. Ecol. Evol. Syst. 2018, 49, 209–230. [Google Scholar] [CrossRef] [Green Version]
  132. Sheet, O.; Grabowski, N.; Klein, G.; Abdulmawjood, A. Development and validation of a loop mediated isothermal amplification (LAMP) assay for the detection of Staphylococcus aureus in bovine mastitis milk samples. Mol. Cell. Probes 2016, 30, 320–325. [Google Scholar] [CrossRef] [PubMed]
  133. Niu, J.H.; Jian, H.; Guo, Q.X.; Chen, C.L.; Wang, X.Y.; Liu, Q.; Guo, Y.D. Evaluation of loop-mediated isothermal amplification (LAMP) assays based on 5S rDNA-IGS2 regions for detecting Meloidogyne enterolobii. Plant. Pathol. 2012, 61, 809–819. [Google Scholar] [CrossRef]
  134. Nzelu, C.O.; Cáceres, A.G.; Guerrero-Quincho, S.; Tineo-Villafuerte, E.; Rodriquez-Delfin, L.; Mimori, T.; Uezato, H.; Katakura, K.; Gomez, E.A.; Guevara, A.G. A rapid molecular diagnosis of cutaneous leishmaniasis by colorimetric malachite green-loop-mediated isothermal amplification (LAMP) combined with an FTA card as a direct sampling tool. Acta Trop. 2016, 153, 116–119. [Google Scholar] [CrossRef]
  135. Amann, R.; Fuchs, B.M. Single-cell identification in microbial communities by improved fluorescence in situ hybridization techniques. Nat. Rev. Microbiol. 2008, 6, 339–348. [Google Scholar] [CrossRef]
  136. Lukumbuzya, M.; Schmid, M.; Pjevac, P.; Daims, H. A Multicolor Fluorescence in situ Hybridization Approach Using an Extended Set of Fluorophores to Visualize Microorganisms. Front. Microbiol. 2019, 10, 1383. [Google Scholar] [CrossRef]
  137. Santiago, P.; Jiménez-Belenguer, A.; García-Hernández, J.; Estellés, R.M.; Hernández Pérez, M.; Castillo López, M.A.; Ferrús, M.A.; Moreno, Y. High prevalence of Salmonella spp. in wastewater reused for irrigation assessed by molecular methods. Int. J. Hyg. Environ. Health 2018, 221, 95–101. [Google Scholar] [CrossRef] [PubMed]
  138. Gwinn, M.; MacCannell, D.; Armstrong, G.L. Next-Generation Sequencing of Infectious Pathogens. JAMA 2019, 321, 893–894. [Google Scholar] [CrossRef] [Green Version]
  139. Behjati, S.; Tarpey, P.S. What is next generation sequencing? Arch. Dis. Child. Educ. Pract. 2013, 98, 236–238. [Google Scholar] [CrossRef]
  140. Treangen, T.J.; Salzberg, S.L. Repetitive DNA and next-generation sequencing: Computational challenges and solutions. Nat. Rev. Genet. 2012, 13, 36–46. [Google Scholar] [CrossRef] [PubMed]
  141. Zhu, X.; Yan, S.; Yuan, F.; Wan, S. The Applications of Nanopore Sequencing Technology in Pathogenic Microorganism Detection. Can. J. Infect. Dis. Med. Microbiol. 2020, 2020, 6675206. [Google Scholar] [CrossRef]
  142. Greninger, A.L.; Naccache, S.N.; Federman, S.; Yu, G.; Mbala, P.; Bres, V.; Stryke, D.; Bouquet, J.; Somasekar, S.; Linnen, J.M.; et al. Rapid metagenomic identification of viral pathogens in clinical samples by real-time nanopore sequencing analysis. Genome Med. 2015, 7, 99. [Google Scholar] [CrossRef] [Green Version]
  143. Schmidt, M.H.W.; Vogel, A.; Denton, A.K.; Istace, B.; Wormit, A.; van de Geest, H.; Bolger, M.E.; Alseekh, S.; Maß, J.; Pfaff, C.; et al. De Novo Assembly of a New Solanum pennellii Accession Using Nanopore Sequencing. Plant. Cell 2017, 29, 2336–2348. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  144. Cao, M.D.; Ganesamoorthy, D.; Elliott, A.G.; Zhang, H.; Cooper, M.A.; Coin, L.J.M. Streaming algorithms for identification of pathogens and antibiotic resistance potential from real-time MinION(TM) sequencing. Gigascience 2016, 5, 32. [Google Scholar] [CrossRef] [Green Version]
  145. Ammar, R.; Paton, T.A.; Torti, D.; Shlien, A.; Bader, G.D. Long read nanopore sequencing for detection of HLA and CYP2D6 variants and haplotypes. F1000Res 2015, 4, 17. [Google Scholar] [CrossRef] [Green Version]
  146. De Siqueira, G.M.V.; Pereira-Dos-Santos, F.M.; Silva-Rocha, R.; Guazzaroni, M.-E. Nanopore Sequencing Provides Rapid and Reliable Insight Into Microbial Profiles of Intensive Care Units. Front. Public Health 2021, 9, 710985. [Google Scholar] [CrossRef]
  147. Wattam, A.R.; Inzana, T.J.; Williams, K.P.; Mane, S.P.; Shukla, M.; Almeida, N.F.; Dickerman, A.W.; Mason, S.; Moriyón, I.; O’Callaghan, D.; et al. Comparative genomics of early-diverging Brucella strains reveals a novel lipopolysaccharide biosynthesis pathway. mBio 2012, 3, e00246-12. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  148. Widmer, A.F.; Frei, R.; Kuijper, J.; Wilcox, M.H.; Schindler, R.; Spaniol, V.; Goldenberger, D.; Egli, A.; Tschudin-Sutter, S. Multicenter Prevalence Study Comparing Molecular and Toxin Assays for Clostridioides difficile Surveillance, Switzerland. Emerg. Infect. Dis. 2020, 26, 2370. [Google Scholar] [CrossRef] [PubMed]
  149. Mao, K.; Zhang, H.; Pan, Y.; Yang, Z. Biosensors for wastewater-based epidemiology for monitoring public health. Water Res. 2021, 191, 116787. [Google Scholar] [CrossRef] [PubMed]
  150. Yildirim, N.; Long, F.; Gu, A.Z. Aptamer based E. coli detection in waste waters by portable optical biosensor system. In Proceedings of the 40th Annual Northeast Bioengineering Conference (NEBEC), Boston, MA, USA, 25–27 April 2014; pp. 1–3. [Google Scholar]
  151. Doria, G.; Conde, J.; Veigas, B.; Giestas, L.; Almeida, C.; Assunção, M.; Rosa, J.; Baptista, P.V. Noble metal nanoparticles for biosensing applications. Sensors 2012, 12, 1657–1687. [Google Scholar] [CrossRef] [PubMed]
  152. Jyoti, A.; Tomar, R.S.; Shanker, R. Nanosensors for the detection of pathogenic bacteria. In Nanoscience in Food and Agriculture 1; Springer: Cham, Switzerland, 2016; pp. 129–150. [Google Scholar]
  153. Ahmed, A.; Rushworth, J.V.; Hirst, N.A.; Millner, P.A. Biosensors for whole-cell bacterial detection. Clin. Microbiol. Rev. 2014, 27, 631–646. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  154. Choi, J.R.; Yong, K.W.; Tang, R.; Gong, Y.; Wen, T.; Li, F.; Pingguan-Murphy, B.; Bai, D.; Xu, F. Advances and challenges of fully integrated paper-based point-of-care nucleic acid testing. TrAC Trends Anal. Chem. 2017, 93, 37–50. [Google Scholar] [CrossRef]
  155. Suea-Ngam, A.; Choopara, I.; Li, S.; Schmelcher, M.; Somboonna, N.; Howes, P.D.; deMello, A.J. In Situ Nucleic Acid Amplification and Ultrasensitive Colorimetric Readout in a Paper-Based Analytical Device Using Silver Nanoplates. Adv. Healthc. Mater. 2021, 10, 2001755. [Google Scholar] [CrossRef]
  156. Mao, K.; Zhang, H.; Yang, Z. Can a Paper-Based Device Trace COVID-19 Sources with Wastewater-Based Epidemiology? Environ. Sci. Technol. 2020, 54, 3733–3735. [Google Scholar] [CrossRef]
  157. Boehle, K.E.; Gilliand, J.; Wheeldon, C.R.; Holder, A.; Adkins, J.A.; Geiss, B.J.; Ryan, E.P.; Henry, C.S. Utilizing Paper-Based Devices for Antimicrobial-Resistant Bacteria Detection. Angew. Chem. Int. Ed. 2017, 56, 6886–6890. [Google Scholar] [CrossRef]
  158. Nishat, S.; Jafry, A.T.; Martinez, A.W.; Awan, F.R. Paper-based microfluidics: Simplified fabrication and assay methods. Sens. Actuators B Chem. 2021, 336, 129681. [Google Scholar] [CrossRef]
  159. Ganaja, K.A.; Chaplan, C.A.; Zhang, J.; Martinez, N.W.; Martinez, A.W. Paper Microzone Plates as Analytical Tools for Studying Enzyme Stability: A Case Study on the Stabilization of Horseradish Peroxidase Using Trehalose and SU-8 Epoxy Novolac Resin. Anal. Chem. 2017, 89, 5333–5341. [Google Scholar] [CrossRef] [Green Version]
  160. Daughton, C.G. Illicit Drugs in Municipal Sewage. In Pharmaceuticals and Care Products in the Environment; American Chemical Society: Washington, DC, USA, 2001; pp. 348–364. [Google Scholar]
  161. Aarestrup, F.M.; Woolhouse, M.E.J. Using sewage for surveillance of antimicrobial resistance. Science 2020, 367, 630–632. [Google Scholar] [CrossRef] [PubMed]
  162. Hellmer, M.; Paxeus, N.; Magnius, L.; Enache, L.; Arnholm, B.; Johansson, A.; Bergstrom, T.; Norder, H. Detection of pathogenic viruses in sewage provided early warnings of hepatitis A virus and norovirus outbreaks. Appl. Env. Microbiol. 2014, 80, 6771–6781. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  163. Park, S.H.; Hanning, I.; Jarquin, R.; Moore, P.; Donoghue, D.J.; Donoghue, A.M.; Ricke, S.C. Multiplex PCR assay for the detection and quantification of Campylobacter spp., Escherichia coli O157:H7, and Salmonella serotypes in water samples. FEMS Microbiol. Lett. 2011, 316, 7–15. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  164. Marangi, M.; Giangaspero, A.; Lacasella, V.; Lonigro, A.; Gasser, R.B. Multiplex PCR for the detection and quantification of zoonotic taxa of Giardia, Cryptosporidium and Toxoplasma in wastewater and mussels. Mol. Cell. Probes 2015, 29, 122–125. [Google Scholar] [CrossRef] [PubMed]
  165. An, X.L.; Su, J.Q.; Li, B.; Ouyang, W.Y.; Zhao, Y.; Chen, Q.L.; Cui, L.; Chen, H.; Gillings, M.R.; Zhang, T.; et al. Tracking antibiotic resistome during wastewater treatment using high throughput quantitative PCR. Environ. Int. 2018, 117, 146–153. [Google Scholar] [CrossRef] [PubMed]
  166. Karkman, A.; Johnson, T.A.; Lyra, C.; Stedtfeld, R.D.; Tamminen, M.; Tiedje, J.M.; Virta, M. High-throughput quantification of antibiotic resistance genes from an urban wastewater treatment plant. FEMS Microbiol. Ecol. 2016, 92, fiw01. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  167. Urbaniak, C.; Sielaff, A.C.; Frey, K.G.; Allen, J.E.; Singh, N.; Jaing, C.; Wheeler, K.; Venkateswaran, K. Detection of antimicrobial resistance genes associated with the International Space Station environmental surfaces. Sci. Rep. 2018, 8, 814. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  168. Mumy, K.L.; Findlay, R.H. Convenient determination of DNA extraction efficiency using an external DNA recovery standard and quantitative-competitive PCR. J. Microbiol. Methods 2004, 57, 259–268. [Google Scholar] [CrossRef] [PubMed]
  169. Gyawali, P.; Ahmed, W.; Jagals, P.; Sidhu, J.P.S.; Toze, S. Comparison of concentration methods for rapid detection of hookworm ova in wastewater matrices using quantitative PCR. Exp. Parasitol. 2015, 159, 160–167. [Google Scholar] [CrossRef] [Green Version]
  170. Lemarchand, K.; Berthiaume, F.; Maynard, C.; Harel, J.; Payment, P.; Bayardelle, P.; Masson, L.; Brousseau, R. Optimization of microbial DNA extraction and purification from raw wastewater samples for downstream pathogen detection by microarrays. J. Microbiol. Methods 2005, 63, 115–126. [Google Scholar] [CrossRef]
  171. Li, A.-D.; Metch, J.W.; Wang, Y.; Garner, E.; Zhang, A.N.; Riquelme, M.V.; Vikesland, P.J.; Pruden, A.; Zhang, T. Effects of sample preservation and DNA extraction on enumeration of antibiotic resistance genes in wastewater. FEMS Microbiol. Ecol. 2017, 94, fix189. [Google Scholar] [CrossRef] [PubMed]
  172. Walden, C.; Carbonero, F.; Zhang, W. Assessing impacts of DNA extraction methods on next generation sequencing of water and wastewater samples. J. Microbiol. Methods 2017, 141, 10–16. [Google Scholar] [CrossRef] [PubMed]
  173. Bonetta, S.; Pignata, C.; Lorenzi, E.; De Ceglia, M.; Meucci, L.; Bonetta, S.; Gilli, G.; Carraro, E. Detection of pathogenic Campylobacter, E. coli O157:H7 and Salmonella spp. in wastewater by PCR assay. Environ. Sci. Pollut. Res. 2016, 23, 15302–15309. [Google Scholar] [CrossRef] [PubMed]
  174. Kibbee, R.J.; Örmeci, B. Development of a sensitive and false-positive free PMA-qPCR viability assay to quantify VBNC Escherichia coli and evaluate disinfection performance in wastewater effluent. J. Microbiol. Methods 2017, 132, 139–147. [Google Scholar] [CrossRef] [PubMed]
  175. Baccari, O.; Elleuch, J.; Barkallah, M.; Boukedi, H.; Ayed, N.B.; Hammami, A.; Fendri, I.; Abdelkafi, S. Development of a new TaqMan-based PCR assay for the specific detection and quantification of Simkania negevensis. Mol. Cell. Probes 2020, 53, 101645. [Google Scholar] [CrossRef]
  176. Liu, P.; Ibaraki, M.; Kapoor, R.; Amin, N.; Das, A.; Miah, R.; Mukhopadhyay, A.K.; Rahman, M.; Dutta, S.; Moe, C.L. Development of Moore Swab and Ultrafiltration Concentration and Detection Methods for Salmonella Typhi and Salmonella Paratyphi A in Wastewater and Application in Kolkata, India and Dhaka, Bangladesh. Front. Microbiol. 2021, 12, 684094. [Google Scholar] [CrossRef]
  177. Jikumaru, A.; Ishii, S.; Fukudome, T.; Kawahara, Y.; Iguchi, A.; Masago, Y.; Nukazawa, K.; Suzuki, Y. Fast, sensitive, and reliable detection of waterborne pathogens by digital PCR after coagulation and foam concentration. J. Biosci. Bioeng. 2020, 130, 76–81. [Google Scholar] [CrossRef] [PubMed]
  178. Pärnänen, K.M.M.; Narciso-da-Rocha, C.; Kneis, D.; Berendonk, T.U.; Cacace, D.; Do, T.T.; Elpers, C.; Fatta-Kassinos, D.; Henriques, I.; Jaeger, T.; et al. Antibiotic resistance in European wastewater treatment plants mirrors the pattern of clinical antibiotic resistance prevalence. Sci. Adv. 2019, 5, eaau9124. [Google Scholar] [CrossRef] [Green Version]
  179. Nathaniel, B.R.; Ghai, M.; Druce, M.; Maharaj, I.; Olaniran, A.O. Development of a loop-mediated isothermal amplification assay targeting lmo0753 gene for detection of Listeria monocytogenes in wastewater. Lett. Appl. Microbiol. 2019, 69, 264–270. [Google Scholar] [CrossRef]
  180. Riquelme, M.V.; Leng, W.; Carzolio, M.; Pruden, A.; Vikesland, P. Stable oligonucleotide-functionalized gold nanosensors for environmental biocontaminant monitoring. J. Environ. Sci. 2017, 62, 49–59. [Google Scholar] [CrossRef] [PubMed]
  181. Tang, R.; Yang, H.; Gong, Y.; You, M.; Liu, Z.; Choi, J.R.; Wen, T.; Qu, Z.; Mei, Q.; Xu, F. A fully disposable and integrated paper-based device for nucleic acid extraction, amplification and detection. Lab. A Chip 2017, 17, 1270–1279. [Google Scholar] [CrossRef] [PubMed]
  182. Guo, J.; Li, J.; Chen, H.; Bond, P.L.; Yuan, Z. Metagenomic analysis reveals wastewater treatment plants as hotspots of antibiotic resistance genes and mobile genetic elements. Water Res. 2017, 123, 468–478. [Google Scholar] [CrossRef] [PubMed]
  183. Yasir, M. Analysis of Microbial Communities and Pathogen Detection in Domestic Sewage Using Metagenomic Sequencing. Diversity 2021, 13, 6. [Google Scholar] [CrossRef]
  184. Numberger, D.; Ganzert, L.; Zoccarato, L.; Mühldorfer, K.; Sauer, S.; Grossart, H.-P.; Greenwood, A.D. Characterization of bacterial communities in wastewater with enhanced taxonomic resolution by full-length 16S rRNA sequencing. Sci. Rep. 2019, 9, 9673. [Google Scholar] [CrossRef] [Green Version]
  185. Oluseyi Osunmakinde, C.; Selvarajan, R.; Mamba, B.B.; Msagati, T.A.M. Profiling Bacterial Diversity and Potential Pathogens in Wastewater Treatment Plants Using High-Throughput Sequencing Analysis. Microorganisms 2019, 7, 506. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  186. Ciesielski, M.; Blackwood, D.; Clerkin, T.; Gonzalez, R.; Thompson, H.; Larson, A.; Noble, R. Assessing sensitivity and reproducibility of RT-ddPCR and RT-qPCR for the quantification of SARS-CoV-2 in wastewater. J. Virol. Methods 2021, 297, 114230. [Google Scholar] [CrossRef]
  187. Nshimyimana, J.P.; Cruz, M.C.; Wuertz, S.; Thompson, J.R. Variably improved microbial source tracking with digital droplet PCR. Water Res. 2019, 159, 192–202. [Google Scholar] [CrossRef] [PubMed]
  188. Gordillo, R.; Rodríguez, A.; Werning, M.L.; Bermúdez, E.; Rodríguez, M. Quantification of viable Escherichia coli O157: H7 in meat products by duplex real-time PCR assays. Meat Sci. 2014, 96, 964–970. [Google Scholar] [CrossRef]
  189. Garrido, A.; Chapela, M.-J.; Román, B.; Ferreira, M.; Lago, J.; Vieites, J.M.; Cabado, A.G. Development of a multiplex real-time PCR method for simultaneous detection of Salmonella enterica, Shigella flexneri and Listeria monocytogenes in processed food samples. Eur. Food Res. Technol. 2012, 234, 571–580. [Google Scholar] [CrossRef]
  190. Collins, S.; Jorgensen, F.; Willis, C.; Walker, J. Real-time PCR to supplement gold-standard culture-based detection of Legionella in environmental samples. J. Appl. Microbiol. 2015, 119, 1158–1169. [Google Scholar] [CrossRef] [PubMed]
  191. Cui, B.; Liang, S. Monitoring Opportunistic Pathogens in Domestic Wastewater from a Pilot-Scale Anaerobic Biofilm Reactor to Reuse in Agricultural Irrigation. Water 2019, 11, 1283. [Google Scholar] [CrossRef] [Green Version]
  192. Wong, Y.P.; Othman, S.; Lau, Y.L.; Radu, S.; Chee, H.Y. Loop-mediated isothermal amplification (LAMP): A versatile technique for detection of micro-organisms. J. Appl. Microbiol. 2018, 124, 626–643. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  193. Yang, L.; Lou, J.; Wang, H.; Wu, L.; Xu, J. Use of an improved high-throughput absolute abundance quantification method to characterize soil bacterial community and dynamics. Sci. Total Environ. 2018, 633, 360–371. [Google Scholar] [CrossRef]
  194. Lou, J.; Yang, L.; Wang, H.; Wu, L.; Xu, J. Assessing soil bacterial community and dynamics by integrated high-throughput absolute abundance quantification. PeerJ 2018, 6, e4514. [Google Scholar] [CrossRef]
  195. Tourlousse, D.M.; Yoshiike, S.; Ohashi, A.; Matsukura, S.; Noda, N.; Sekiguchi, Y. Synthetic spike-in standards for high-throughput 16S rRNA gene amplicon sequencing. Nucleic Acids Res. 2017, 45, e23. [Google Scholar] [CrossRef] [Green Version]
  196. Wang, X.; Howe, S.; Deng, F.; Zhao, J. Current Applications of Absolute Bacterial Quantification in Microbiome Studies and Decision-Making Regarding Different Biological Questions. Microorganisms 2021, 9, 1797. [Google Scholar] [CrossRef]
  197. Ejeian, F.; Etedali, P.; Mansouri-Tehrani, H.-A.; Soozanipour, A.; Low, Z.-X.; Asadnia, M.; Taheri-Kafrani, A.; Razmjou, A. Biosensors for wastewater monitoring: A review. Biosens. Bioelectron. 2018, 118, 66–79. [Google Scholar] [CrossRef]
  198. Garner, E.; Davis, B.C.; Milligan, E.; Blair, M.F.; Keenum, I.; Maile-Moskowitz, A.; Pan, J.; Gnegy, M.; Liguori, K.; Gupta, S.; et al. Next generation sequencing approaches to evaluate water and wastewater quality. Water Res. 2021, 194, 116907. [Google Scholar] [CrossRef]
  199. Shapiro, E.; Biezuner, T.; Linnarsson, S. Single-cell sequencing-based technologies will revolutionize whole-organism science. Nat. Rev. Genet. 2013, 14, 618–630. [Google Scholar] [CrossRef]
  200. Xue, J.; Schmitz, B.W.; Caton, K.; Zhang, B.; Zabaleta, J.; Garai, J.; Taylor, C.M.; Romanchishina, T.; Gerba, C.P.; Pepper, I.L.; et al. Assessing the spatial and temporal variability of bacterial communities in two Bardenpho wastewater treatment systems via Illumina MiSeq sequencing. Sci. Total Environ. 2019, 657, 1543–1552. [Google Scholar] [CrossRef]
  201. Stamps, B.W.; Spear, J.R. Identification of Metagenome-Assembled Genomes Containing Antimicrobial Resistance Genes, Isolated from an Advanced Water Treatment Facility. Microbiol. Resour. Announc. 2020, 9, e00003-20. [Google Scholar] [CrossRef] [Green Version]
  202. Alneberg, J.; Karlsson, C.M.G.; Divne, A.-M.; Bergin, C.; Homa, F.; Lindh, M.V.; Hugerth, L.W.; Ettema, T.J.G.; Bertilsson, S.; Andersson, A.F.; et al. Genomes from uncultivated prokaryotes: A comparison of metagenome-assembled and single-amplified genomes. Microbiome 2018, 6, 173. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  203. Pascault, N.; Loux, V.; Derozier, S.; Martin, V.; Debroas, D.; Maloufi, S.; Humbert, J.-F.; Leloup, J. Technical challenges in metatranscriptomic studies applied to the bacterial communities of freshwater ecosystems. Genetica 2015, 143, 157–167. [Google Scholar] [CrossRef] [PubMed]
  204. Yergeau, E.; Masson, L.; Elias, M.; Xiang, S.; Madey, E.; Huang, H.; Brooks, B.; Beaudette, L.A. Comparison of Methods to Identify Pathogens and Associated Virulence Functional Genes in Biosolids from Two Different Wastewater Treatment Facilities in Canada. PLoS ONE 2016, 11, e0153554. [Google Scholar] [CrossRef] [PubMed]
  205. World Health Organization. Global Antimicrobial Resistance Surveillance System: Manual for Early Implementation; World Health Organization: Geneva, Switzerland, 2015. [Google Scholar]
  206. Bitton, G. Microbiology of Drinking Water Production and Distribution; John Wiley & Sons: Hoboken, NJ, USA, 2014; p. 312. [Google Scholar]
  207. Sun, Y.; Shen, Y.-X.; Liang, P.; Zhou, J.; Yang, Y.; Huang, X. Multiple antibiotic resistance genes distribution in ten large-scale membrane bioreactors for municipal wastewater treatment. Bioresour. Technol. 2016, 222, 100–106. [Google Scholar] [CrossRef] [PubMed]
  208. Mao, D.; Yu, S.; Rysz, M.; Luo, Y.; Yang, F.; Li, F.; Hou, J.; Mu, Q.; Alvarez, P. Prevalence and proliferation of antibiotic resistance genes in two municipal wastewater treatment plants. Water Res. 2015, 85, 458–466. [Google Scholar] [CrossRef]
  209. Nesme, J.; Cécillon, S.; Delmont, T.O.; Monier, J.-M.; Vogel, T.M.; Simonet, P. Large-scale metagenomic-based study of antibiotic resistance in the environment. Curr. Biol. 2014, 24, 1096–1100. [Google Scholar] [CrossRef] [Green Version]
  210. Xia, Y.; Li, A.-D.; Deng, Y.; Jiang, X.-T.; Li, L.-G.; Zhang, T. MinION Nanopore Sequencing Enables Correlation between Resistome Phenotype and Genotype of Coliform Bacteria in Municipal Sewage. Front. Microbiol. 2017, 8, 2105. [Google Scholar] [CrossRef]
  211. Che, Y.; Xia, Y.; Liu, L.; Li, A.-D.; Yang, Y.; Zhang, T. Mobile antibiotic resistome in wastewater treatment plants revealed by Nanopore metagenomic sequencing. Microbiome 2019, 7, 44. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  212. Law, J.W.-F.; Ab Mutalib, N.-S.; Chan, K.-G.; Lee, L.-H. Rapid methods for the detection of foodborne bacterial pathogens: Principles, applications, advantages and limitations. Front. Microbiol. 2015, 5, 770. [Google Scholar] [CrossRef] [PubMed] [Green Version]
Table 1. Pathogenic bacteria detected in wastewater with high significance to public health.
Table 1. Pathogenic bacteria detected in wastewater with high significance to public health.
Pathogenic BacteriaRelated DiseaseInfectivity aPersistence bDensity in WWTP InfluentReference
Enteric pathogensE. coliO157:H7GastroenteritisHighModerate101–106 CFU/100 mL[18]
Campylobacter spp.GastroenteritisModerateModerate102–105 MPN/100 mL[19]
Shigellaspp.ShigellosisHighShort10–107 MPN/100 mL[20,21]
Salmonellaspp.Salmonellosis; Typhoid feverLowMay multiply1–107 MPN/100 mL[20,22]
ClostridioidesdifficileSevere diarrhea and colitisHighLong-[23,24]
Non-enteric pathogensLegionellaspp.Acute
respiratory
illness,
legionellosis
ModerateMay multiply107–1010 cells/100 mL[25,26]
Mycobacteriumspp.Pulmonary
disease, skin
infection
LowMay multiply105 gene
copies/100 mL
[27]
Note: a [6]; b [17]; MPN: most probable number.
Table 2. Molecular detection methods and example of applications in the analysis of pathogenic bacteria.
Table 2. Molecular detection methods and example of applications in the analysis of pathogenic bacteria.
Molecular MethodTarget
Bacteria/Genes
Sample TypeLimit of
Detection
References
Nucleic acid targeting methodsPolymerase
chain reaction (PCR)-based method
Multiplex-PCR (mPCR)EnteropathogensWastewater-[88]
Single qPCRSalmonellaSalmonella
Isolates
-[89]
Multiplex
qPCR
Taqman methodinvA of Salmonella spp.; the paratose synthase (prt) gene, and the tyvelose epimerase (tyv) gene of group D and group A Salmonella, the Salmonella-differentiating fragment 1 (Sdf-1) sequence of S. EnteritidisEnvironmental
Samples
1 copies/reaction b[90]
SYBR greenmcr-1 geneWastewater12 copies/reaction b[91]
Microfluidic
quantitative PCR
Antibiotic resistance and heavy metal resistance genesWastewater-[92]
Droplet digital PCR (ddPCR)VBNC E. coli O157:H7/rfbEFood5–6 copies/μL b[93]
DNA MicroarraySalmonella enterica, Shigella flexneri, E. coli O157:H7, and Listeria monocytogenesFood102 CFU/mL a[94]
LAMPV. parahaemolyticusFlatfish1 CFU/mL in
buffer b; 10 CFU/g in fish sample a
[95]
FISHSalmonellaMinced lamb meat10 CFU/g a[96]
SequencingPyrosequencingBacterial communitiesSputum-[97]
Illumina technology16S rRNA geneWell-characterized bacterial reference sample-[98]
Oxford Nanopore Technologies
Whole-genome
sequencing (WGS)
381 different resistance genesWastewater-[99]
Immunology-based
methods
Enzyme-linked immunosorbent assay (ELISA)S. enterica typhimurium.River water9.2 × 103 CFU/mL a[100]
Biosensor-based
methods
Cross-linking reaction between antibody and water-soluble
cf-GQDs (carboxyl functionalized graphene quantum dots)
E. coli O157: H7Milk102 CFU/mL a, b[101]
Paper-based deviceOrigami paper-based deviceE. coliBacteria culture103 CFU/mL b within 35 min[102]
Paper-based ELISAE. coli O157:H7Food1 × 104 CFU/mL a, b[9]
Note: a method LoD (based on sample volume/mass); b assay LoD (based on reaction or L); a, b both method and assay LoD.
Table 3. Comparison studies of DNA/RNA extraction methods for various downstream molecular methods of wastewater and sediment samples.
Table 3. Comparison studies of DNA/RNA extraction methods for various downstream molecular methods of wastewater and sediment samples.
Downstream AnalysisTargetsBest/Limited Fragment LengthSuggested
Extraction Kits/Methods
Storage and
Pretreatment
of Samples
Recovery
Efficiency
Sample TypeReference
PCR-based methodLambda DNA-FastDNA
Spin Kit for Soil
Stored at −70 °C15.5% to 43.3%Sediment[168]
Ancylostoma caninum ova-MO Bio Power Max® Soil DNA Extraction Kit (MO BIO Laboratories Inc, Carlsbad, CA USA); FiltrationStored at 4 °C in the darkTreated wastewater: 39–50%
Raw wastewater: 7.1–12%
Wastewater[169]
Microarray16S Rdna, cpn60, and wecEDetection
sensitivity is
optimal when DNA
targets > 500 bp
Bead beating separation and ammonium acetate purificationCentrifuged at 3000× g for 16 min at room temperature; stored at −20 °C81 μg DNA/mLWastewater[170]
NGSARG150 bp
(Limitation of the sequencing length)
FastDNA Spin Kit for SoilEthanol fixation (50%); filter-concentrated using 0.22-μm mixed cellulose ester filters;
stored at –20 °C
10.3 ± 3.6 μg/sampleWastewater[171]
16S rRNA
amplicons
Qiagen Mini Kit and MO Bio PowerSoil KitCentrifuged at 10,000× g for
5 min to pellet; filtered through 0.22 μm cellulose nitrate
membrane filters
-Wastewater[172]
Table 4. Applications of molecular methods in the analysis of pathogenic bacteria in wastewater.
Table 4. Applications of molecular methods in the analysis of pathogenic bacteria in wastewater.
Detection MethodCultivationDNA/RNA ExtractionTarget
Pathogen
BiomarkerSample TypeLimit of
Detection (LoD)
References
PCRYesYesCampylobacter spp., C. jejuni, C. coli16S rRNA, mapA, ceuEWastewater2 CFU/100 mL[173]
E. coli O157:H7stx2
S. typhimuriumstx1
Real-time PCRNoYesE. coliuidA geneWastewater10 gc/reaction (standard curve)[174]
NoSimkania
negevensis
16S rRNA geneWastewater5 gc/reaction (standard curve)[175]
NoYesS. enterica serovar
Typhi
stgAWastewater0.05–0.005 CFU/mL of seeded wastewater[176]
S. enterica serovar Paratyphi ASSPAI
Droplet digital PCR (ddPCR)YesYesShiga toxin-producing E. colistx2River water, wastewater6 gc/reaction of standard curve; 32 copies/100 mL in river water[177]
qPCR arrayNoYesAll285 ARGs and nine transposase genesWastewater-[166]
NoYesAll229 ARGs and 25 mobile genetic elementsWastewater-[178]
Microfluidic
qPCR
NoYesAllARGs, heavy metal resistance genes, genes encoding the integrase, and 16S rRNA genesWastewater, drinking water-[92]
LAMPYesYesListeria
monocytogenes
lmo0753 geneWastewater65 fg/µL of DNA and 38 CFU/mL in cell culture[179]
FISHYesNoSalmonella spp.23S rRNAWastewater102, 10, and 1 CFU/mL for 0 h,
6 h, and 24 h of enrichment in Rappaport-Vassiliadis broth, respectively.
[137]
Biosensor-based deviceNoNoARGmecA geneARG-spiked wastewater effluent70 pM
(4 × 107 gc/μL) by bootstrapping
[180]
Paper-based deviceNoNoSalmonella
typhimurium
fimASpiked wastewater102 CFU/mL[181]
YesNoβ-lactamase-expressing bacteriaβ-lactamaseWastewater3.8 × 106 CFU/mL[157]
SequencingNoYesAllARGsSludge-[182]
NoYesShotgun metagenomic for
microbial community analysis and pathogen detection
WastewaterDetected
87 pathogenic/opportunistic
Bacteria, with most having <1% abundance.
[183]
YesYesNanopore and Illumina
metagenomics analysis for mobile antibiotic resistome
Wastewater-[25]
NoYesFull-length 16S rRNAWastewater-[184]
NoYes16S-rRNAWastewater-[185]
Table 5. PCR primer-probe sets available for the detection and quantification of typical pathogenic bacteria in wastewater and other samples.
Table 5. PCR primer-probe sets available for the detection and quantification of typical pathogenic bacteria in wastewater and other samples.
Pathogenic
Bacteria
Available PCR Primers and Probes
(5′—3′)
SensitivityReference
E. coliO157:H7RFBEO157-F GGATGACAAATATCTGCGCTGC
RFBEO157-R GGTGATTCCTTAATTCCTCTCTTTCC
RFBEO157-P HEX-TACAAGTCCACAAGGAAAG-BHQ1
1 CFU/g of seeded meat products after 4 h enrichment period at 37 °C[188]
Rfb-F GTGTCCATTTATACGGACATCCATG
Rfb-R CCTATAACGTCATGCCAATATTGCC
2 CFU/100 mL of raw sewage[173]
Campylobacter spp.16S-F CCTGAMGCAGCAACGCC
16S-R CGGAGTTAGCCGGTGCTTATT
16S-P FAM-CTCCGAAAAGTGTCATCCT –MGB
3.2 gene copies/reaction[19,40]
C. jejunihipO-F CTTGCGGTCATGCTGGACATAC
hipO-R AGCACCACCCAAACCCTCTTCA
hipO-P VIC-ATTGCTTGCTGCAAAGT- MGB
2.0 × 102 CFU/g of feces[31]
C. coliglyA-F AAACCAAAGCTTATCGTGTGC
glyA-R AGTGCAGCAATGTGTGCAATG
glyA-P FAM-CAACTTCATCCGCAAT- MGB
2.5 × 102 CFU/g of feces
C. lariglyA-F CAGGCTTGGTTGTAGCAGGTG
glyA-R ACCCCTTGGACCTCTTAAAGTTTT
glyA-P TET-CATCCTAGTCCATTCCCTTATGCTC
ATGTT-TAMRA
2.1 gene copies/reaction[19]
Shigellaspp.ipaH-F CGCAATACCTCCGGATTCC
ipaH-R TCCGCAGAGGCACTGAGTT
ipaH-P FAM- AACAGGTCGCTGCATGGCTGGAA-BHQ1
10−5 ng/μL for genomic DNA templates, 10−1 CFU/mL for Shigella bacteria culture[113]
Salmonellaspp.invA-F AACGTGTTTCCGTGCGTAAT
invA-R TCCATCAAATTAGCGGAGGC
invA-P TGGAAGCGCTCGCATTGTGG
9–15 CFU/25 g food sample[189]
S. TyphistgA-F TATCGGCAACCCTGCTAATG
stgA-R TATCCGCGCGG TTGTAAAT
stgA-P FAM-CCATTACAG CATCTGGCGTAGCGA-BHQ1
0.05–0.005 CFU/mL of wastewater[176]
S. entericaserovarParatyphi ASSPAI-F ACCATCCGCAGGACAAATC
SSPAI-R GGGAGATTACTGATGGAGAGATTAC
SSPAI-P Cy5-AGAGTGCAAGTGGAGTGCCTCAAA-BHQ2
C. difficiletpi-F AAAGAAGCTACTAAGGGTACAAA
tpi-R CATAATATTGGGTCTATTCCTAC
For simultaneous identification and toxigenic type characterization (fecal and urban water
samples)
[59,66]
tcdB-F GGAAAAGAGAATGGTTTTATTAA
tcdB-R ATCTTTAGTTATAACTTTGACATCTTT
tcdA-F AGATTCCTATATTTACATGACAATAT
tcdA-R GTATCAGGCATAAAGTAATATACTTT
Legionellaspp.PanLeg-F GGCGACCTGGCTTC
PanLeg-R1 GGTCATCGTTTGCATTTATATTTA
PanLeg-P1 FAM-ACGTGGGTTGCAA-MGBNFQ
5 genome units (GU)/reaction with water sample[190]
L.
pneumophila
Lp-F TTGTCTTATAGCATTGGTGCCG
Lp-R CCAATTGAGCGCCACTCATAG
Lp-P Quasar670-CGGAAGCAATGGCTAAAGGCATGCA-BHQ3
L. pneumophila sg1Lp1-F TGCCTCTGGCTTTGCAGTTA
Lp1-R CACACAGGCACAGCAGAAACA
Lp1-P VIC-TTTATTACTCCACTCCAGCGAT-MGBNFQ
Mycobacteriumspp.16S rRNA-F: ATGCACCACCTGCACACAGG
16S rRNA-R: GGTGGTTTGTCGCGTTGTTC
10–100 copies of template
plasmid/reaction (raw wastewater)
[191]
Table 6. Advantages, limitations, and prospects of various molecular methods in analyzing pathogenic bacteria for WBE applications.
Table 6. Advantages, limitations, and prospects of various molecular methods in analyzing pathogenic bacteria for WBE applications.
Molecular MethodBiomarkersAdvantagesLimitationsReference
Nucleic acid targeting methodsDNA/RNA- High sensitivity
- High specificity
- Multiple targets detection and quantification
- Fast community profiling
- Require sample storage and processing
- Require DNA/RNA extraction, which can cause DNA/RNA loss
- Sensitive to inhibitors
- High cost for large number of samples
- Usually need specialized instruments
[11,212]
Immunology-based methodsProteins- Low cost
- Can be automated
- Can detect bacterial toxins
- Require pre-enrichment
- Low sensitivity
- Require labeling of antibodies and antigens
[9]
Biosensor-based methodsDNA/RNA, proteins, chemicals- High sensitivity
- Real-time detection
- Label free
- High cost
- Require specialized instruments
- Low specificity
- Not suitable for simultaneous detection of various organisms
- Low reproducibility and insufficient stability
[197]
Paper-based deviceDNA/RNA, proteins, chemicals- Cost effective
- Instrument free
- Detection limit (LoD) is usually high due to the traditional colorimetry
- Limitations of the structure and material of paper device
[13,154,156]
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Zhang, S.; Li, X.; Wu, J.; Coin, L.; O’Brien, J.; Hai, F.; Jiang, G. Molecular Methods for Pathogenic Bacteria Detection and Recent Advances in Wastewater Analysis. Water 2021, 13, 3551. https://doi.org/10.3390/w13243551

AMA Style

Zhang S, Li X, Wu J, Coin L, O’Brien J, Hai F, Jiang G. Molecular Methods for Pathogenic Bacteria Detection and Recent Advances in Wastewater Analysis. Water. 2021; 13(24):3551. https://doi.org/10.3390/w13243551

Chicago/Turabian Style

Zhang, Shuxin, Xuan Li, Jiangping Wu, Lachlan Coin, Jake O’Brien, Faisal Hai, and Guangming Jiang. 2021. "Molecular Methods for Pathogenic Bacteria Detection and Recent Advances in Wastewater Analysis" Water 13, no. 24: 3551. https://doi.org/10.3390/w13243551

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

Article Metrics

Back to TopTop