Next Article in Journal
Uncovering Key Transcription Factors Driving Chilling Stress Tolerance in Rice Germination
Previous Article in Journal
Cell Senescence and the DNA Single-Strand Break Damage Repair Pathway
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

DNA-Based Technology for Herpesvirus Detection

Department of Environmental and Prevention Sciences, University of Ferrara, 44121 Ferrara, Italy
*
Author to whom correspondence should be addressed.
These authors contributed equally to this work.
DNA 2024, 4(4), 553-581; https://doi.org/10.3390/dna4040037
Submission received: 29 October 2024 / Revised: 2 December 2024 / Accepted: 5 December 2024 / Published: 13 December 2024

Abstract

:
The detection of viral DNA is considered crucial in both diagnosis and prognosis. Nowadays, molecular diagnostic approaches represent the most promising tools for the clinical detection of viral infections. This review aims to investigate the most used and promising DNA-based technologies for viral detection, focusing on herpesviruses because of their ability to undergo latent and reactivation cycles, persisting lifelong in the host in association with several diseases. Molecular technologies, such as PCR-based assays, enhance sensitivity and specificity in identifying viral DNA from clinical samples such as blood, cerebrospinal fluid and saliva, indicating PCR and its derivatives as the gold standard methods for herpesvirus detection. In conclusion, this review underscores the need for continuous innovation in diagnostic methodologies to address the complexities of herpesvirus identification in different clinical samples.

1. Introduction

Viruses pose a serious threat to humans and are a significant global concern [1]. Given the huge impact of viral infections on human health, virologists and clinical biologists are increasingly focused on designing effective and precise detection methods, primarily based on molecular biology techniques. Herpesviruses represent a large family of DNA viruses characterized by their ability to persist lifelong within the host, undergoing latency/reactivation cycles [2] which make diagnosis a considerable challenge.
During primary infection, herpesviruses typically cause active lytic infection, characterized by viral replication, cell lysis, and occasionally clinically evident symptoms [3].
Following the primary infection, the virus enters a latent phase in specific cell types. During the latency phase, the viral genome persists in infected cells with minimal to no gene expression, allowing the virus to evade the immune response and persist for the lifetime of the host. The ability of herpesviruses to periodically reactivate leads to recurrent infection that results in severe dissemination and complication, especially in immunocompromised subjects.
In light of recent evidence suggesting a potential role for the herpesvirus latency/reactivation cycle in the development or worsening of several diseases and clinical conditions—including cancer, autoimmunity, and neurological diseases [4]—the use of specific and rapid DNA-based detection methods has become extremely relevant.
In this review, we explore the main DNA-based molecular diagnostic methods currently employed for the detection of human herpesviruses.

2. Diagnostic Method for Virus Detection

Virus detection methods primarily rely on molecular-based techniques to improve efficiency and reduce diagnostic turnaround time. These approaches use immunological detection techniques or specific oligonucleotide probes designed to bind selectively to viral nucleic acids [5,6] (Table 1). This section provides an overview of the main molecular techniques used for virus detection.

2.1. Viral Molecular Detection Techniques

Molecular techniques are increasingly employed in clinical settings due to their rapid and precise identification of viral agents [2]. These methods mainly consist in the identification of viral genomes or specific genes by nucleic acid amplification techniques (NAATs), hybridization, and sequencing.

2.1.1. PCR-Based Methods

Polymerase Chain Reaction (PCR)

PCR is the most widely used nucleic acid-based technique for viral detection. It is fast, sensitive, and specific, enabling the amplification of target DNA sequences via DNA polymerase. This exponential amplification ensures high sensitivity, with amplicons visualized through gel electrophoresis [7]. PCR can detect both DNA and RNA viruses, with the latter requiring reverse transcription prior to amplification.

Real-Time PCR (qPCR)

Although conventional PCR provides qualitative or semi-quantitative results, real-time quantitative PCR (rtPCR) addresses the need for accurate viral load quantification in clinical diagnostics. This technique uses fluorescent dyes or specific probes to monitor DNA amplification in real time. Fluorescence intensity is plotted as a function of amplification cycles, providing quantitative data [8]. Common dyes include SYBR Green, which binds to all double-stranded DNA (dsDNA), though it may produce non-specific signals. To overcome this, fluorophore-bound probes are used, emitting fluorescence only when hybridized with complementary sequences, enhancing specificity.

Multiplex PCR

Multiplex PCR employs multiple primers and probes to detect different viral targets within the same reaction. This method is especially valuable in respiratory infections where co-infections are common. Some assays can identify up to eight different viruses in a single test, reducing time and costs [9,10,11].

Droplet Digital PCR (ddPCR)

A significant advancement in PCR technology is droplet digital PCR (ddPCR). This method partitions DNA into millions of microdroplets, amplifying each droplet independently. Absolute quantification is achieved without standard curves by applying the Poisson distribution to the fluorescence signal [12,13]. ddPCR is particularly suited for detecting low viral loads and provides high sensitivity and precision. However, limitations include its cost, sensitivity to contamination, and the need for specialized equipment [14].

2.1.2. Isothermal Amplification (LAMP)

Loop-mediated isothermal amplification (LAMP) amplifies DNA at a constant temperature, eliminating the need for thermal cycling. This method uses primers designed to form stem–loop structures, enabling rapid and efficient amplification [15,16]. LAMP is highly sensitive and specific and is widely used in field settings due to its simplicity and minimal equipment requirements. However, its specificity may be lower than PCR, as primer design is critical to prevent non-specific amplifications [17,18].

2.1.3. Hybridization Techniques: Microarrays and Biosensors

Microarrays

Microarray technology allows for the simultaneous hybridization of target DNA to thousands of immobilized probes on a solid surface. This approach is ideal for detecting multiple pathogens or identifying novel variants through conserved gene sequences [19,20]. Techniques like using clinical array technology (CLART) enable the sensitive and automated detection of various viruses, including HPV and herpesviruses, offering cost-effective solutions for high-throughput diagnostics [20,21,22].

Biosensors

Biosensors utilize DNA nanotechnology to detect viral DNA by converting molecular interactions into measurable signals, such as electrical or optical outputs [23,24]. These devices are portable, cost-effective, and rapid, making them suitable for point-of-care diagnostics. Recent advancements include aptamers and DNAzymes, which enhance sensitivity and specificity while offering therapeutic potential. Biosensors hold promise for both diagnostics and therapeutic applications, but further development is needed to expand their clinical utility [25,26,27].

2.1.4. CRISPR-Based Detection

CRISPR technology has revolutionized viral diagnostics. Systems like CRISPR-Cas12 and CRISPR-Cas13 provide high sensitivity and specificity for DNA or RNA detection [28,29]. Platforms such as DETECTR leverage CRISPR-Cas12 for fluorescence-based or colorimetric detection, making these methods suitable for field diagnostics or point-of-care applications [30,31].
CRISPR-based diagnostics rival PCR in sensitivity but often require the pre-amplification of target nucleic acids, increasing complexity and contamination risks. Future improvements aim to simplify workflows, minimize contamination through closed-tube reactions, and enable multiplex detection for broader clinical adoption [30].

2.1.5. Next-Generation Sequencing (NGS)

NGS is a transformative tool for comprehensive viral genome analysis. In complex clinical cases requiring viral discrimination, additional laboratory procedures, such as restriction enzyme analysis or complete genome sequencing, may be necessary using next-generation sequencing (NGS) technologies [32]. These massively parallel sequencing platforms can sequence highly diverse genetic materials with excellent sensitivity [33]. Furthermore, these platforms are sequence-independent, making them exceptional tools for virus identification. They provide unparalleled sensitivity for detecting low viral loads, co-infections, and unknown pathogens. Metagenomic NGS (mNGS) has proven particularly useful in diagnosing respiratory infections, central nervous system infections, and other complex cases [34,35].
NGS also facilitates detailed virome analysis, enabling the identification of mutations and the monitoring of emerging variants [36]. However, its adoption is limited by high costs, labor-intensive sample preparation, and the need for bioinformatics expertise. Despite these limitations, NGS remains invaluable for uncovering viral interactions and advancing diagnostics in complex clinical scenarios.
Molecular techniques are integral to efficient and accurate herpesvirus detection. PCR-based methods, including qPCR, remain the gold standard due to their reliability and widespread use. However, emerging technologies like CRISPR and NGS are expanding the boundaries of viral diagnostics by offering more comprehensive pathogen detection and insights into viral behavior [37], improving the possibility to discriminate between lytic and latent infections [32]. The integration of these advanced methods into clinical practice will require addressing cost, accessibility, and workflow complexity, ensuring broader applicability across healthcare settings.
In the next chapters of this review, we will describe the main DNA-based methods used for viral identification of herpesviruses.

3. Herpesviruses

The identification of herpesviruses in clinical samples of different origin is critical due to their widespread prevalence [38] and their ability to establish lifelong latent infections in the host, often associated with serious diseases [39].
Herpesviruses infect a wide range of animals, with more than 200 species identified, including nine that infect humans (HHVs) [40]. These viruses are classified into three subfamilies based on their tropism and latency sites, namely Alphaherpesvirinae, Betaherpesvirinae, and Gammaherpesvirinae [41,42] (Table 2).
The Alphaherpesvirinae subfamily include herpes simplex virus type 1 and 2 (HSV-1 and HSV-2) and Varicella-zoster virus (VZV). These viruses are characterized by the ability to replicate in a wide variety of host tissues and then establish a latent infection in the ganglia [43,44] (Table 2).
On the contrary, Betaherpesvirinae show a restricted host range, establishing latent infection in lymphocytes, secretory glands, cells of the reticuloendothelial system, and kidneys. The viruses that take part of this subfamily are cytomegalovirus (CMV), human herpesvirus-6 (HHV-6) A and -6B, and human herpesvirus 7 (HHV-7) (Table 2).
Gammaherpesvirinae have the most limited host range. They replicate in lymphoblastoid cells, leading to lytic infections and establishing latent infections in lymphoid tissue. Epstein–Barr virus (EBV) and human herpesvirus 8 (HHV-8) are members of this sub- family (Table 2).
Table 2. Characteristics of herpesvirus subfamilies.
Table 2. Characteristics of herpesvirus subfamilies.
SubfamilyVirusPrimary Sites of ReplicationSite of LatencyReferences
AlphaherpesvirusHSV-1Epithelial cells in and around mouth, ocular, genital areaSensory nerve cells[44]
HSV-2Epithelial cells in and around genital areaSensory nerve cells[45]
VZVEpithelial cells in skinNeurons of trigeminal ganglia and dorsal root ganglia[46,47]
BetaherpesvirusCMVEpithelial cells of salivary glands, kidneys, genital tractCD34+ myeloid progenitor, CD14+ monocytes[48,49,50]
HHV-6A/BEpithelial cells of salivary glands, CD4+ T lymphocytesCD34+ stem cells, monocytes[51,52,53,54]
HHV-7Epithelial cells of salivary glands, CD4+ T lymphocytes CD4+ T lymphocytes[55]
GammaherpesvirusHHV-8Epithelial cells in oropharynxB cells[27,56,57]
EBVEpithelial cells in oropharynx, genital tractB cells[58,59]

Latency and Reactivation

After primary infection, HHVs can establish latency following two main strategies. The viral genome can persist as a circular episome in the cell nucleus due to the expression of latency-associated viral genes [60,61] or, less frequently, viral DNA can integrate into human genomic DNA [62]. Reactivation occurs in response to various stimuli, including stress, hormonal changes, UV light exposure, and immunosuppression [1,7,63,64]. Although most reactivations are clinically asymptomatic, they play a crucial role in maintaining a viral reservoir and can lead to severe diseases, such as Alzheimer’s disease, multiple sclerosis, or lupus, especially in immunocompromised individuals [3].

4. DNA-Based Technology for Herpesviruses Detection

The early and accurate identification of herpesvirus DNA in clinical samples is critical for managing and treating infections. In fact, a prompt detection enables early antiviral therapy, reducing the risk of complications, disease progression, and transmission [5]. Regular monitoring is especially important for immunocompromised patients, as viral reactivation can significantly impact disease outcomes. Molecular tools, such as PCR, qPCR, and next-generation sequencing (NGS), remain the most effective methods for herpesvirus diagnosis.
Given the clinical and genomic peculiarities that characterize each herpesvirus, this review will report the most effective molecular tools currently used for HHV diagnosis (Table 3).

4.1. Herpes Simplex Virus (HSV)

HSV is a neurotropic virus which consists of HSV-1 and HSV-2 that cause a spectrum of diseases ranging from asymptomatic infections to life-threatening conditions like herpes simplex encephalitis and neonatal herpes [102]. HSV-1 is typically transmitted via the oral route and is associated with upper body infections, including oral and ocular herpes, while HSV-2 is a sexually transmitted infection that primarily causes genital herpes [103]. The choice of clinical sample for HSV detection depends on the infection site (Table 3), such as saliva for oral herpes, genital swabs for genital herpes, or cerebrospinal fluid for suspected encephalitis [104].
Additional diagnostic tests for HSV include the collection of sore fluid from oral, genital, or ocular lesions for direct detection in cell cultures. However, this labor-intensive procedure has been largely replaced by PCR analysis of genetic material extracted from the sample. PCR provides 100% sensitivity and specificity compared to viral culture methods, which achieve only 50% sensitivity [67].
HSV can also infect ocular cells, leading to complications in the retina, cornea, and other parts of the eye [105]. In cases of HSV-associated ocular infections, such as uveitis—a serious sight-threatening condition commonly caused by HSV—samples from intraocular specimens must be collected and analyzed for the presence of the virus.
For cases of herpes simplex encephalitis (HSE), HSV detection can be performed on CSF samples using molecular assays. In more complex clinical cases, metagenomic next-generation sequencing (mNGS) can also be employed to identify the virus [71,106].

4.1.1. Detection of HSVs in Oral and Vaginal Samples

As mentioned above, HSV-1 is commonly associated with oral herpes and only in rare cases, genital herpes, which is most commonly due to HSV-2 infection [107,108]. The most frequent complications associated with HSV mucosal infection include the appearance of highly painful sores and scabs, which resolve within few weeks. For proper diagnosis, it is important to take a swab sample from these lesions before they resolve in order to collect intact virions [103]. Molecular techniques like PCR have replaced traditional methods such as viral culture [105,109], providing higher sensitivity and specificity for detecting HSV in oral and genital lesions [110]. The gold standard molecular technique for HVS diagnosis is PCR [5], particularly qPCR [111]. The fluorescence-based real-time PCR technique has completely revolutionized PCR-based systems for HSV detection in various types of clinical samples. This method also allows for the discrimination between HSV-1 and HSV-2 using multiplex PCR assays [112]. The enhanced sensitivity of multiplex qPCR for HSV detection was confirmed in a study [75], which reported successful HSV detection in genital samples from four patients who had tested negative using the viral culture method. HSV typing was performed using multiplex qPCR with specific primers targeting the TK3 and POL genes for HSV-1 and HSV-2, respectively, demonstrating the specificity and reliability of this technique for diagnosing genital herpes [82]. The same method has also been used to detect HSV in oral mucocutaneous samples, further confirming its diagnostic efficacy [113,114].
Other molecular approaches have explored the use of a restriction fragment length polymorphism (RFLP) analysis of HSV PCR products. This technique enables the detection of variations at specific single nucleotide polymorphism (SNP) loci through enzymatic restriction, providing valuable insights into viral populations. Such information is critical for viral classification and epidemiological studies that investigate host–pathogen interactions [115]. The DNA polymerase I genes of HSV-1 and HSV-2 exhibit 93% sequence identity; therefore, polymorphisms in this viral gene can be identified using RFLP analysis. This approach utilizes sequence data from public databases to design restriction endonuclease recognition sites for the specific identification of HSV strains.

4.1.2. Detection of HSV from CSF Samples

Although the most accurate method for diagnosing herpes simplex encephalitis (HSE) involves the isolation of HSV or the detection of its viral antigens in brain tissue, the detection of HSV DNA in cerebrospinal fluid (CSF) is critical for diagnosis [116] (Table 3).
Currently, while no FDA-approved assays exist for prenatal HSV meningitis or HSE screening in CSF, many molecular diagnostic tests are in use [117,118,119,120]. Detecting HSV DNA by PCR in CSF samples is highly effective, with a sensitivity of 74–98% and a specificity of 100%, making it a valuable tool for diagnosing HSE or neonatal HSV infections [121]. However, the routine clinical use of semi-quantitative PCR assays for HSV detection in CSF remains uncertain, particularly regarding their ability to discriminate between HSV-1 and HSV-2 [122,123,124].
This limitation has been addressed by employing multiplex droplet digital PCR (ddPCR) for CSF samples. Xunhua Zhu et al. suggested that the detection of HSV DNA using ddPCR may serve as a useful diagnostic method for HSE, correlating disease severity and patient age with the number of HSV DNA copies detected in CSF [69]. In this study, CSF samples from patients with viral central nervous system (CNS) infections were analyzed using quadruplex ddPCR, which proved to be an accurate and specific method for detecting both HSV-1 and HSV-2. The results were comparable to those obtained using qPCR and quantitative reverse transcription PCR (qRT-PCR) assays through commercial kits [69].
Next-generation sequencing (NGS) has also emerged as a powerful tool for detecting HSV in CSF samples, particularly in cases of suspected viral infections affecting the CNS [71]. NGS provides detailed genomic information that can identify mutations or variations within viral strains. This is particularly valuable when the causative agent is unknown or when multiple infections are suspected. For instance, a study demonstrated that NGS could identify various herpesviruses in patients with neurological symptoms, offering improved diagnostic accuracy compared to conventional PCR methods [125,126].

4.1.3. Detection of HSV in Intraocular Samples

HSV can cause serious ocular complications, such as keratitis and uveitis, which may lead to blindness if left untreated [127,128]. These manifestations can result from either primary ocular HSV infection or recurrent episodes. During herpes simplex keratitis or infectious uveitis, the presence of HSV is typically confirmed using a multiplex qPCR assay, which can differentiate between HSV-1, HSV-2, VZV, and CMV [129]. Although molecular diagnostic tests for HSV infections in ophthalmology are widely available and highly sensitive, recent years have seen the adoption of metagenomic deep sequencing. This advanced technique allows for the detection of infections from less than 50 μL of intraocular fluid, providing an innovative approach to diagnostics [127,130,131].

4.2. Varicella-Zoster Virus (VZV)

VZV, an Alphaherpesvirus, causes primary chickenpox infection and remains latent in dorsal root ganglia. Reactivation results in herpes zoster (shingles), often with complications like post-herpetic neuralgia [132]. Detecting VZV DNA is particularly important during pregnancy and in CNS infections. Approximately 90% of the adult population tests positive for VZV antibodies, and studies on pregnant women report seropositivity rates between 80% and 91% [133,134,135]. Primary infection with VZV during pregnancy increases the risk of clinical complications for both the mother and fetus. The complications and sequelae of gestational VZV infection include pneumonia, an increased risk of premature abortion, congenital varicella syndrome (CVS), neonatal varicella, and herpes zoster during the first year of life [136,137]. The risk of adverse effects is greater for the mother in the third trimester of pregnancy, whereas it is higher for the fetus in the first and second trimesters [138]. For these reasons, the detection of VZV, especially during pregnancy, is crucial to prevent adverse outcomes, guiding antiviral therapy and preventive measures, ensuring proper prenatal care [137]. VZV can also lead to encephalitis, a relatively rare condition that can cause significant morbidity and mortality, particularly in vulnerable populations such as the elderly and immunocompromised individuals. Recent studies indicate that the incidence of VZV encephalitis is higher than previously estimated, occurring at a rate of approximately 5.3 cases per million people annually [139], mainly affecting elderly adults [140]. These findings highlight the importance of detecting VZV DNA in cerebrospinal fluid (CSF) samples as a key diagnostic method [141].
In summary, depending on the symptomatology, clinical samples for VZV detection may include vesicle/skin swabs, CSF, the respiratory system or eye swabs, genital swabs, blood, or other body fluids (Table 3). Once collected, these samples are typically analyzed using PCR-based assays, which represent the most sensitive methods for detecting VZV across a variety of specimens in cases of both invasive disease and localized lesions [142].

4.2.1. Detection of VZV in Vesicle Fluids Swabs, Crusts, or Fixed Tissue Samples

Vesicle fluid swabs, dried crusts, skin biopsy, or fixed tissue specimens are the best source of VZV DNA for molecular assays [75,143].
Currently, PCR is the most useful laboratory test to confirm suspected varicella and herpes zoster (Table 3). PCR can detect VZV DNA rapidly and sensitively in skin lesions, vesicles, scabs, and maculopapular lesions [144,145]. Modifications of the basic PCR technique have been used to increase assay sensitivity, including nested PCR, real-time PCR, and multiplex assays.
Among these methods, the Lyra Direct assay (Lyra™ Direct HSV 1 + 2/VZV Assay; Quidel Corp. CA, USA) is a multiplex real-time PCR for the detection and differentiation of HSV-1, HSV-2, and VZV in cutaneous or mucocutaneous lesion samples from symptomatic patients, approved by the FDA in 2014 [76]. This assay identifies and differentiates HSV-1, HSV-2, and VZV using species-specific probes (TaqMan-based probes) under optimized conditions in a single reaction tube, generating distinct amplicons for each virus [76]. To confirm these results, positive amplicons are cloned and validated through sequencing [146].

4.2.2. Detection of VZV in CSF Samples

As with other HHV infections, molecular tests have also rapidly become the gold standard for CSF sample analysis. Numerous studies have confirmed the high sensitivity of PCR-based molecular methods for detecting VZV in various clinical specimens [147], including CSF for diagnosing herpetic encephalitis and meningitis [148,149], as well as amniotic fluid [144,150]. Rapid laboratory diagnosis is particularly crucial when the CNS is involved, especially in cases of clinically ambiguous varicella-related skin manifestations that may suggest VZV infection. Specific molecular analyses for VZV play a critical role in guiding therapeutic decisions and preventing CNS complications as the infection progresses. Real-time PCR assays have proven to be as sensitive as nested PCR for detecting VZV, while being faster, easier to use, and significantly reducing the risk of molecular contamination [151]. In recent years, next-generation sequencing has emerged as a new promising diagnostic tool for VZV-related CNS infections in CSF samples. NGS provides high sensitivity, the capability to detect multiple pathogens simultaneously, and the ability to identify unexpected co-infections. Recent studies have shown that NGS, particularly metagenomic NGS (mNGS), provides higher sensitivity for detecting VZV in CSF compared to conventional methods such as PCR and antibody tests. In one study involving patients with suspected VZV CNS infections, mNGS identified VZV DNA in 16 patients, with some samples showing co-infection with other pathogens like cytomegalovirus and herpes simplex virus. This ability is crucial in clinical settings where rapid and accurate diagnosis is essential for effective clinical management [80,81] (Table 2).

4.2.3. Detection of VZV in Amniotic Fluid Samples

The detection of VZV in amniotic fluid samples is crucial for diagnosing congenital varicella syndrome, particularly when maternal infection occurs during pregnancy [144]. PCR is the most effective method for detection, demonstrating significantly higher sensitivity compared to traditional cell culture techniques. It represents a vital tool for prenatal diagnosis, enabling the timely intervention and management of potential congenital infections [79]. In a study involving 107 women with clinical varicella before 24 weeks of gestation, PCR detected VZV in 8.4% of samples, whereas only 1.8% tested positive using cell culture methods [144].
In 2021, a case report described a mother diagnosed with varicella at 12 weeks of gestation who underwent amniocentesis. VZV was detected in the amniotic fluid via PCR, confirming congenital varicella syndrome. The newborn exhibited severe abnormalities and, unfortunately, did not survive long after delivery [152]. These findings underscore the importance of early VZV detection in pregnant women, with qPCR remaining the gold-standard diagnostic technique.

4.3. Epstein–Barr Virus (EBV)

EBV infects over 95% of adults worldwide and is linked to diseases like infectious mononucleosis and several cancers, including Burkitt’s lymphoma and nasopharyngeal carcinoma [153,154,155]. EBV can infect a wide range of cells and tissues, including T and B lymphocytes, nasopharynx and oropharynx squamous epithelial cells, salivary and stomach glands, thyroid glandular epithelial cells, and smooth muscle and follicular dendritic cells [59,60] (Table 2). The quantitative measurement of EBV DNA is necessary to distinguish between healthy carriers and patients with EBV-related diseases [156]. Various molecular techniques have been established and used for the identification of EBV DNA and to measure viral load [156]. To date, in situ hybridization (ISH), protein-based assays, qPCR, and immunoblotting have been utilized in the diagnosis and staging of EBV infection. Although these methods assist in diagnosis, the lack of standardization means that the observed differences in sensitivity and specificity among laboratories should be systematically addressed [157,158].

4.3.1. Detection of EBV from Gingival Swabs and Salivary Swabs

Gingival crevicular fluid (GCF) and saliva have been identified as the primary source of EBV (Table 3). Sampling is achieved by rubbing a swab along the gum line [159]. There are several methods to detect and quantify EBV from these samples, including in situ hybridization, an EBV clonality assay, immunohistochemistry, ELISA, and PCR. In particular, qPCR performed with specific labeled probes provides a precise and sensitive method for EBV quantification in oral samples [83]. Another disease caused by EBV is oral hairy leukoplakia, which is characterized by white patches on the lateral borders of the tongue. It is most common in immunocompromised individuals, particularly those with HIV/AIDS. The detection of EBV in cases of oral hairy leukoplakia can be performed using molecular techniques, such as multiplex PCR to detect EBV DNA in tissue samples from the lesions [82].

4.3.2. Detection of EBV from CSF and Peripheral Blood

The chronic persistence of EBV-infected cells in the peripheral circulation and/or in the CNS is possibly associated with lytic viral reactivation. This clinical aspect has received increasing attention in recent years as a potential cause of MS onset and progression [160]. A real-time PCR assay was shown to be the best technique for the detection and quantification of EBV viral load in this type of sample (Table 3). Furthermore, to establish whether the presence of the virus is associated with latently infected cells or viral lytic reactivation, EBV DNA detection could be performed on both cell-associated CSF fractions and peripheral blood mononuclear cells (PBMCs) to investigate the possible role of EBV systemic infection in the disease. This method allows for the identification of lymphomonocytes acting as carriers of viral particles from the periphery to the CSF or the assessment of the release of a free virus following lytic infection [161].

4.4. Human Cytomegalovirus (CMV)

CMV is a highly prevalent Betaherpesvirus, with seroprevalence rates ranging from 60% to over 90% worldwide [162]. However, most infected individuals remain asymptomatic because of the development of an effective memory immune response [163]. Following initial infection, the virus can remain latent in myeloid lineage cells, such as CD34+ progenitor cells and peripheral blood monocytes. CMV is acquired most commonly early in life, during childhood to early adulthood, through exposure to saliva, tears, urine, stool, breast milk, semen, and other body secretions from infected individuals [164]. It can also be transmitted efficiently via organ and tissue transplantation and blood transfusions [165], making the screening for CMV positivity crucial to avoid graft.
Upon the resolution of primary infection, similar to other herpesviruses, CMV establishes lifelong latency, but in contrast to HSVs, genes expressed during CMV latency are not latency-specific, as their expression was observed also during the lytic cycle [166,167].
A critical aspect of CMV infection is gestational infection. Primary CMV infection occurs in 0.7 to 4.1% of all pregnancies and is asymptomatic in approximately 75 to 95% of mothers. In some cases, it may manifest as a mild mononucleosis or flu-like syndrome with persistent fever and fatigue [49,168]. CMV is the most frequent cause of congenital infections, sensorineural hearing loss, and congenital malformations. Complications due to fetal CMV infection may also involve calcifications around the brain’s ventricles, enlargement of the cerebral ventricles, microcephaly, hepatosplenomegaly, cerebellar hypoplasia, restricted fetal growth, pleural effusion, and placental enlargement [166,168]. To prevent these issues, it is crucial to promptly conduct diagnosis. Furthermore, CMV infection continues to be a major concern in post-solid organ transplant (SOT), affecting both the allograft and recipient survival by causing CMV disease, drug-related side effects, bacterial and opportunistic infections, and transplant rejection. Moreover, it may negatively impact the quality of life for recipients of solid organ transplants [169].
In addition to plasma, where viremia and specific antibodies can be detected, CMV is often found in CSF and urine (Table 3). For these reasons, monitoring CMV viral load in these samples is necessary, particularly in critical care patients [170]. Many of the current techniques for CMV detection and quantification are either imprecise, have poor sensitivity, or require long processing times, as seen in ELISA serological analysis [171,172]. However, qPCR with automated DNA extraction has the potential to improve these deficiencies [173,174].

4.4.1. Detection of CMV in Oral, Urine, and Amniotic Fluid Samples

The standard method to diagnose CMV consists of virus isolation from urine or saliva in tissue cultures, but this procedure is labor-intensive, costly and not suitable for high-volume screening purposes. Recent large-scale studies have demonstrated that CMV real-time PCR performed on saliva is a sensitive, acceptable, and feasible method for hospital newborn screening [86,175] (Table 3). In fact, the rapid identification of newborns with CMV congenital infection after birth is crucial to promptly address possible CNS concerns, hearing loss, and developmental delays in order to perform proper diagnosis, monitoring, and intervention [176,177]. Urine samples are commonly utilized for congenital CMV screening infection because in these samples, infected infants often exhibit high virus levels [178]. In fact, Yamamoto et al. demonstrated that urine samples can be as effective as saliva samples in identifying CMV-infected infants through molecular screening programs [176]. Anyway, even if molecular assays are effective in detecting CMV in both saliva and urine during screening, these methods need to be further improved [176]. Another approach for the early identification of congenital CMV infection is the use of amniotic fluid. Amniocentesis is usually performed between 20 and 24 weeks of gestation when there is suspicion of CMV infection, such as when a fetal ultrasound shows signs consistent with infection. Amniotic fluid collected during amniocentesis can be analyzed by qPCR to allow for the early detection of CMV for a better management of affected pregnancies [178,179].

4.4.2. Detection of CMV in Blood Samples and Aqueous Humor

Recent studies have suggested that plasma-based PCR assays result in a delayed detection of CMV DNA due to the lower sensitivity of these assays compared to cell-based and whole-blood assays [179,180]. However, plasma does not require lengthy cell separation procedures and offers a much better opportunity to detect CMV viremia during periods of severe cytopenia when cell-based assays perform poorly. Plasma CMV viremia is associated with overall mortality in stem cell transplant recipients [181] and HIV-infected individuals [182]. Recently, the detection of CMV DNA in EDTA whole-blood samples has been suggested as the most sensitive method for monitoring CMV infection in immunosuppressed patients [183]. A high-sensitivity molecular assay may detect the latent virus, but the clinical relevance of low-level CMV DNA in whole blood is unresolved. Nevertheless, the early detection of CMV DNA has the advantage of warning clinicians of the evolution of infection and carefully monitoring viral load kinetics [184]. Razonable et al. reported the use of an automated qPCR to simultaneously measure the CMV DNA levels in whole blood (WB), plasma (PL), peripheral blood leukocytes (PBLs), and peripheral blood mononucleated cells (PBMCs). Higher levels of CMV DNA were observed in WB than PLs, and CMV DNA levels of PBLs and PBMCs were highly comparable. In spite of the adequacy of all blood compartments for CMV DNA quantification, the higher sensitivity of WB and its yield of higher CMV DNA render it an optimal sample for monitoring CMV DNA load during CMV disease, especially in immunocompromised patients [185]. The analysis of aqueous humor is essential for diagnosing ocular diseases associated with CMV. PCR is the primary method used to detect CMV DNA in aqueous humor. It offers high sensitivity and specificity. For example, studies have reported that PCR can detect CMV DNA in 93% of aqueous humor samples from eyes with active retinitis compared to 100% detection rates with next-generation sequencing [186,187] (Table 3).

4.5. Human Herpesvirus 6 (HHV-6)

HHV-6 belongs to the Betaherpesvirus subfamily (Table 2) [188]. HHV-6 is one of the most prevalent herpesviruses in humans. It spreads primarily through saliva, making close contact a common way of transmission. The diagnosis of HHV-6 infection can be performed by serology (indirect method) or real-time PCR, viral culture, in situ hybridization, and immunohistochemistry (direct methods). However, the most prominent technique is the quantification of viral DNA in blood, other body fluids, and organs using real-time PCR (Table 3). Although qPCR is useful for HHV-6 diagnosis and in determining viral load, serological tests, such as immunofluorescence and ELISA assays, have the potential to differentiate latent from lytic infection and can detect past exposure [88]. PCR-based methods are financially accessible, quick, safe, and currently widespread. In addition, these approaches readily permit the differentiation of the two species of HHV-6, even in cases of mixed infection [89].
Recently, HHV-6 has been subdivided into two different viruses, namely HHV-6A and -6B [189,190]. Even if the two viruses share about 90% of the genome, they have been differentiated based on their epidemiology, pathology association, in vitro growth properties, reactivity with monoclonal antibodies, and restriction endonuclease mapping [188,191]. For this reason, discrimination among the two HHV-6 viruses also represents a crucial clue in diagnosis.

4.5.1. HHV-6A and HHV-6B

HHV-6A and HHV-6B have distinct prevalence and clinical associations. HHV-6A is more common in specific geographic regions, such as sub-Saharan Africa [192], while HHV-6B, which is primarily linked to roseola infantum, a common childhood illness characterized by high fever followed by a rash [193], is diffused worldwide.
While HHV-6B is predominantly associated with childhood illnesses and is better understood, HHV-6A’s knowledge is more limited, even if its emerging link to serious neurological conditions highlights its significance in medical research [194]. HHV-6A has been described in association with neurological disorders, such as MS, systemic sclerosis (SSc), and Alzheimer’s disease [89,195,196]. Moreover, HHV-6A endometrial infection has also been described as potentially involved in women with unexplained infertility [197], which seems to be due to the modulation of natural killer cells by the virus [198]. Although both viruses can cause mild infections, HHV-6B may lead to more severe complications in immunocompromised patients, including encephalitis and organ inflammation [199]. Following initial infection, both viruses can enter a latent phase within the body via episome formation. Since both HHV-6A and HHV-6B possess telomere-like repeats at the terminal regions of their genomes, latency may also occur by integration into the host telomeres [51,199]. HHV-6B often integrates into the DNA of immune cells, particularly T-cells, which can subsequently reactivate, leading to symptoms such as a fever and rash. Understanding the behavior of these viruses, especially their latency and reactivation mechanisms, is crucial for managing their health impacts.
To date, several molecular assays are available to distinguish HHV-6A and HHV-6B in different clinical samples.

4.5.2. Chromosomally Integrated HHV-6

Chromosomally integrated HHV-6 (ciHHV-6) occurs when HHV-6 integrates its genetic material into the human genome, typically at chromosome telomeres. This condition differs from standard latent or active infections, as the viral DNA becomes a permanent part of the host’s chromosomes and can be inherited [199].
In fact, HHV-6 can integrate into the chromosomes of germline cells and be vertically transmitted [200]. The integrated viral genome is inherited in a Mendelian manner, meaning there is a 50% chance of passing it on to offspring. Consequently, individuals with ciHHV-6 may have one or more copies of the viral genome present in every nucleated cell in their body [201]. About 1% of the population carries ciHHV-6 [200], which results in high viral loads in blood tests, not due to active infection but due to the presence of integrated viral DNA in every cell.
Despite the majority of subjects carrying ciHHV-6 remaining asymptomatic, ongoing research is investigating the implications of its presence on health to improve clinical diagnostics [202,203]. ciHHV-6 diagnosis is primarily performed through PCR, where a persistently high viral load without symptoms may indicate ciHHV-6 [201]. There are concerns about the potential reactivation of the virus, especially in immunocompromised individuals. For instance, considering organ transplant patients, distinguishing ciHHV-6 from reactivated infections is crucial to avoid unnecessary treatments. The detection of ciHHV-6 poses challenges due to the presence of viral DNA in various body fluids, which does not reliably indicate active infection [199]. Reverse transcription PCR (RT-PCR) is effective for identifying active replication by detecting HHV-6 mRNA encoding structural proteins transcribed during the late phase of viral replication [204]. This qualitative assay is informative only during active viral replication, with a sensitivity of 90% and a specificity of 98% [194,205]. Real-time qPCR helps in measuring viral load to differentiate between primary and latent infections, although overlapping values can complicate interpretations [206]. A comprehensive diagnostic approach based on multiple assays has been developed to identify ciHHV-6 with high sensitivity [92,207,208,209,210,211]. Specifically, this is composed by qPCR to categorize viral load and, if necessary, RT-PCR to confirm active replication. Additionally, ddPCR has been created to detect ciHHV-6 with high sensitivity [92,207,208,209,210,211].

4.5.3. Detection of HHV-6 in Oral and Ocular Samples

A practical method to detect HHV-6 involves saliva sample collection using small filter paper strips followed by qPCR. The sensitivity and specificity of this technique are comparable to those of the standard method of saliva collection, even after prolonged drying of the specimens, without affecting the results [212]. Recent studies have shown that qualitative nested PCR of the HHV-6 genome derived from saliva is useful to distinguish among HHV-6A, HHV-6B, and chromosomally integrated viral DNA [213]. Besides nested PCR, other more advanced assays have been developed to differentiate between HHV-6A and HHV-6B in several fluids, including saliva. These specific PCR techniques can accurately identify both viruses even in mixed infections, with sensitivities as low as one copy per well for HHV-6B [93]. PCR-based assays, particularly multiplex PCR and nested PCR, have proven to be successful to detect HHV-6 also in ocular samples from patients with inflammatory disorders, suggesting its possible involvement in the development of inflammation in the eye [214].

4.5.4. Detection of HHV-6 in CSF Samples

As other HHVs, HHV-6 can also invade the CNS and persist longitudinally in the brain [215]. HHV-6 establishes latency in the nervous tissue, in particular, in several cell types including glial cells, oligodendrocytes, and astrocytes [52,53,54,55,188]. The detection of herpetic DNA, including the HHV-6 genome, in CSF by PCR is considered as a diagnostic marker of active infection in the CNS (Table 3) [216]. Recently, the use of multiplex PCR panels for HHV-6 detection in CFS was evaluated. Despite its advantages, this method can be challenging, as the detection of latency or subclinical HHV-6 reactivation may lead to incorrect assumptions of the cause of CNS disease, potentially harmful therapies, and delayed true diagnosis [217]. The interpretation of HHV-6 detection from CSF is further complicated by the fact that HHV-6 can exist as ciHHV-6. Recently, to improve the accuracy of identifying viral infections such as acute encephalitis and encephalopathy caused by HHV-6, next-generation sequencing has been implemented. NGS has been utilized for identifying viruses and finding new viruses in clinical specimens (Table 3). While it has been difficult to identify viral pathogens in patients with encephalitis/encephalopathy, multiple studies have demonstrated the effectiveness of NGS in identifying viral pathogens through the detection of virus-induced DNA or RNA sequences in CSF [218,219].

4.5.5. Detection of HHV-6 in Blood and Tissue Samples

The diagnosis of HHV-6 infection in blood samples poses a significant challenge. Conventional PCR assays for detecting viral DNA in peripheral blood leukocytes cannot distinguish between latent infection—present in the vast majority of healthy individuals—and active infection [220]. Only qPCR assays capable of detecting free HHV-6 DNA in biological fluids offer reliable tools for tracking active HHV-6 infection [194] (Table 3). However, the routine use of these assays in large epidemiological studies has been limited by their technical complexity and labor-intensive nature.
A novel highly sensitive qPCR test has been developed to precisely measure the levels of HHV-6 DNA in cell suspensions from tissues and body fluids. This assay employs a 5′ nuclease fluorogenic test with real-time monitoring of PCR amplification products using the ABI PRISM 7700 sequence detector system [206]. The sensitivity of this method matches that of a nested PCR protocol for both A and B subgroups of HHV-6. Additionally, the assay demonstrated a broader detection range (1 to 106 viral genome equivalents per test) and greater accuracy, repeatability, and reproducibility compared to a quantitative competitive PCR assay using the same reference DNA. This method is flexible and exhibits equal sensitivity and dynamic range when analyzing viral DNA from various fluids (e.g., culture medium or plasma) or cell suspensions obtained from tissues [201]. Nested PCR assays remain available for detecting HHV-6 viral DNA in a variety of body fluids and tissues, including plasma, peripheral blood mononuclear cells (PBMCs), and whole blood [213]. Recently, the ability to differentiate between HHV-6A and HHV-6B has been enhanced by ddPCR assays [196]. Furthermore, PCR-based techniques are also used to detect chromosomally integrated HHV-6 (ciHHV-6) in blood or other tissues (Table 3).

4.6. Human Herpesvirus 7 (HHV-7)

Human herpesvirus 7 (HHV-7) belongs to the Betaherpesvirus subfamily. It is genetically, epidemiologically, and clinically closely related to the more extensively studied HHV-6 [221]. HHV-7 primarily infects CD4+ T-lymphocytes, with CD8+ and immature T-cells being infected less commonly [55]. Upon the resolution of primary infection, HHV-7 establishes long-life latency in CD34+ hematopoietic progenitor stem cells (HPCs) [55]. HHV-7 is widely spread, and the initial infection typically occurs in young children, reaching its highest incidence between 18 and 36 months of age. In children, HHV-7 infection can manifest in various ways, including exanthema subitum, fever without rash, febrile seizures, and prolonged febrile seizures [222,223].
HHV-7 has also been reported to enter the CNS, although the mechanism by which it crosses the blood–brain barrier (BBB) remains unclear [224]. In the past, there have been reports of individual cases and groups of cases of encephalitis or encephalopathy related to HHV-7 in both immune-competent and immune-compromised children and adults [225]. The range of symptoms related to involving the CNS is varied and distinct from other neurological conditions. Symptoms can include febrile seizures, encephalitis, meningoencephalitis, facial palsy, vestibular neuritis, severe headache, drowsiness, fatigue, nausea, vomiting, sensitivity to light, loss of coordination, and unconsciousness [225,226,227,228].
HHV-7 is frequently linked to myalgic encephalomyelitis/chronic fatigue syndrome (ME/CFS), an illness characterized by long-lasting or repeated disabling fatigue [229]. Even though research has not identified one specific pathogen as the direct cause of ME/CFS [230], HHV-7 infection is commonly found in subjects with the condition. However, the HHV-7 DNA detection rate is similar in ME/CFS and healthy individuals [231]. Therefore, the connection between HHV-7 infection/reactivation and ME/CFS requires further investigation. While large-scale evidence is lacking, a recent study suggests that HHV-7 may also have the ability for chromosomal integration, sharing similar genomic regions to HHV-6, including direct repeats [229,231]. Prusty et al. explored the possibility of germ-line transmission of HHV-7 by developing a qPCR capable of detecting as few as 10 viral genome copies within complex human genomic DNA samples.

4.6.1. Detection of HHV-7 from Saliva

The detection of HHV-7 from saliva specimens has emerged as a promising approach to understand the virus’s role in various conditions, including ME/CFS [229] (Table 3). In a study involving participants with ME/CFS, saliva samples were collected monthly over six months, as well as during episodes of symptom exacerbation [229]. The viral load of HHV-7, along with other herpesviruses, was quantified using ddPCR, which allows for the highly sensitive detection of viral DNA [97]. The outcomes revealed that HHV-7 was detectable in saliva, with higher viral loads observed in patients with ME/CFS compared to healthy controls [97]. This correlation suggests that fluctuations in HHV-7 DNA levels may be associated with the severity of symptoms experienced by patients, highlighting the potential significance of saliva as a non-invasive diagnostic sample for monitoring herpesvirus reactivation and its implications in chronic illnesses.

4.6.2. Detection of HHV-7 from CSF

As mentioned above, HHV-7 can enter and infect the CNS. HHV-7 DNA was found in post-mortem brain tissue samples [224] and in the CSF of children with acute/subacute neurological symptoms using real-time PCR [221]. Yet, this approach fails to differentiate between the primary infection and subsequent viral reactivation or reinfection.
Primary HHV-7 infection typically happens in kids, often without or with mild symptoms, and is commonly linked to neurological issues like febrile seizures in children [223,232]. The presence of HHV-7 DNA in CSF has been observed in children with CNS diseases, indicating that neuroinvasion possibly occurs during initial infection [233,234]. In clinical settings, diagnosing a potential CNS disease due to HHV-7 infection typically relies on detecting HHV-7 DNA in CSF by qPCR. However, proving the diagnostic role of this amplification could be challenging, since HHV-7 reactivation in the CSF could be a side effect linked to other inflammatory or non-inflammatory CNS diseases. To overcome the limitations of conventional PCR, multiplex PCR can be employed for a more precise detection of HHV-7. Research by Leveque et al. showed that multiplex PCR is an accurate and specific method, enabling the rapid and simultaneous identification of HHV-7 from small volumes of CSF samples. This molecular diagnostic tool has the potential to enhance the standard virological diagnosis and treatment of CNS infections by identifying multiple viral infections that could lead to more severe cases of meningitis or encephalitis [235].

4.6.3. Detection of HHV-7 from Blood Samples

HHV-7 can be found in peripheral blood during initial infections and reactivations from latency [236]. Although various studies have confirmed the presence of HHV-7 DNA in peripheral blood lymphocytes (PBLs) using PCR, its clinical relevance remains uncertain because it is frequently detected in healthy individuals [236]. This highlights the need for improved molecular methods to detect HHV-7 in blood and plasma samples. While transmission through blood or blood products may not pose a significant risk to the general population due to widespread immunity, it could present potential dangers for young children, non-immune individuals, or immunocompromised patients [237]. Conventional PCR techniques, however, have limitations regarding sensitivity. Multiplex PCR offers a valuable alternative for screening blood donations for viral pathogens, including HHV-7. For example, a study by Zhang et al. demonstrated the effectiveness of a multiplex qPCR assay in detecting HHV-7 in blood samples from donors and patients with pityriasis rosea. In this study, 55% of blood donor samples tested positive for HHV-7, indicating a significant presence of the virus in the population. This method exhibited high specificity and sensitivity, establishing it as a reliable approach to ensuring blood safety [95].

4.7. Human Herpesvirus 8 (HHV-8)

HHV-8 belongs to the Alphaherpesvirus subfamily and is the first known member of the genus Rhadinovirus (Table 2) [238]. Through the use of classical epidemiological studies and laboratory techniques, scientists were able to associate this virus with Kaposi’s sarcoma (KS) [238,239], multicentric Castleman’s disease [240], primary effusion lymphoma (PEL) [241], and human immunodeficiency virus (HIV) [242]. HHV-8 establishes latency in B cells, and after primary infection, it can trigger symptomatic reactivations in immunocompromised patients [243,244]. The virus’s genetic material has been detected in various proportions in the oropharyngeal epithelium, saliva, semen, and blood, although its precise transmission mechanisms remain unclear [245,246,247,248].

4.7.1. Detection of HHV-8 from Oral Swabs and Saliva

The exact mechanism underlying the progression of HHV-8 infection to HHV-8-associated diseases remains unclear.
HHV-8 is most commonly detected in the oropharynx, with studies identifying its DNA in 15–57% of salivary samples from asymptomatic seropositive individuals [249]. These findings suggest that salivary transmission could represent a significant infection route and that oral replication likely serves as a critical reservoir contributing to systemic dissemination and, ultimately, the development of KS [249]. Taylor et al. found that 40% of the 1074 HHV-8 seropositive women enrolled in their study had HHV-8 DNA detected by PCR [250]. Specifically, the virus was found more frequently in samples taken from the mouth compared to samples taken from the genitals. Detection rates were 34% in oral specimens and 6% in genital specimens, with 32% of saliva samples and 28% of mouth swab samples testing positive for HHV-8 [250]. Furthermore, Derafshi et al. demonstrated that nested PCR or qPCR techniques can effectively amplify the highly conserved ORF73 gene in saliva samples to identify HHV-8 [100]. These findings indicate that oral specimens are effective in detecting HHV-8. However, further advancements in molecular methods are necessary to achieve a faster and more precise detection of HHV-8 infections.

4.7.2. Detection of HHV-8 from Semen and Blood Samples

HHV-8 is considered a significant risk factor for disease development only in the presence of additional cofactors, such as sexual transmission (e.g., concurrent HIV infection) or nonsexual routes [250,251]. In particular, HHV-8 DNA has been identified in sperm as well as in normal and malignant prostate tissues, reinforcing the sexual route as a critical mechanism for viral spread [252]. Recently, nested PCR demonstrated its potential for assessing HHV-8 DNA prevalence in semen samples (Table 2). Conversely, when analyzing blood samples, the SYBR Green qPCR method is preferred for detecting and quantifying HHV-8 DNA [100,251].

5. Emerging Molecular Approaches and Future Perspectives

To date, several DNA-based methods are already used in the diagnosis of viral infections, such as the detection and genotyping of human papillomaviruses (HPVs) and members of the Herpesviridae family, including HHV-6, HHV-8, VZV, and CMV [253,254,255,256,257].
Recent advancements in molecular biology have significantly enhanced the design of new assays for detecting and diagnosing herpesviruses. This review explores the latest DNA-based technologies shaping the future of herpesvirus detection.
Currently, many molecular assays for virus detection rely on PCR [258,259]. They provide sample-to-result workflows, requiring minimal sample preparation, offering fast and simple reports, and demanding limited interpretation by laboratory personnel with minimal technical expertise [260].
However, existing methods for detecting viral nucleic acids, such as droplet digital PCR and other digital PCR technologies, face limitations that hinder their broad application in clinical settings. While ddPCR offers significant advantages in sensitivity and specificity, the high cost of equipment and reagents can be a substantial barrier [14]. This high cost may limit access for smaller laboratories, especially in developing countries or those in resource-limited settings [261], where ddPCR is less feasible for routine use compared to conventional PCR methods [262]. Although ddPCR simplifies some aspects of viral nucleic acid quantification by eliminating the need for standard curves, the overall workflow remains complex, introducing operational challenges, such as errors during sample preparation and the need for specialized training [14,263]. To address these limitations, ongoing research and recent innovations focus on enhancing the accessibility and efficiency of ddPCR and related technologies. Efforts include developing more affordable reagents, optimizing protocols to streamline workflows, creating portable ddPCR systems for diverse settings like point-of-care diagnostics, and integrating microfluidics and automation to simplify procedures [14].
Among the most promising technologies for improving infection diagnosis and control is NGS. When applied to whole-genome sequencing (WGS), NGS has demonstrated significant advancements in epidemiology, phylogenetics, virulence determinants, and the evolution of antimicrobial resistance [264]. However, barriers to the widespread adoption of NGS in infection control include concerns over data quality and the effective implementation of bioinformatics in clinical laboratories [265,266].
Emerging technologies like biosensors are transforming viral diagnostics. These devices detect biological analytes, including viruses, by converting biological interactions into measurable electrical signals [267]. Their rapid response times, sensitivity, and portability make them ideal for point-of-care diagnostics. Innovations in DNA nanotechnology and aptamer-based detection further enhance their specificity and versatility, promising significant advancements in viral diagnostics. Biosensors offer substantial advantages over traditional diagnostic methods, including rapid results, high sensitivity, and the potential for point-of-care testing.
Different biosensor types have been utilized for virus detection. For example, electrochemical biosensors exploit electrochemical reactions to detect viral components like proteins or nucleic acids. These sensors can achieve extremely low detection limits (femtomolar concentrations) and are frequently used for viruses such as HIV and SARS-CoV-2 [267]. Moreover, the recent integration of nanomaterials enhances their sensitivity and specificity [268]. Optical biosensors, which employ mechanisms such as surface plasmon resonance (SPR) or surface-enhanced Raman scattering (SERS), use light interactions with viral particles for real-time detection [269]. These methods are highly sensitive and can detect viruses without the need for specific labeling. For instance, plasmonic biosensors provide cost-effective and rapid virus identification due to their tunable properties [270].
The most innovative biosensor-based devices available are nanobiosensors. These biosensors employ nanomaterials like gold nanoparticles and graphene quantum dots to improve performance [271]. These materials improve sensitivity and specificity by providing a high surface area for interactions with viral targets. Nanobiosensors are also highly miniaturizable, making them suitable for diverse settings [23,270].
Although DNA nanotechnology-based biosensors show significant potential for virus identification, further studies are required in this emerging area of research [272,273].
For example, CRISPR-based diagnostics are evolving, with ongoing research addressing detection limitations. Innovations include developing PAM-independent systems [274], exploring alternative Cas proteins [274], exploring other alternative Cas proteins (e.g., Cas14) [275,276], and integrating metagenomics [277]. These advancements aim to simplify workflows, enable the multiplex detection of multiple pathogens, and reduce contamination risks, thereby improving the robustness of CRISPR in diagnostics.
Despite these advancements, universal detection across diverse herpesvirus species remains challenging. The need for high specificity and sensitivity, reducing direct and indirect associated costs, and the importance to improve consistency in clinical diagnostics by standardizing DNA-based herpesvirus detection methods across laboratories continue to drive research on novel molecular assays capable of adapting to emerging viral strains and facilitate point-of-care testing.

6. Conclusions

In this review, we reported how DNA-based molecular technologies have evolved for the identification of viruses, focusing on herpesvirus detection. Methods for identifying herpesviruses primarily rely on PCR-based or related techniques, which enable the amplification of viral DNA fragments from various samples with high specificity and sensitivity.
Advanced techniques, such as ddPCR and biosensors, have significantly improved the management of herpesvirus-related diseases, enabling faster interventions and delivering more accurate prognoses. These new laboratory tools offer the ability to detect latent infections, promising not only faster and more precise diagnostics but also deeper insights into herpesvirus epidemiology and pathogenesis.
Despite the high diagnostic performance achieved, challenges remain. Enhancing sensitivity, reducing costs, and enabling broader accessibility—particularly in low-resource settings—remain critical areas of focus in clinical research. Addressing these challenges will be essential to expanding the global impact of these molecular diagnostic tools on public health.

Author Contributions

Conceptualization, D.B. and V.G.; data curation, G.M. and G.C.; writing—original draft, G.M., G.C. and M.F.; writing—review and editing, D.B. and V.G.; supervision, D.B. All authors have read and agreed to the published version of the manuscript.

Funding

This work was supported by Unife FAR (2023-FAR.L_chimiche_BD_001 to D.B.).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Acknowledgments

We thank Iva Pivanti and Niccolò Caivano for their technical and conceptual support.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Taylor, T.J.; Brockman, M.A.; McNamee, E.E.; Knipe, D.M. Herpes simplex virus. Front. Biosci. 2002, 7, 752–764. [Google Scholar] [CrossRef] [PubMed]
  2. Cassedy, A.; Parle-McDermott, A.; O’Kennedy, R. Virus detection: A review of the current and emerging molecular and immunological methods. Front. Mol. Biosci. 2001, 8, 637559. [Google Scholar] [CrossRef]
  3. Whitley, R.J.; Roizman, B.J.T.L. Herpes simplex virus infections. Lancet 2001, 357, 1513–1518. [Google Scholar] [CrossRef] [PubMed]
  4. De Francesco, M.A.J.V. Herpesviridae, Neurodegenerative Disorders and Autoimmune Diseases: What Is the Relationship between Them? Viruses 2024, 16, 133. [Google Scholar] [CrossRef]
  5. Nath, P.; Kabir, A.; Doust, S.K.; Ray, A. Diagnosis of herpes simplex virus: Laboratory and point-of-care techniques. Infect. Dis. Rep. 2021, 13, 518–539. [Google Scholar] [CrossRef] [PubMed]
  6. LeGoff, J.; Péré, H.; Bélec, L. Diagnosis of genital herpes simplex virus infection in the clinical laboratory. Virol. J. 2014, 11, 83. [Google Scholar] [CrossRef] [PubMed]
  7. Morshed, M.G.; Lee, M.K.; Jorgensen, D.; Isaac-Renton, J.L. Molecular methods used in clinical laboratory: Prospects and pitfalls. FEMS Immunol. Med. Microbiol. 2007, 49, 184–191. [Google Scholar] [CrossRef] [PubMed]
  8. Navarro, E.; Serrano-Heras, G.; Castaño, M.J.; Solera, J.J.C.C.A. Real-time PCR detection chemistry. Clin. Chim. Acta 2015, 439, 231–250. [Google Scholar] [CrossRef] [PubMed]
  9. Broude, N.E.; Zhang, L.; Woodward, K.; Englert, D.; Cantor, C.R. Multiplex allele-specific target amplification based on PCR suppression. Proc. Natl. Acad. Sci. USA 2001, 98, 206–211. [Google Scholar] [CrossRef] [PubMed]
  10. Elnifro, E.M.; Ashshi, A.M.; Cooper, R.J.; Klapper, P.E. Multiplex PCR: Optimization and application in diagnostic virology. Clin. Microbiol. Rev. 2000, 13, 559–570. [Google Scholar] [CrossRef]
  11. Poritz, M.A.; Blaschke, A.J.; Byington, C.L.; Allen, L.; Nilsson, K.; Jones, D.E.; Thatcher, S.A.; Robbins, T.; Lingenfelter, B.; Amiott, E.; et al. FilmArray, an automated nested multiplex PCR system for multi-pathogen detection: Development and application to respiratory tract infection. PLoS ONE 2011, 6, e26047. [Google Scholar] [CrossRef]
  12. Li, H.; Bai, R.; Zhao, Z.; Tao, L.; Ma, M.; Ji, Z.; Jian, M.; Ding, Z.; Dai, X.; Bao, F.; et al. Application of droplet digital PCR to detect the pathogens of infectious diseases. Biosci. Rep. 2018, 38, BSR20181170. [Google Scholar] [CrossRef] [PubMed]
  13. Taylor, S.C.; Laperriere, G.; Germain, H. Droplet Digital PCR versus qPCR for gene expression analysis with low abundant targets: From variable nonsense to publication quality data. Sci. Rep. 2017, 7, 2409. [Google Scholar] [CrossRef] [PubMed]
  14. Kojabad, A.A.; Farzanehpour, M.; Galeh, H.E.G.; Dorostkar, R.; Jafarpour, A.; Bolandian, M.; Nodooshan, M.M. Droplet digital PCR of viral DNA/RNA, current progress, challenges, and future perspectives. J. Med. Virol. 2021, 93, 4182–4197. [Google Scholar] [CrossRef]
  15. Notomi, T.; Mori, Y.; Tomita, N.; Kanda, H. Loop-mediated isothermal amplification (LAMP): Principle, features, and future prospects. J. Microbiol. 2015, 53, 1–5. [Google Scholar] [CrossRef]
  16. Kurosaki, Y.; Martins, D.B.G.; Kimura, M.; Catena, A.D.S.; Borba, M.A.C.S.M.; Mattos, S.D.S.; Abe, H.; Yoshikawa, R.; de Lima Filho, J.L.; Yasuda, J. Development and evaluation of a rapid molecular diagnostic test for Zika virus infection by reverse transcription loop-mediated isothermal amplification. Sci. Rep. 2017, 7, 13503. [Google Scholar] [CrossRef] [PubMed]
  17. Francois, P.; Tangomo, M.; Hibbs, J.; Bonetti, E.J.; Boehme, C.C.; Notomi, T.; Perkins, M.D.; Schrenzel, J. Robustness of a loop-mediated isothermal amplification reaction for diagnostic applications. FEMS Immunol. Med. Microbiol. 2011, 62, 41–48. [Google Scholar] [CrossRef]
  18. Silva, S.J.R.D.; Paiva, M.H.S.; Guedes, D.R.D.; Krokovsky, L.; Melo, F.L.D.; Silva, M.A.L.D.; Silva, A.; Ayres, C.F.J.; Pena, L.J. Development and validation of reverse transcription loop-mediated isothermal amplification (RT-LAMP) for rapid detection of ZIKV in mosquito samples from Brazil. Sci. Rep. 2019, 9, 4494. [Google Scholar] [CrossRef] [PubMed]
  19. Jeba, J.M.P.; Deepalakshmi, P. Selection of Robust Feature Selection Methods Used for Gene Expression Analysis of Microarray Data. In Proceedings of the 2024 IEEE 5th International Conference on Image Processing and Capsule Networks (ICIPCN), Dhulikhel, Nepal, 3–4 July 2024; pp. 918–924. [Google Scholar]
  20. Földes-Papp, Z.; Egerer, R.; Birch-Hirschfeld, E.; Striebel, H.-M.; Demel, U.; Tilz, G.P.; Wutzler, P. Detection of multiple human herpes viruses by DNA microarray technology. Mol. Diagn. 2004, 8, 1–9. [Google Scholar] [CrossRef]
  21. Matteoli, B.; Ceccherini-Nelli, L. Viral DNA and cDNA Array in the Diagnosis of Respiratory Tract Infections. In Biomedical Tissue Culture; IntechOpen: Rijeka, Croatia, 2012. [Google Scholar]
  22. Bonde, J.; Rebolj, M.; Ejegod, D.M.; Preisler, S.; Lynge, E.; Rygaard, C. HPV prevalence and genotype distribution in a population-based split-sample study of well-screened women using CLART HPV2 human papillomavirus genotype microarray system. BMC Infect. Dis. 2014, 14, 413. [Google Scholar] [CrossRef]
  23. Shand, H.; Dutta, S.; Rajakumar, S.; James Paulraj, S.; Mandal, A.K.; KT, R.D.; Ghorai, S. New age detection of viruses: The nano-biosensors. Front. Nanotechnol. 2022, 3, 814550. [Google Scholar] [CrossRef]
  24. Zhai, J.; Cui, H.; Yang, R. DNA based biosensors. Biotechnol. Adv. 1997, 15, 43–58. [Google Scholar] [CrossRef]
  25. Yuwen, L.; Zhang, S.; Chao, J. Recent advances in DNA nanotechnology-enabled biosensors for virus detection. Biosensors 2023, 13, 822. [Google Scholar] [CrossRef]
  26. Chakraborty, B.; Das, S.; Gupta, A.; Xiong, Y.; TV, V.; Kizer, M.E.; Duan, J.; Chandrasekaran, A.R.; Wang, X. Aptamers for viral detection and inhibition. ACS Infect. Dis. 2022, 8, 667–692. [Google Scholar] [CrossRef]
  27. Song, J.; Cha, B.; Moon, J.; Jang, H.; Kim, S.; Jang, J.; Yong, D.; Kwon, H.J.; Lee, I.C.; Lim, E.K.; et al. Smartphone-based SARS-CoV-2 and variants detection system using colorimetric DNAzyme reaction triggered by loop-mediated isothermal amplification (LAMP) with clustered regularly interspaced short palindromic repeats (CRISPR). ACS Nano 2022, 16, 11300–11314. [Google Scholar] [CrossRef]
  28. Li, L.; Li, S.; Wu, N.; Wu, J.; Wang, G.; Zhao, G.; Wang, J. HOLMESv2: A CRISPR-Cas12b-assisted platform for nucleic acid detection and DNA methylation quantitation. ACS Synth. Biol. 2019, 8, 2228–2237. [Google Scholar] [CrossRef]
  29. Zhang, Y.; Odiwuor, N.; Xiong, J.; Sun, L.; Nyaruaba, R.O.; Wei, H.; Tanner, N.A. Rapid molecular detection of SARS-CoV-2 (COVID-19) virus RNA using colorimetric LAMP. MedRxiv 2020. [Google Scholar] [CrossRef]
  30. Chen, J.S.; Ma, E.; Harrington, L.B.; Da Costa, M.; Tian, X.; Palefsky, J.M.; Doudna, J.A. CRISPR-Cas12a target binding unleashes indiscriminate single-stranded DNase activity. Science 2018, 360, 436–439. [Google Scholar] [CrossRef]
  31. Li, S.Y.; Cheng, Q.X.; Liu, J.K.; Nie, X.Q.; Zhao, G.P.; Wang, J. CRISPR-Cas12a has both cis-and trans-cleavage activities on single-stranded DNA. Cell Res. 2018, 28, 491–493. [Google Scholar] [CrossRef] [PubMed]
  32. Kazim, I.; Gande, T.; Reyher, E.; Bhutia, K.G.; Dhingra, K.; Verma, S. Advancements in sequencing technologies: From genomic revolution to single-cell insights in precision medicine. J. Knowl. Learn. Sci. Technol. 2024, 3, 108–124. [Google Scholar] [CrossRef]
  33. Datta, S.; Budhauliya, R.; Das, B.; Chatterjee, S.; Veer, V. Next-generation sequencing in clinical virology: Discovery of new viruses. World J. Virol. 2015, 4, 265. [Google Scholar] [CrossRef] [PubMed]
  34. Sip, M.; Bystricka, D.; Kmoch, S.; Noskova, L.; Hartmannova, H.; Dedic, P. Detection of viral infections by an oligonucleotide microarray. J. Virol. Methods 2010, 165, 64–70. [Google Scholar] [CrossRef] [PubMed]
  35. Satam, H.; Joshi, K.; Mangrolia, U.; Waghoo, S.; Zaidi, G.; Rawool, S.; Thakare, R.P.; Banday, S.; Mishra, A.K.; Das, G.; et al. Next-generation sequencing technology: Current trends and advancements. Biology 2023, 12, 997. [Google Scholar] [CrossRef]
  36. Ilié, M.; Benzaquen, J.; Hofman, V.; Long-Mira, E.; Lassalle, S.; Boutros, J.; Bontoux, C.; Lespinet-Fabre, V.; Bordone, O.; Tanga, V.; et al. Accurate Detection of SARS-CoV-2 by Next-Generation Sequencing in Low Viral Load Specimens. Int. J. Mol. Sci. 2023, 24, 3478. [Google Scholar] [CrossRef] [PubMed]
  37. Visser, M.; Bester, R.; Burger, J.T.; Maree, H.J. Next-generation sequencing for virus detection: Covering all the bases. Virol. J. 2016, 13, 85. [Google Scholar] [CrossRef] [PubMed]
  38. Rechenchoski, D.Z.; Faccin-Galhardi, L.C.; Linhares, R.E.C.; Nozawa, C. Herpesvirus: An underestimated virus. Folia Microbiol. 2017, 62, 151–156. [Google Scholar] [CrossRef]
  39. Knipe, D.M.; Cliffe, A. Chromatin control of herpes simplex virus lytic and latent infection. Nat. Rev. Microbiol. 2008, 6, 211–221. [Google Scholar] [CrossRef] [PubMed]
  40. Mettenleiter, T.C.; Ehlers, B.; Müller, T.; Yoon, K.J.; Teifke, J.P. Herpesviruses. In Diseases of Swine; Wiley-Blackwell: Hoboken, NJ, USA, 2019; pp. 548–575. [Google Scholar]
  41. Kukhanova, M.; Korovina, A.; Kochetkov, S.J.B. Human herpes simplex virus: Life cycle and development of inhibitors. Biochemistry 2014, 79, 1635–1652. [Google Scholar] [CrossRef]
  42. Roizman, B.; Pellett, P.E. The family Herpesviridae: A brief introduction. In Fields Virology, 4th ed.; Knipe, D.M., Howley, P.M., Eds.; Lippincott Williams & Wilkins: Philadelphia, PA, USA, 2001; Volume 2, pp. 2381–2397. [Google Scholar]
  43. Gershon, A.A.; Breuer, J.; Cohen, J.I.; Cohrs, R.J.; Gershon, M.D.; Gilden, D.; Grose, C.; Hambleton, S.; Kennedy, P.G.E.; Oxman, M.N.; et al. Varicella zoster virus infection. Nat. Rev. Dis. Primers 2015, 1, 15016. [Google Scholar] [CrossRef] [PubMed]
  44. Schelhaas, M.; Jansen, M.; Haase, I.; Knebel-Mörsdorf, D. Herpes simplex virus type 1 exhibits a tropism for basal entry in polarized epithelial cells. J. Gen. Virol. 2003, 84, 2473–2484. [Google Scholar] [CrossRef] [PubMed]
  45. Gupta, R.; Warren, T.; Wald, A. Genital herpes. Lancet 2007, 370, 2127–2137. [Google Scholar] [CrossRef]
  46. Gilden, D.H.; Vafai, A.; Shtram, Y.; Becker, Y.; Devlin, M.; Wellish, M. Varicella-zoster virus DNA in human sensory ganglia. Nature 1983, 306, 478–480. [Google Scholar] [CrossRef] [PubMed]
  47. Laing, K.J.; Ouwendijk, W.J.D.; Koelle, D.M.; Verjans, G.M.G.M. Immunobiology of Varicella-Zoster Virus Infection. J. Infect. Dis. 2018, 218 (Suppl. S2), S68–S74. [Google Scholar] [CrossRef] [PubMed]
  48. Hummel, M.; Abecassis, M.M. A model for reactivation of CMV from latency. J. Clin. Virol. 2002, 25, 123–136. [Google Scholar] [CrossRef]
  49. Leng, S.X.; Kamil, J.; Purdy, J.G.; Lemmermann, N.A.; Reddehase, M.J.; Goodrum, F.D. Recent advances in CMV tropism, latency, and diagnosis during aging. GeroScience 2017, 39, 251–259. [Google Scholar] [CrossRef]
  50. Liu, X.F.; Wang, X.; Yan, S.; Zhang, Z.; Abecassis, M.; Hummel, M. Epigenetic control of cytomegalovirus latency and reactivation. Viruses 2013, 5, 1325–1345. [Google Scholar] [CrossRef]
  51. Pantry, S.N.; Medveczky, P.G.J.V. Latency, integration, and reactivation of human herpesvirus-6. Viruses 2017, 9, 194. [Google Scholar] [CrossRef] [PubMed]
  52. Hannolainen, L.; Pyöriä, L.; Pratas, D.; Lohi, J.; Skuja, S.; Rasa-Dzelzkaleja, S.; Murovska, M.; Hedman, K.; Jahnukainen, T.; Perdomo, M.F. Reactivation of a Transplant Recipient’s Inherited Human Herpesvirus 6 and Implications to the Graft. J. Infect. Dis. 2024, jiae268. [Google Scholar] [CrossRef]
  53. Rotola, A.; Ravaioli, T.; Gonelli, A.; Dewhurst, S.; Cassai, E.; Di Luca, D. U94 of human herpesvirus 6 is expressed in latently infected peripheral blood mononuclear cells and blocks viral gene expression in transformed lymphocytes in culture. Proc. Natl. Acad. Sci. USA 1998, 95, 13911–13916. [Google Scholar] [CrossRef]
  54. Lusso, P.; Markham, P.D.; Tschachler, E.; Veronese, F.d.M.; Salahuddin, S.Z.; Ablashi, D.V.; Pahwa, S.; Krohn, K.; Gallo, R.C. In vitro cellular tropism of human B-lymphotropic virus (human herpesvirus-6). J. Exp. Med. 1988, 167, 1659–1670. [Google Scholar] [CrossRef]
  55. Berneman, Z.N.; Ablashi, D.V.; Li, G.; Eger-Fletcher, M.; Reitz, M.S., Jr.; Hung, C.L.; Brus, I.; Komaroff, A.L.; Gallo, R.C. Human herpesvirus 7 is a T-lymphotropic virus and is related to, but significantly different from, human herpesvirus 6 and human cytomegalovirus. Proc. Natl. Acad. Sci. USA 1992, 89, 10552–10556. [Google Scholar] [PubMed]
  56. Corey, L.; Brodie, S.; Huang, M.; Koelle, D.M.; Wald, A. HHV-8 infection: A model for reactivation and transmission. Rev. Med. Virol. 2002, 12, 47–63. [Google Scholar] [CrossRef] [PubMed]
  57. Ueda, K. KSHV genome replication and maintenance in latency. Adv. Exp. Med. Biol. 2018, 1045, 299–320. [Google Scholar]
  58. Dunmire, S.K.; Verghese, P.S.; Balfour, H.H. Primary Epstein-Barr virus infection. J. Clin. Virol. 2018, 102, 84–92. [Google Scholar] [CrossRef] [PubMed]
  59. Guo, R.; Liang, J.H.; Zhang, Y.; Lutchenkov, M.; Li, Z.; Wang, Y.; Trujillo-Alonso, V.; Puri, R.; Giulino-Roth, L.; Gewurz, B.E. Methionine metabolism controls the B cell EBV epigenome and viral latency. Cell Metab. 2022, 34, 1280–1297.e9. [Google Scholar] [CrossRef] [PubMed]
  60. Jeffery-Smith, A.; Riddell, A. Herpesviruses. Medicine 2021, 49, 780–784. [Google Scholar] [CrossRef]
  61. Whitley, R.J. Herpesviruses. In Medical Microbiology, 4th ed.; University of Texas Medical Branch at Galveston: Galveston, TX, USA, 1996. [Google Scholar]
  62. Roizman, B.; Whitley, R.J. An inquiry into the molecular basis of HSV latency and reactivation. Annu. Rev. Microbiol. 2013, 67, 355–374. [Google Scholar] [CrossRef] [PubMed]
  63. Crawford, L.B.; Microbiology, I. Hematopoietic stem cells and betaherpesvirus latency. Front. Cell Infect. Microbiol. 2023, 13, 1189805. [Google Scholar] [CrossRef]
  64. Arvin, A.; Campadelli-Fiume, G.; Mocarski, E.; Moore, P.S.; Roizman, B.; Whitley, R.; Yamanishi, K. Human Herpesviruses: Biology, Therapy, and Immunoprophylaxis; Cambridge University Press: Cambridge, UK, 2007. [Google Scholar]
  65. Kessler, H.H.; Mühlbauer, G.; Rinner, B.; Stelzl, E.; Berger, A.; Dörr, H.-W.; Santner, B.; Marth, E.; Rabenau, H. Detection of herpes simplex virus DNA by real-time PCR. J. Clin. Microbiol. 2000, 38, 2638–2642. [Google Scholar] [CrossRef]
  66. Lopez Roa, P.; Alonso, R.; de Egea, V.; Usubillaga, R.; Muñoz, P.; Bouza, E. PCR for detection of herpes simplex virus in cerebrospinal fluid: Alternative acceptance criteria for diagnostic workup. J. Clin. Microbiol. 2013, 51, 2880–2883. [Google Scholar] [CrossRef] [PubMed]
  67. Druce, J.; Catton, M.; Chibo, D.; Minerds, K.; Tyssen, D.; Kostecki, R.; Maskill, B.; Leong-Shaw, W.; Gerrard, M.; Birch, C. Utility of a multiplex PCR assay for detecting herpesvirus DNA in clinical samples. J. Clin. Microbiol. 2002, 40, 1728–1732. [Google Scholar] [CrossRef]
  68. Xiu, L.; Zhang, C.; Li, Y.; Wang, F.; Peng, J. Simultaneous detection of eleven sexually transmitted agents using multiplexed PCR coupled with MALDI-TOF analysis. Infect. Drug Resist. 2019, 12, 2671–2682. [Google Scholar] [CrossRef]
  69. Zhu, X.; Liu, P.; Lu, L.; Zhong, H.; Xu, M.; Jia, R.; Su, L.; Cao, L.; Sun, Y.; Guo, M.; et al. Development of a multiplex droplet digital PCR assay for detection of enterovirus, parechovirus, herpes simplex virus 1 and 2 simultaneously for diagnosis of viral CNS infections. Virol. J. 2022, 19, 70. [Google Scholar] [CrossRef]
  70. Liu, W.; Wang, C.; Pan, F.; Shao, J.; Cui, Y.; Han, D.; Zhang, H. Clinical application of a multiplex droplet digital PCR in the rapid diagnosis of children with suspected bloodstream infections. Pathogens 2023, 12, 719. [Google Scholar] [CrossRef] [PubMed]
  71. Shi, L.; Xia, H.; Moore, M.D.; Deng, C.; Li, N.; Ren, H.; Chen, Y.; Liu, J.; Du, F.; Zheng, G.; et al. Metagenomic next-generation sequencing in the diagnosis of HHV-1 reactivation in a critically ill COVID-19 patient: A case report. Front. Med. 2021, 8, 715519. [Google Scholar] [CrossRef] [PubMed]
  72. Modhusudon, S.; Bithi, R.; Islam, M.A.J.F. Detection of herpes simplex virus 2: A SYBR-Green-based real-time PCR assay. F1000Res 2021, 10, 655. [Google Scholar]
  73. Liu, J.; Yi, Y.; Chen, W.; Si, S.; Yin, M.; Jin, H.; Liu, J.; Zhou, J.; Zhang, J. Development and evaluation of the quantitative real-time PCR assay in detection and typing of herpes simplex virus in swab specimens from patients with genital herpes. Int. J. Clin. Exp. Med. 2015, 8, 18758. [Google Scholar]
  74. Navti, O.B.; Al-Belushi, M.; Konje, J.C. Cytomegalovirus infection in pregnancy—An update. Eur. J. Obstet. Gynecol. Reprod. Biol. 2021, 258, 216–222. [Google Scholar] [CrossRef] [PubMed]
  75. Nahass, G.T.; Mandel, M.J.; Cook, S.; Fan, W.; Leonardi, C.L. Detection of herpes simplex and varicella-zoster infection from cutaneous lesions in different clinical stages with the polymerase chain reaction. J. Am. Acad. Dermatol. 1995, 32, 730–733. [Google Scholar] [CrossRef]
  76. Fan, F.; Stiles, J.; Mikhlina, A.; Lu, X.; Babady, N.E.; Tang, Y.-W. Clinical validation of the Lyra direct HSV 1 + 2/VZV assay for simultaneous detection and differentiation of three herpesviruses in cutaneous and mucocutaneous lesions. J. Clin. Microbiol. 2014, 52, 3799–3801. [Google Scholar] [CrossRef]
  77. Guo, M.; Deng, L.; Liang, H.; Du, Y.; Gao, W.; Tian, N.; Bi, Y.; Li, J.; Ma, T.; Zhang, Y.; et al. Development and preliminary application of a droplet digital PCR assay for quantifying the oncolytic herpes simplex virus type 1 in the clinical-grade production. Viruses 2023, 15, 178. [Google Scholar] [CrossRef] [PubMed]
  78. Si, Z.; Li, L.; Han, J. Analysis of metagenomic next-generation sequencing (mNGS) in the diagnosis of herpes simplex virus (HSV) encephalitis with normal cerebrospinal fluid (CSF). Infect. Drug Resist. 2023, 16, 3431–3439. [Google Scholar] [CrossRef] [PubMed]
  79. Hartung, J.; Enders, G.; Chaoui, R.; Arents, A.; Tennstedt, C.; Bollmann, R. Prenatal diagnosis of congenital varicella syndrome and detection of varicella-zoster virus in the fetus: A case report. Prenat. Diagn. 1999, 19, 163–166. [Google Scholar] [CrossRef]
  80. Zhu, Y.; Xu, M.; Ding, C.; Peng, Z.; Wang, W.; Sun, B.; Cheng, J.; Chen, C.; Chen, W.; Wei, H.; et al. Metagenomic next-generation sequencing vs. traditional microbiological tests for diagnosing varicella-zoster virus central nervous system infection. Front. Public Health 2022, 9, 738412. [Google Scholar] [CrossRef]
  81. Han, J.; Si, Z.; Wei, N.; Cao, D.; Ji, Y.; Kang, Z.; Zhu, J. Next-generation sequencing of cerebrospinal fluid for the diagnosis of VZV-associated rhombencephalitis. J. Integr. Neurosci. 2023, 22, 36. [Google Scholar] [CrossRef]
  82. Farisyi, M.A.; Sufiawati, I.J.O.D. Detection of Epstein–Barr virus DNA in saliva of HIV-1-infected individuals with oral hairy leukoplakia. Oral Dis. 2020, 26, 158–160. [Google Scholar] [CrossRef]
  83. Idesawa, M.; Sugano, N.; Ikeda, K.; Oshikawa, M.; Takane, M.; Seki, K.; Ito, K. Detection of Epstein–Barr virus in saliva by real-time PCR. Oral Microbiol. Immunol. 2004, 19, 230–232. [Google Scholar] [CrossRef] [PubMed]
  84. Kim, K.Y.; Le, Q.-T.; Yom, S.S.; Pinsky, B.A.; Bratman, S.V.; Ng, R.H.W.; El Mubarak, H.S.; Chan, K.C.A.; Sander, M.; Conley, B.A. Current state of PCR-based Epstein-Barr virus DNA testing for nasopharyngeal cancer. J. Natl. Cancer Inst. 2017, 109, djx007. [Google Scholar] [CrossRef]
  85. Wang, S.; Xiong, H.; Yan, S.; Wu, N.; Lu, Z. Identification and characterization of Epstein-Barr virus genomes in lung carcinoma biopsy samples by next-generation sequencing technology. Sci. Rep. 2016, 6, 26156. [Google Scholar] [CrossRef] [PubMed]
  86. Ross, S.A.; Ahmed, A.; Palmer, A.L.; Michaels, M.G.; Sánchez, P.J.; Bernstein, D.I.; Tolan, R.W., Jr.; Novak, Z.; Chowdhury, N.; Fowler, K.B.; et al. Detection of congenital cytomegalovirus infection by real-time polymerase chain reaction analysis of saliva or urine specimens. J. Infect. Dis. 2014, 210, 1415–1418. [Google Scholar]
  87. Mallory, M.A.; Hymas, W.C.; Simmon, K.E.; Pyne, M.T.; Stevenson, J.B.; Barker, A.P.; Hillyard, D.R.; Hanson, K.E. Development and validation of a next-generation sequencing assay with open-access analysis software for detecting resistance-associated mutations in CMV. J. Clin. Microbiol. 2023, 61, e00829-23. [Google Scholar] [CrossRef]
  88. Al-Sadeq, D.W.; Zedan, H.T.; Aldewik, N.; Elkhider, A.; Hicazi, A.; Younes, N.; Ayoub, H.H.; Abu Raddad, L.; Yassine, H.M.; Nasrallah, G.K. Human herpes simplex virus-6 (HHV-6) detection and seroprevalence among Qatari nationals and immigrants residing in Qatar. IJID Reg. 2021, 2, 90–95. [Google Scholar] [CrossRef] [PubMed]
  89. Boutolleau, D.; Duros, C.; Bonnafous, P.; Caïola, D.; Karras, A.; De Castro, N.; Ouachée, M.; Narcy, P.; Gueudin, M.; Agut, H.; et al. Identification of human herpesvirus 6 variants A and B by primer-specific real-time PCR may help to revisit their respective role in pathology. J. Clin. Virol. 2006, 35, 257–263. [Google Scholar] [CrossRef] [PubMed]
  90. Kidd, I.; A Clark, D.; Bremner, J.A.; Pillay, D.; Griffiths, P.D.; Emery, V.C. A multiplex PCR assay for the simultaneous detection of human herpesvirus 6 and human herpesvirus 7, with typing of HHV-6 by enzyme cleavage of PCR products. J. Virol. Methods 1998, 70, 29–36. [Google Scholar] [CrossRef]
  91. Agut, H.; Bonnafous, P.; Gautheret-Dejean, A. Human Herpesviruses 6A, 6B, and 7. Microbiol. Spectr. 2016, 4, 157–176. [Google Scholar] [CrossRef] [PubMed]
  92. Sedlak, R.H.; Cook, L.; Huang, M.-L.; Magaret, A.; Zerr, D.M.; Boeckh, M.; Jerome, K.R. Identification of chromosomally integrated human herpesvirus 6 by droplet digital PCR. Clin. Chem. 2014, 60, 765–772. [Google Scholar] [CrossRef]
  93. Sharp, C.; Golubchik, T.; Gregory, W.F.; McNaughton, A.L.; Gow, N.; Selvaratnam, M.; Mirea, A.; Foster, D.; Andersson, M.; Klenerman, P.; et al. Human Herpes Virus 6 (HHV-6)-Pathogen or Passenger? A Pilot Study of Clinical Laboratory Data and Next Generation Sequencing; Cold Spring Harbor Laboratory: Laurel Hollow, NY, USA, 2017; p. 236083. [Google Scholar]
  94. Deback, C.; Agbalika, F.; Scieux, C.; Marcelin, A.; Gautheret-Dejean, A.; Cherot, J.; Hermet, L.; Roger, O.; Agut, H. Detection of human herpesviruses HHV-6, HHV-7 and HHV-8 in whole blood by real-time PCR using the new CMV, HHV-6, 7, 8 R-gene™ kit. J. Virol. Methods 2008, 149, 285–291. [Google Scholar] [CrossRef] [PubMed]
  95. Zheng, Y.; Zhao, Y.; Wang, Y.; Rao, J. A multiplex real-time PCR quantitation of human herpesvirus-6, 7, 8 viruses: Application in blood transfusions. Virol. J. 2021, 18, 38. [Google Scholar] [CrossRef] [PubMed]
  96. Shirahama, S.; Tanaka, R.; Kaburaki, T. Anterior Uveitis Associated with Human Herpesvirus 7 Infection Diagnosed by Multiplex Polymerase Chain Reaction Assay: A Case Report. Ocul. Immunol. Inflamm. 2023, 31, 474–476. [Google Scholar] [CrossRef]
  97. Hindson, B.J.; Ness, K.D.; Masquelier, D.A.; Belgrader, P.; Heredia, N.J.; Makarewicz, A.J.; Bright, I.J.; Lucero, M.Y.; Hiddessen, A.L.; Legler, T.C.; et al. High-throughput droplet digital PCR system for absolute quantitation of DNA copy number. Anal. Chem. 2011, 83, 8604–8610. [Google Scholar] [CrossRef]
  98. Lallemand, F.; Desire, N.; Rozenbaum, W.; Nicolas, J.-C.; Marechal, V. Quantitative Analysis of Human Herpesvirus 8 Viral Load Using a Real-Time PCR Assay. J. Clin. Microbiol. 2000, 38, 1404–1408. [Google Scholar] [CrossRef]
  99. Pellett, P.E.; Spira, T.J.; Bagasra, O.; Boshoff, C.; Corey, L.; de Lellis, L.; Huang, M.-L.; Lin, J.-C.; Matthews, S.; Monini, P.; et al. Multicenter Comparison of PCR Assays for Detection of Human Herpesvirus 8 DNA in Semen. J. Clin. Microbiol. 1999, 37, 1298–1301. [Google Scholar] [CrossRef]
  100. Derafshi, R.; Ghapanchi, J.; Rezazadeh, F.; Kalantari, M.H.; Naeeni, A.M.; Farzin, M.; Moattari, A. PCR Detection of HHV8 DNA in the Saliva of Removable Denture Wearers Compared to Dentate Cases in Shiraz, South of Iran. BioMed Res. Int. 2020, 2020, 9358947. [Google Scholar] [CrossRef] [PubMed]
  101. Zhang, E.; Cotton, V.E.; Hidalgo-Bravo, A.; Huang, Y.; Bell, A.J.; Jarrett, R.F.; Wilkie, G.S.; Davison, A.J.; Nacheva, E.P.; Siebert, R.; et al. HHV-8-unrelated primary effusion-like lymphoma associated with clonal loss of inherited chromosomally-integrated human herpesvirus-6A from the telomere of chromosome 19q. Sci. Rep. 2016, 6, 22730. [Google Scholar] [CrossRef] [PubMed]
  102. Whitley, R. Neonatal herpes simplex virus infections. J. Med. Virol. 1993, 41 (Suppl. S1), 13–21. [Google Scholar] [CrossRef]
  103. Lafferty, W.E.; Downey, L.; Celum, C.; Wald, A. Herpes simplex virus type 1 as a cause of genital herpes: Impact on surveillance and prevention. J. Infect. Dis. 2000, 181, 1454–1457. [Google Scholar] [CrossRef]
  104. Nahmias, A.J.; Keyserling, H.; Lee, F.K. Herpes simplex viruses 1 and 2. Viral Infect. Hum. Epidemiol. Control 1989, 393–417. [Google Scholar]
  105. Anderson, N.W.; Buchan, B.W.; Ledeboer, N.A. Light microscopy, culture, molecular, and serologic methods for detection of herpes simplex virus. J. Clin. Microbiol. 2014, 52, 2–8. [Google Scholar] [CrossRef] [PubMed]
  106. Dominguez, S.R.; Pretty, K.; Hengartner, R.; Robinson, C.C. Comparison of herpes simplex virus PCR with culture for virus detection in multisource surface swab specimens from neonates. J. Clin. Microbiol. 2018, 56, e00632-18. [Google Scholar] [CrossRef]
  107. Bagga, B.; Kate, A.; Joseph, J.; Dave, V.P. Herpes simplex infection of the eye: An introduction. Community Eye Health 2020, 33, 68. [Google Scholar]
  108. Wald, A. Genital HSV-1 Infections. Sex Transm. Infect. 2006, 82, 189–190. [Google Scholar] [CrossRef]
  109. Singh, A.; Preiksaitis, J.; Romanowski, B. The laboratory diagnosis of herpes simplex virus infections. Can. J. Infect. Dis. Med. Microbiol. 2005, 16, 92–98. [Google Scholar] [CrossRef] [PubMed]
  110. Scoular, A.; Gillespie, G.; Carman, W. Polymerase chain reaction for diagnosis of genital herpes in a genitourinary medicine clinic. Sex. Transm. Infect. 2002, 78, 21–25. [Google Scholar] [CrossRef] [PubMed]
  111. Strick, L.B.; Wald, A. Diagnostics for Herpes Simplex Virus. Mol. Diagn. Ther. 2006, 10, 17–28. [Google Scholar] [CrossRef] [PubMed]
  112. Yuan, L.; Xia, D.; Zhou, Q.; Xu, W.; Xu, S.; Yin, Y. An evaluation of a multiplex PCR assay for the detection of Treponema pallidum, HSV-1, and HSV-2. Diagn. Microbiol. Infect. Dis. 2023, 106, 115958. [Google Scholar] [CrossRef]
  113. Jain, S.; Wyatt, D.; McCaughey, C.; O’Neill, H.J.; Coyle, P.V. Nested multiplex polymerase chain reaction for the diagnosis of cutaneous herpes simplex and herpes zoster infections and a comparison with electronmicroscopy. J. Med. Virol. 2001, 63, 52–56. [Google Scholar] [CrossRef] [PubMed]
  114. Nogueira, M.; Amorim, J.B.; Oliveira, J.G.; Bonjardim, C.A.; Ferreira, P.C.; Kroon, E.G. Comparison of virus isolation and various polymerase chain reaction methods in the diagnosis of mucocutaneous herpesvirus infection. Acta Virol. 2000, 44, 61–66. [Google Scholar]
  115. Aryee, E.A.; Bailey, R.L.; Natividad-Sancho, A.; Kaye, S.; Holland, M.J. Detection, quantification and genotyping of Herpes Simplex Virus in cervicovaginal secretions by real-time PCR: A cross sectional survey. Virol. J. 2005, 2, 61. [Google Scholar] [CrossRef] [PubMed]
  116. AK, A.K.; Bhutta, B.S.; Mendez, M.D. Herpes simplex encephalitis. In StatPearls; StatPearls Publishing: Treasure Island, FL, USA, 2024. [Google Scholar]
  117. Tong, Y.; McCarthy, K.; Kong, H.; Lemieux, B. Development and comparison of a rapid isothermal nucleic acid amplification test for typing of herpes simplex virus types 1 and 2 on a portable fluorescence detector. J. Mol. Diagn. 2012, 14, 569–576. [Google Scholar] [CrossRef] [PubMed]
  118. Legoff, J.; Bouhlal, H.; Grésenguet, G.; Weiss, H.; Khonde, N.; Hocini, H.; Désiré, N.; Si-Mohamed, A.; de Dieu Longo, J.; Chemin, C.; et al. Real-time PCR quantification of genital shedding of herpes simplex virus (HSV) and human immunodeficiency virus (HIV) in women coinfected with HSV and HIV. J. Clin. Microbiol. 2006, 44, 423–432. [Google Scholar] [CrossRef]
  119. Domingues, R.; Lakeman, F.D.; Mayo, M.S.; Whitley, R.J. Application of competitive PCR to cerebrospinal fluid samples from patients with herpes simplex encephalitis. J. Clin. Microbiol. 1998, 36, 2229–2234. [Google Scholar] [CrossRef]
  120. Tanaka, T.; Kogawa, K.; Sasa, H.; Nonoyama, S.; Furuya, K.; Sato, K. Rapid and simultaneous detection of 6 types of human herpes virus (herpes simplex virus, varicella-zoster virus, Epstein-Barr virus, cytomegalovirus, human herpes virus 6A/B, and human herpes virus 7) by multiplex PCR assay. Biomed. Res. 2009, 30, 279–285. [Google Scholar] [CrossRef] [PubMed]
  121. Yap, T.; Khor, S.; Kim, J.S.; Kim, J.; Yun, J.S.W.; Kern, J.S.; Martyres, R.; Varigos, G.; Chan, H.T.; McCullough, M.J.; et al. Intraoral human herpes viruses detectable by PCR in majority of patients. Oral Dis. 2021, 27, 378–387. [Google Scholar] [CrossRef] [PubMed]
  122. Ando, Y.; Kimura, H.; Miwata, H.; Kudo, T.; Shibata, M.; Morishima, T. Quantitative analysis of herpes simplex virus DNA in cerebrospinal fluid of children with herpes simplex encephalitis. J. Med. Virol. 1993, 41, 170–173. [Google Scholar] [CrossRef] [PubMed]
  123. Kimura, H.; Futamura, M.; Kito, H.; Ando, T.; Goto, M.; Kuzushima, K.; Shibata, M.; Morishima, T. Detection of viral DNA in neonatal herpes simplex virus infections: Frequent and prolonged presence in serum and cerebrospinal fluid. J. Infect. Dis. 1991, 164, 289–293. [Google Scholar] [CrossRef] [PubMed]
  124. Troendle-Atkins, J.; Demmler, G.J.; Buffone, G.J. Buffone, Rapid diagnosis of herpes simplex virus encephalitis by using the polymerase chain reaction. J. Pediatr. 1993, 123, 376–380. [Google Scholar] [CrossRef]
  125. Benjamin, L.A.; Kelly, M.; Cohen, D.; Neuhann, F.; Galbraith, S.; Mallewa, M.; Hopkins, M.; Hart, I.J.; Guiver, M.; Lalloo, D.G.; et al. Detection of herpes viruses in the cerebrospinal fluid of adults with suspected viral meningitis in Malawi. Infection 2012, 41, 27–31. [Google Scholar] [CrossRef]
  126. Studahl, M.; Hagberg, L.; Rekabdar, E.; Bergström, T. Herpesvirus DNA detection in cerebral spinal fluid: Differences in clinical presentation between alpha-, beta-, and gamma-herpesviruses. Scand. J. Infect. Dis. 2000, 32, 237–248. [Google Scholar] [PubMed]
  127. Doan, T.; Acharya, N.R.; Pinsky, B.A.; Sahoo, M.K.; Chow, E.D.; Banaei, N.; Budvytiene, I.; Cevallos, V.; Zhong, L.; Zhou, Z.; et al. Metagenomic DNA sequencing for the diagnosis of intraocular infections. Ophthalmology 2017, 124, 1247–1248. [Google Scholar] [CrossRef] [PubMed]
  128. Musa, M.; Enaholo, E.; Aluyi-Osa, G.; Atuanya, G.N.; Spadea, L.; Salati, C.; Zeppieri, M. Herpes simplex keratitis: A brief clinical overview. World J. Virol. 2024, 13, 89934. [Google Scholar] [CrossRef]
  129. Joseph, J.; Guda, S.J.M.; Sontam, B.; Bagga, B.; Ranjith, K.; Sharma, S. Evaluation of multiplex real-time polymerase chain reaction for the detection of herpes simplex virus-1 and 2 and varicella-zoster virus in corneal cells from normal subjects and patients with keratitis in India. Indian J. Ophthalmol. 2019, 67, 1040–1046. [Google Scholar] [CrossRef]
  130. Kobayashi, T.; Suzuki, T.; Okajima, Y.; Aoki, K.; Ishii, Y.; Tateda, K.; Hori, Y. Metagenome techniques for detection of pathogens causing ocular infection. Reports 2021, 4, 6. [Google Scholar] [CrossRef]
  131. Doan, T.; Wilson, M.R.; Crawford, E.D.; Chow, E.D.; Khan, L.M.; Knopp, K.A.; O’donovan, B.D.; Xia, D.; Hacker, J.K.; Stewart, J.M.; et al. Illuminating uveitis: Metagenomic deep sequencing identifies common and rare pathogens. Genome Med. 2016, 8, 90. [Google Scholar] [CrossRef]
  132. Gilden, D.; Mahalingam, R.; Nagel, M.A.; Pugazhenthi, S.; Cohrs, R.J. The neurobiology of varicella zoster virus infection. Neuropathol. Appl. Neurobiol. 2011, 37, 441–463. [Google Scholar] [CrossRef]
  133. O’Riavdan, M.; O’Gorman, C.; Morgan, C. Seroprevelence of varicella zoster virus in pregnant women in Dublin. J. Med. Sci. 2000, 169, 288. [Google Scholar]
  134. Leikin, E.; Figueroa, R.; Bertkau, A.; Lysikiewicz, A.; Visintainer, P.; Tejani, N. Seronegativity to varicella-zoster virus in a tertiary care obstetric population. Obstet. Gynecol. 1997, 90, 511–513. [Google Scholar] [CrossRef] [PubMed]
  135. Miller, E.; Cradock-Watson, T.; Ridehalgh, M.S.J.T.L. Outcome in newborn babies given anti-varicella-zoster immunoglobulin after perinatal maternal infection with varicella-zoster virus. Lancet 1989, 334, 371–373. [Google Scholar] [CrossRef] [PubMed]
  136. Gershon, A.A.; Raker, R.; Steinberg, S.; Topf-Olstein, B.; Drusin, L.M. Antibody to varicella-zoster virus in parturient women and their offspring during the first year of life. Pediatrics 1976, 58, 692–696. [Google Scholar] [CrossRef]
  137. Chapman, S.J. Varicella in pregnancy. In Seminars in Perinatology; Elsevier: Amsterdam, The Netherlands, 1998. [Google Scholar]
  138. Mendelson, E.; Aboudy, Y.; Smetana, Z.; Tepperberg, M.; Grossman, Z. Laboratory assessment and diagnosis of congenital viral infections: Rubella, cytomegalovirus (CMV), varicella-zoster virus (VZV), herpes simplex virus (HSV), parvovirus B19 and human immunodeficiency virus (HIV). Reprod. Toxicol. 2006, 21, 350–382. [Google Scholar] [CrossRef]
  139. Herlin, L.K.; Hansen, K.S.; Bodilsen, J.; Larsen, L.; Brandt, C.; Andersen, C.; Hansen, B.R.; Lüttichau, H.R.; Helweg-Larsen, J.; Wiese, L.; et al. Varicella Zoster Virus Encephalitis in Denmark from 2015 to 2019—A Nationwide Prospective Cohort Study. Clin. Infect. Dis. 2020, 72, 1192–1199. [Google Scholar] [CrossRef]
  140. Mirouse, A.; Sonneville, R.; Razazi, K.; Merceron, S.; Argaud, L.; Bigé, N.; Faguer, S.; Perez, P.; Géri, G.; Guérin, C.; et al. Neurologic outcome of VZV encephalitis one year after ICU admission: A multicenter cohort study. Ann. Intensiv. Care 2022, 12, 32. [Google Scholar] [CrossRef] [PubMed]
  141. Lizzi, J.; Hill, T.; Jakubowski, J. Varicella zoster virus encephalitis. Clin. Pract. Cases Emerg. Med. 2019, 3, 380–382. [Google Scholar] [CrossRef] [PubMed]
  142. Finnström, N.; Bergsten, K.; Ström, H.; Sundell, T.; Martin, S.; Wutzler, P.; Sauerbrei, A. Analysis of varicella-zoster virus and herpes simplex virus in various clinical samples by the use of different PCR assays. J. Virol. Methods 2009, 160, 193–196. [Google Scholar] [CrossRef] [PubMed]
  143. Sauerbrei, A.; Eichhorn, U.; Schacke, M.; Wutzler, P. Laboratory diagnosis of herpes zoster. J. Clin. Virol. 1999, 14, 31–36. [Google Scholar] [CrossRef] [PubMed]
  144. Mouly, F.; Mirlesse, V.; Méritet, J.F.; Rozenberg, F.; Poissonier, M.H.; Lebon, P.; Daffos, F. Prenatal diagnosis of fetal varicella-zoster virus infection with polymerase chain reaction of amniotic fluid in 107 cases. Am. J. Obstet. Gynecol. 1997, 177, 894–898. [Google Scholar] [CrossRef] [PubMed]
  145. Puchhammer-Stöckl, E.; Kunz, C.; Wagner, G.; Enders, G. Detection of varicella zoster virus (VZV) DNA in fetal tissue by polymerase chain reaction. J. Perinat. Med. 1994, 22, 65–69. [Google Scholar] [CrossRef]
  146. Tang, Y.W.; Espy, M.J.; Persing, D.H.; Smith, T.F. Molecular evidence and clinical significance of herpesvirus coinfection in the central nervous system. J. Clin. Microbiol. 1997, 35, 2869–2872. [Google Scholar] [CrossRef]
  147. Mehta, S.K.; Tyring, S.K.; Cohrs, R.J.; Gilden, D.; Feiveson, A.H.; Lechler, K.J.; Pierson, D.L. Rapid and sensitive detection of varicella zoster virus in saliva of patients with herpes zoster. J. Virol. Methods 2013, 193, 128–130. [Google Scholar] [CrossRef] [PubMed]
  148. Rowley, A.; Whitley, R.J.; Lakeman, F.D.; Wolinsky, S.M. Rapid detection of herpes-simplex-virus DNA in cerebrospinal fluid of patients with herpes simplex encephalitis. Lancet 1990, 335, 440–441. [Google Scholar] [CrossRef] [PubMed]
  149. Tang, Y.-W.; Mitchell, P.S.; Espy, M.J.; Smith, T.F.; Persing, D.H. Molecular diagnosis of herpes simplex virus infections in the central nervous system. J. Clin. Microbiol. 1999, 37, 2127–2136. [Google Scholar] [CrossRef]
  150. Weidmann, M.; Meyer-König, U.; Hufert, F.T. Rapid detection of herpes simplex virus and varicella-zoster virus infections by real-time PCR. J. Clin. Microbiol. 2003, 41, 1565–1568. [Google Scholar] [CrossRef] [PubMed]
  151. Mendonça, C.; Zauli, D.J.C.C. A-308 The Molecular Diagnosis of Varicella Zoster Virus (VZV): A Validation of a Real-Time PCR (qPCR) Assay for the Qualitative Detection. Clin. Chem. 2023, 69 (Suppl. S1), hvad097.273. [Google Scholar] [CrossRef]
  152. Leung, J. Notes from the field: Congenital varicella syndrome case—Illinois, 2021. MMWR Morb. Mortal. Wkly. Rep. 2022, 71, 390–392. [Google Scholar] [CrossRef]
  153. Abusalah, M.A.H.; Gan, S.H.; Al-Hatamleh, M.A.; Irekeola, A.A.; Shueb, R.H.; Yean Yean, C. Recent advances in diagnostic approaches for epstein–barr virus. Pathogens 2020, 9, 226. [Google Scholar] [CrossRef]
  154. Gulley, M.L. Molecular diagnosis of Epstein-Barr virus-related diseases. J. Mol. Diagn. 2001, 3, 1–10. [Google Scholar] [CrossRef]
  155. Gulley, M.L.; Tang, W. Laboratory assays for Epstein-Barr virus-related disease. J. Mol. Diagn. 2008, 10, 279–292. [Google Scholar] [CrossRef]
  156. Kimura, H.; Ito, Y.; Suzuki, R.; Nishiyama, Y. Measuring Epstein–Barr virus (EBV) load: The significance and application for each EBV-associated disease. Rev. Med. Virol. 2008, 18, 305–319. [Google Scholar] [CrossRef]
  157. Smatti, M.K.; Al-Sadeq, D.W.; Ali, N.H.; Pintus, G.; Abou-Saleh, H.; Nasrallah, G.K. Epstein–Barr virus epidemiology, serology, and genetic variability of LMP-1 oncogene among healthy population: An update. Front. Oncol. 2018, 8, 211. [Google Scholar] [CrossRef]
  158. De Paschale, M.; Clerici, P. Serological diagnosis of Epstein-Barr virus infection: Problems and solutions. World J. Virol. 2012, 1, 31–43. [Google Scholar] [CrossRef]
  159. Grimm-Geris, J.; Dunmire, S.K.; Duval, L.M.; Filtz, E.A.; Leuschen, H.J.; Schmeling, D.O.; Kulasingam, S.L.; Balfour, H.H. Screening for Epstein–Barr virus (EBV) infection status in university freshmen: Acceptability of a gingival swab method. Epidemiol. Infect. 2019, 147, e140. [Google Scholar] [CrossRef]
  160. Owens, G.P.; Bennett, J.L. Trigger, pathogen, or bystander: The complex nexus linking Epstein–Barr virus and multiple sclerosis. Mult. Scler. 2012, 18, 1204–1208. [Google Scholar] [CrossRef]
  161. Cocuzza, C.E.; Piazza, F.; Musumeci, R.; Oggioni, D.; Andreoni, S.; Gardinetti, M.; Fusco, L.; Frigo, M.; Banfi, P.; Rottoli, M.R.; et al. Quantitative Detection of Epstein-Barr Virus DNA in Cerebrospinal Fluid and Blood Samples of Patients with Relapsing-Remitting Multiple Sclerosis. PLoS ONE 2014, 9, e94497. [Google Scholar] [CrossRef]
  162. Zuhair, M.; Smit, G.S.A.; Wallis, G.; Jabbar, F.; Smith, C.; Devleesschauwer, B.; Griffiths, P. Estimation of the worldwide seroprevalence of cytomegalovirus: A systematic review and meta-analysis. Rev. Med. Virol. 2019, 29, e2034. [Google Scholar] [CrossRef]
  163. Griffiths, P.; Baraniak, I.; Reeves, M. The pathogenesis of human cytomegalovirus. J. Pathol. 2015, 235, 288–297. [Google Scholar] [CrossRef]
  164. Dioverti, M.V.; Razonable, R.R. Cytomegalovirus. Microbiol. Spectr. 2016, 4, 97–125. [Google Scholar] [CrossRef]
  165. Ho, M.; Suwansirikul, S.; Dowling, J.N.; Youngblood, L.A.; Armstrong, J.A. The transplanted kidney as a source of cytomegalovirus infection. N. Engl. J. Med. 1975, 293, 1109–1112. [Google Scholar] [CrossRef]
  166. Krstanović, F.; Britt, W.J.; Jonjić, S.; Brizić, I. Cytomegalovirus Infection and Inflammation in Developing Brain. Viruses 2021, 13, 1078. [Google Scholar] [CrossRef]
  167. Umbach, J.L.; Kramer, M.F.; Jurak, I.; Karnowski, H.W.; Coen, D.M.; Cullen, B.R. MicroRNAs expressed by herpes simplex virus 1 during latent infection regulate viral mRNAs. Nature 2008, 454, 780–783. [Google Scholar] [CrossRef]
  168. Nigro, G. Maternal–fetal cytomegalovirus infection: From diagnosis to therapy. J. Matern. Fetal. Neonatal. Med. 2009, 22, 169–174. [Google Scholar] [CrossRef]
  169. Grossi, P.A.; Peghin, M. Recent advances in cytomegalovirus infection management in solid organ transplant recipients. Curr. Opin. Organ Transplant. 2024, 29, 131–137. [Google Scholar] [CrossRef]
  170. Boeckh, M.; Boivin, G. Quantitation of Cytomegalovirus: Methodologic Aspects and Clinical Applications. Clin. Microbiol. Rev. 1998, 11, 533–554. [Google Scholar] [CrossRef]
  171. Hildenbrand, C.; Wedekind, L.; Li, G.; vonRentzell, J.E.; Shah, K.; Rooney, P.; Harrington, A.T.; Zhao, R.Y. Clinical evaluation of Roche COBAS® AmpliPrep/COBAS® TaqMan® CMV test using nonplasma samples. J. Med. Virol. 2018, 90, 1611–1619. [Google Scholar] [CrossRef]
  172. Boeckh, M.; Huang, M.; Ferrenberg, J.; Stevens-Ayers, T.; Stensland, L.; Nichols, W.G.; Corey, L. Optimization of Quantitative Detection of Cytomegalovirus DNA in Plasma by Real-Time PCR. J. Clin. Microbiol. 2004, 42, 1142–1148. [Google Scholar] [CrossRef]
  173. Satou, J.; Funato, T.; Satoh, N.; Abe, Y.; Ishii, K.; Sasaki, T.; Kaku, M. Quantitative PCR determination of human cytomegalovirus in blood cells. J. Clin. Lab. Anal. 2001, 15, 122–126. [Google Scholar] [CrossRef]
  174. Gault, E.; Michel, Y.; Dehée, A.; Belabani, C.; Nicolas, J.-C.; Garbarg-Chenon, A. Quantification of Human Cytomegalovirus DNA by Real-Time PCR. J. Clin. Microbiol. 2001, 39, 772–775. [Google Scholar] [CrossRef]
  175. Shlonsky, Y.; Smair, N.S.; Mubariki, R.; Bamberger, E.; Hemo, M.; Cohen, S.; Riskin, A.; Srugo, I.; Bader, D.; Golan-Shany, O. Pooled saliva CMV DNA detection: A viable laboratory technique for universal CMV screening of healthy newborns. J. Clin. Virol. 2021, 138, 104798. [Google Scholar] [CrossRef]
  176. Yamamoto, A.Y.; Mussi-Pinhata, M.M.; Marin, L.J.; Brito, R.M.; Oliveira, P.F.C.; Coelho, T.B. Is saliva as reliable as urine for detection of cytomegalovirus DNA for neonatal screening of congenital CMV infection? J. Clin. Virol. 2006, 36, 228–230. [Google Scholar] [CrossRef]
  177. Demmler, G.J. Screening for congenital cytomegalovirus infection: A tapestry of controversies. J. Pediatr. 2005, 146, 162–164. [Google Scholar] [CrossRef]
  178. Halwachs-Baumann, G.; Genser, B.; Pailer, S.; Engele, H.; Rosegger, H.; Schalk, A.; Kessler, H.H.; Truschnig-Wilders, M. Human cytomegalovirus load in various body fluids of congenitally infected newborns. J. Clin. Virol. 2002, 25, 81–87. [Google Scholar] [CrossRef]
  179. Boeckh, M.; Gallez-Hawkins, G.M.; Myerson, D.; Zaia, J.A.; Bowden, R.A. Plasma polymerase chain reaction for cytomegalovirus DNA after allogeneic marrow transplantation: Comparison with Polymerase Chain Reaction Using Peripheral Blood Leukocytes, pp65 Antigenemia, and Viral Culture. Transplantation 1997, 64, 108–113. [Google Scholar] [CrossRef]
  180. Boivin, G.; Handfield, J.; Toma, E.; Murray, G.; Lalonde, R.; Bergeron, M.G. Comparative Evaluation of the Cytomegalovirus DNA Load in Polymorphonuclear Leukocytes and Plasma of Human Immunodeficiency Virus-Infected Subjects. J. Infect. Dis. 1998, 177, 355–360. [Google Scholar] [CrossRef]
  181. Boeckh, M.; Leisenring, W.; Riddell, S.R.; Bowden, R.A.; Huang, M.-L.; Myerson, D.; Stevens-Ayers, T.; Flowers, M.E.D.; Cunningham, T.; Corey, L. Late cytomegalovirus disease and mortality in recipients of allogeneic hematopoietic stem cell transplants: Importance of viral load and T-cell immunity. Blood 2003, 101, 407–414. [Google Scholar] [CrossRef] [PubMed]
  182. Spector, S.A.; Hsia, K.; Crager, M.; Pilcher, M.; Cabral, S.; Stempien, M.J. Cytomegalovirus (CMV) DNA Load Is an Independent Predictor of CMV Disease and Survival in Advanced AIDS. J. Virol. 1999, 73, 7027–7030. [Google Scholar] [CrossRef]
  183. Deback, C.; Fillet, A.; Dhedin, N.; Barrou, B.; Varnous, S.; Najioullah, F.; Bricaire, F.; Agut, H. Monitoring of human cytomegalovirus infection in immunosuppressed patients using real-time PCR on whole blood. J. Clin. Virol. 2007, 40, 173–179. [Google Scholar] [CrossRef]
  184. Garrigue, I.; Doussau, A.; Asselineau, J.; Bricout, H.; Couzi, L.; Rio, C.; Merville, P.; Fleury, H.; Lafon, M.-E.; Thiébaut, R. Prediction of Cytomegalovirus (CMV) Plasma Load from Evaluation of CMV Whole-Blood Load in Samples from Renal Transplant Recipients. J. Clin. Microbiol. 2008, 46, 493–498. [Google Scholar] [CrossRef]
  185. Razonable, R.R.; Brown, R.A.; Wilson, J.; Groettum, C.; Kremers, W.; Espy, M.; Smith, T.F.; Paya, C.V. The clinical use of various blood compartments for cytomegalovirus (CMV) DNA quantitation in transplant recipients with CMV disease. Transplantation 2002, 73, 968–973. [Google Scholar] [CrossRef]
  186. Wu, W.; Jiang, H.; Zhang, Y.; Zhou, Y.; Bai, G.; Shen, L.; Zhou, H.; Chen, X.; Hu, L. Clinical metagenomic next-generation sequencing for diagnosis of secondary glaucoma in patients with cytomegalovirus-induced corneal endotheliitis. Front. Microbiol. 2022, 13, 940818. [Google Scholar] [CrossRef]
  187. Smith, I.L.; Macdonald, J.C.; Freeman, W.R.; Shapiro, A.M.; Spector, S.A. Cytomegalovirus (CMV) retinitis activity is accurately reflected by the presence and level of CMV DNA in aqueous humor and vitreous. J. Infect. Dis. 1999, 179, 1249–1253. [Google Scholar] [CrossRef]
  188. Bolle, L.D.; Naesens, L.; Clercq, E.D. Update on Human Herpesvirus 6 Biology, Clinical Features, and Therapy. Clin. Microbiol. Rev. 2005, 18, 217–245. [Google Scholar] [CrossRef]
  189. Ablashi, D.; Agut, H.; Alvarez-Lafuente, R.; Clark, D.A.; Dewhurst, S.; DiLuca, D.; Flamand, L.; Frenkel, N.; Gallo, R.; Gompels, U.A.; et al. Classification of HHV-6A and HHV-6B as distinct viruses. Arch. Virol. 2013, 159, 863–870. [Google Scholar] [CrossRef] [PubMed]
  190. Burbelo, P.D.; Bayat, A.; Wagner, J.; Nutman, T.B.; Baraniuk, J.N.; Iadarola, M.J. No serological evidence for a role of HHV-6 infection in chronic fatigue syndrome. Am. J. Transl. Res. 2012, 4, 443. [Google Scholar]
  191. Agut, H.; Bonnafous, P.; Gautheret-Dejean, A. Laboratory and Clinical Aspects of Human Herpesvirus 6 Infections. Clin. Microbiol. Rev. 2015, 28, 313–335. [Google Scholar] [CrossRef]
  192. Campadelli-Fiume, G.; Mirandola, P.; Menotti, L. Human herpesvirus 6: An emerging pathogen. Emerg. Infect. Dis. 1999, 5, 353–366. [Google Scholar] [CrossRef]
  193. Mullins, T.B.; Krishnamurthy, K. Roseola infantum. In StatPearls; StatPearls Publishing: Treasure Island, FL, USA, 2023. [Google Scholar]
  194. Norton, R.A.; Caserta, M.T.; Hall, C.B.; Schnabel, K.; Hocknell, P.; Dewhurst, S. Detection of human herpesvirus 6 by reverse transcription-PCR. J. Clin. Microbiol. 1999, 37, 3672–3675. [Google Scholar] [CrossRef]
  195. Bortolotti, D.; Gentili, V.; Rotola, A.; Caselli, E.; Rizzo, R. HHV-6A infection induces amyloid-beta expression and activation of microglial cells. Alzheimer’s Res. Ther. 2019, 11, 104. [Google Scholar] [CrossRef]
  196. Leibovitch, E.C.; Brunetto, G.S.; Caruso, B.; Fenton, K.; Ohayon, J.; Reich, D.S.; Jacobson, S. Coinfection of human herpesviruses 6A (HHV-6A) and HHV-6B as demonstrated by novel digital droplet PCR assay. PLoS ONE 2014, 9, e92328. [Google Scholar] [CrossRef]
  197. Marci, R.; Gentili, V.; Bortolotti, D.; Lo Monte, G.; Caselli, E.; Bolzani, S.; Rotola, A.; Di Luca, D.; Rizzo, R. Presence of HHV-6A in endometrial epithelial cells from women with primary unexplained infertility. PLoS ONE 2016, 11, e0158304. [Google Scholar] [CrossRef] [PubMed]
  198. Caselli, E.; Bortolotti, D.; Marci, R.; Rotola, A.; Gentili, V.; Soffritti, I.; D’Accolti, M.; Monte, G.L.; Sicolo, M.; Barao, I.; et al. HHV-6A infection of endometrial epithelial cells induces increased endometrial NK cell-mediated cytotoxicity. Front. Microbiol. 2017, 8, 2525. [Google Scholar] [CrossRef]
  199. Morissette, G.; Flamand, L. Herpesviruses and chromosomal integration. J. Virol. 2010, 84, 12100–12109. [Google Scholar] [CrossRef]
  200. Pellett, P.E.; Ablashi, D.V.; Ambros, P.F.; Agut, H.; Caserta, M.T.; Descamps, V.; Flamand, L.; Gautheret-Dejean, A.; Hall, C.B.; Kamble, R.T.; et al. Chromosomally integrated human herpesvirus 6: Questions and answers. Rev. Med. Virol. 2011, 22, 144–155. [Google Scholar] [CrossRef]
  201. Wang, H.; Tomatis-Souverbielle, C.; Everhart, K.; Oyeniran, S.J.; Leber, A.L. Detection of human herpesvirus 6 in pediatric CSF samples: Causing disease or incidental distraction? Diagn. Microbiol. Infect. Dis. 2023, 107, 116029. [Google Scholar] [CrossRef]
  202. Kamble, R.T.; A Clark, D.; Leong, H.N.; E Heslop, H.; Brenner, M.K.; Carrum, G. Transmission of integrated human herpesvirus-6 in allogeneic hematopoietic stem cell transplantation. Bone Marrow Transplant. 2007, 40, 563–566. [Google Scholar] [CrossRef]
  203. Zerr, D.M.; Corey, L.; Kim, H.W.; Huang, M.; Nguy, L.; Boeckh, M. Clinical outcomes of human herpesvirus 6 reactivation after hematopoietic stem cell transplantation. Clin. Infect. Dis. 2005, 40, 932–940. [Google Scholar] [CrossRef]
  204. Strenger, V.; Caselli, E.; Lautenschlager, I.; Schwinger, W.; Aberle, S.W.; Loginov, R.; Gentili, V.; Nacheva, E.; DiLuca, D.; Urban, C. Detection of HHV-6-specific mRNA and antigens in PBMCs of individuals with chromosomally integrated HHV-6 (ciHHV-6). Clin. Microbiol. Infect. 2014, 20, 1027–1032. [Google Scholar] [CrossRef]
  205. Yoshikawa, T.; Akimoto, S.; Nishimura, N.; Ozaki, T.; Ihira, M.; Ohashi, M.; Morooka, M.; Suga, S.; Asano, Y.; Takemoto, M.; et al. Evaluation of active human herpesvirus 6 infection by reverse transcription-PCR. J. Med. Virol. 2003, 70, 267–272. [Google Scholar] [CrossRef]
  206. Locatelli, G.; Santoro, F.; Veglia, F.; Gobbi, A.; Lusso, P.; Malnati, M.S. Real-Time Quantitative PCR for Human Herpesvirus 6 DNA. J. Clin. Microbiol. 2000, 38, 4042–4048. [Google Scholar] [CrossRef]
  207. Kainth, M.K.; Caserta, M.T. Molecular diagnostic tests for human herpesvirus 6. Pediatr. Infect. Dis. J. 2011, 30, 604–605. [Google Scholar] [CrossRef]
  208. Chiu, S.S.; Cheung, C.Y.; Tse, C.Y.C.; Peiris, M. Early diagnosis of primary human herpesvirus 6 infection in childhood: Serology, polymerase chain reaction, and virus load. J. Infect. Dis. 1998, 178, 1250–1256. [Google Scholar] [CrossRef]
  209. Clark, D.A.; Ait-Khaled, M.; Wheeler, A.C.; Kidd, I.M.; McLaughlin, J.E.; Johnson, M.A.; Griffiths, P.D.; Emery, V.C. Quantification of human herpesvirus 6 in immunocompetent persons and post-mortem tissues from AIDS patients by PCR. J. Gen. Virol. 1996, 77, 2271–2275. [Google Scholar] [CrossRef]
  210. Ward, K.N.; Leong, H.N.; Thiruchelvam, A.D.; Atkinson, C.E.; Clark, D.A. Human herpesvirus 6 DNA levels in cerebrospinal fluid due to primary infection differ from those due to chromosomal viral integration and have implications for diagnosis of encephalitis. J. Clin. Microbiol. 2007, 45, 1298–1304. [Google Scholar] [CrossRef] [PubMed]
  211. Clark, D.A.; Kidd, I.M.; E Collingham, K.; Tarlow, M.; Ayeni, T.; Riordan, A.; Griffiths, P.D.; Emery, V.C.; Pillay, D. Diagnosis of primary human herpesvirus 6 and 7 infections in febrile infants by polymerase chain reaction. Arch. Dis. Child. 1997, 77, 42–45. [Google Scholar] [CrossRef] [PubMed]
  212. Zerr, D.M.; Huang, M.-L.; Corey, L.; Erickson, M.; Parker, H.L.; Frenkel, L.M. Sensitive Method for Detection of Human Herpesviruses 6 and 7 in Saliva Collected in Field Studies. J. Clin. Microbiol. 2000, 38, 1981–1983. [Google Scholar] [CrossRef] [PubMed]
  213. Flamand, L.; Gravel, A.; Boutolleau, D.; Alvarez-Lafuente, R.; Jacobson, S.; Malnati, M.S.; Kohn, D.; Tang, Y.-W.; Yoshikawa, T.; Ablashi, D. Multicenter comparison of PCR assays for detection of human herpesvirus 6 DNA in serum. J. Clin. Microbiol. 2008, 46, 2700–2706. [Google Scholar] [CrossRef]
  214. Sugita, S.; Shimizu, N.; Watanabe, K.; Ogawa, M.; Maruyama, K.; Usui, N.; Mochizuki, M. Virological Analysis in Patients with Human Herpes Virus 6–Associated Ocular Inflammatory Disorders. Investig. Opthalmology Vis. Sci. 2012, 53, 4692–4698. [Google Scholar] [CrossRef] [PubMed]
  215. Theodore, W.H.; Epstein, L.; Gaillard, W.D.; Shinnar, S.; Wainwright, M.S.; Jacobson, S. Human herpes virus 6B: A possible role in epilepsy? Epilepsia 2008, 49, 1828–1837. [Google Scholar] [CrossRef]
  216. Perlejewski, K.; Popiel, M.; Laskus, T.; Nakamura, S.; Motooka, D.; Stokowy, T.; Lipowski, D.; Pollak, A.; Lechowicz, U.; Cortés, K.C.; et al. Next-generation sequencing (NGS) in the identification of encephalitis-causing viruses: Unexpected detection of human herpesvirus 1 while searching for RNA pathogens. J. Virol. Methods 2015, 226, 1–6. [Google Scholar] [CrossRef]
  217. Gomez, C.A.; Pinsky, B.A.; Liu, A.; Banaei, N. Delayed Diagnosis of Tuberculous Meningitis Misdiagnosed as Herpes Simplex Virus-1 Encephalitis with the FilmArray Syndromic Polymerase Chain Reaction Panel. Open Forum Infect. Dis. 2016, 4, ofw245. [Google Scholar] [CrossRef] [PubMed]
  218. Kawada, J.-I.; Okuno, Y.; Torii, Y.; Okada, R.; Hayano, S.; Ando, S.; Kamiya, Y.; Kojima, S.; Ito, Y. Identification of Viruses in Cases of Pediatric Acute Encephalitis and Encephalopathy Using Next-Generation Sequencing. Sci. Rep. 2016, 6, 33452. [Google Scholar] [CrossRef] [PubMed]
  219. Chan, B.K.; Wilson, T.; Fischer, K.F.; Kriesel, J.D. Deep sequencing to identify the causes of viral encephalitis. PLoS ONE 2014, 9, e93993. [Google Scholar] [CrossRef] [PubMed]
  220. Lusso, P.; Gallo, R.C. 9 Human herpesvirus 6. Baillieres Clin. Haematol. 1995, 8, 201–223. [Google Scholar] [CrossRef]
  221. Foiadelli, T.; Rossi, V.; Paolucci, S.; Rovida, F.; Novazzi, F.; Orsini, A.; Brambilla, I.; Marseglia, G.L.; Baldanti, F.; Savasta, S. Human herpes virus 7-related encephalopathy in children. Acta Biomed. 2022, 92 (Suppl. S4), e2021415. [Google Scholar]
  222. Tanaka, K.; Kondo, T.; Torigoe, S.; Okada, S.; Mukai, T.; Yamanishi, K. Human herpesvirus 7: Another causal agent for roseola (exanthem subitum). J. Pediatr. 1994, 125, 1–5. [Google Scholar] [CrossRef]
  223. Epstein, L.G.; Shinnar, S.; Hesdorffer, D.C.; Nordli, D.R.; Hamidullah, A.; Benn, E.K.T.; Pellock, J.M.; Frank, L.M.; Lewis, D.V.; Moshe, S.L.; et al. Human herpesvirus 6 and 7 in febrile status epilepticus: The FEBSTAT study. Epilepsia 2012, 53, 1481–1488. [Google Scholar] [CrossRef] [PubMed]
  224. Chapenko, S.; Roga, S.; Skuja, S.; Rasa, S.; Cistjakovs, M.; Svirskis, S.; Zaserska, Z.; Groma, V.; Murovska, M. Detection frequency of human herpesviruses-6A-6B, and-7 genomic sequences in central nervous system DNA samples from post-mortem individuals with unspecified encephalopathy. J. Neurovirol. 2016, 22, 488–497. [Google Scholar] [CrossRef]
  225. Ongrádi, J.; Ablashi, D.V.; Yoshikawa, T.; Stercz, B.; Ogata, M. Roseolovirus-associated encephalitis in immunocompetent and immunocompromised individuals. J. Neurovirol. 2017, 23, 1–19. [Google Scholar] [CrossRef] [PubMed]
  226. Luzzi, S.; Lucifero, A.G.; Brambilla, I.; Mantelli, S.S.; Mosconi, M.; Foiadelli, T.; Savasta, S. Targeting the medulloblastoma: A molecular-based approach. Acta Biomed. 2020, 91 (Suppl. S7), 79. [Google Scholar]
  227. Ward, K.N.; Andrews, N.J.; Verity, C.M.; Miller, E.; Ross, E.M. Human herpesviruses-6 and-7 each cause significant neurological morbidity in Britain and Ireland. Arch. Dis. Child. 2005, 90, 619–623. [Google Scholar] [CrossRef] [PubMed]
  228. Chan, P.K.; Chik, K.; To, K.; Li, C.; Shing, M.M.; Ng, K.; Yuen, P.M.; Cheng, A.F. Case report: Human herpesvirus 7 associated fatal encephalitis in a peripheral blood stem cell transplant recipient. J. Med. Virol. 2002, 66, 493–496. [Google Scholar] [CrossRef] [PubMed]
  229. Rasa, S.; Nora-Krukle, Z.; Henning, N.; Eliassen, E.; Shikova, E.; Harrer, T.; Scheibenbogen, C.; Murovska, M.; Prusty, B.K. Chronic viral infections in myalgic encephalomyelitis/chronic fatigue syndrome (ME/CFS). J. Transl. Med. 2018, 16, 268. [Google Scholar] [CrossRef]
  230. Wallace, H.L.; Natelson, B.; Gause, W.; Hay, J. Human Herpesviruses in Chronic Fatigue Syndrome. Clin. Diagn. Lab. Immunol. 1999, 6, 216–223. [Google Scholar] [CrossRef]
  231. Prusty, B.K.; Gulve, N.; Rasa, S.; Murovska, M.; Hernandez, P.C.; Ablashi, D.V. Possible chromosomal and germline integration of human herpesvirus 7. J. Gen. Virol. 2017, 98, 266–274. [Google Scholar] [CrossRef]
  232. Ward, K.N. The natural history and laboratory diagnosis of human herpesviruses-6 and -7 infections in the immunocompetent. J. Clin. Virol. 2005, 32, 183–193. [Google Scholar] [CrossRef] [PubMed]
  233. Van den Berg, J.S.P.; van Zeijl, J.; Rotteveel, J.; Melchers, W.G.; Gabrels, F.M.; Galama, J.D. Neuroinvasion by human herpesvirus type 7 in a case of exanthem subitum with severe neurologic manifestations. Neurology 1999, 52, 1077. [Google Scholar] [CrossRef] [PubMed]
  234. Yoshikawa, T.; Farooqi, I.S.; O’Rahilly, S. Invasion by human herpesvirus 6 and human herpesvirus 7 of the central nervous system in patients with neurological signs and symptoms. Arch. Dis. Child. 2000, 83, 170–171. [Google Scholar] [CrossRef]
  235. Leveque, N.; Van Haecke, A.; Renois, F.; Boutolleau, D.; Talmud, D.; Andreoletti, L. Rapid Virological Diagnosis of Central Nervous System Infections by Use of a Multiplex Reverse Transcription-PCR DNA Microarray. J. Clin. Microbiol. 2011, 49, 3874–3879. [Google Scholar] [CrossRef]
  236. Osman, H.; Peiris, J.S.; Taylor, C.E.; Karlberg, J.P.; Madeley, C.R. Correlation between the detection of viral DNA by the polymerase chain reaction in peripheral blood leukocytes and serological responses to human herpesvirus 6, human herpesvirus 7, and cytomegalovirus in renal allograft recipients. J. Med. Virol. 1997, 53, 288–294. [Google Scholar] [CrossRef]
  237. Wilborn, F.; Schmidt, C.A.; Lorenz, F.; Peng, R.; Gelderblom, H.; Huhn, D.; Siegert, W. Human herpesvirus type 7 in blood donors: Detection by the polymerase chain reaction. J. Med. Virol. 1995, 47, 65–69. [Google Scholar] [CrossRef]
  238. Schulz, T.F. Epidemiology of Kaposi’s sarcoma-associated herpesvirus/human herpesvirus 8. Adv. Cancer Res. 1999, 76, 121–160. [Google Scholar] [PubMed]
  239. Mariggiò, G.; Koch, S.; Schulz, T.F. Kaposi sarcoma herpesvirus pathogenesis. Philos. Trans. R. Soc. B Biol. Sci. 2017, 372, 20160275. [Google Scholar] [CrossRef] [PubMed]
  240. Liu, A.Y.; Nabel, C.S.; Finkelman, B.S.; Ruth, J.R.; Kurzrock, R.; van Rhee, F.; Krymskaya, V.P.; Kelleher, D.; Rubenstein, A.H.; Fajgenbaum, D.C. Idiopathic multicentric Castleman’s disease: A systematic literature review. Lancet Haematol. 2016, 3, e163–e175. [Google Scholar] [CrossRef]
  241. Calabrò, M.L.; Sarid, R. Human herpesvirus 8 and lymphoproliferative disorders. Mediterr. J. Hematol. Infect. Dis. 2018, 10, e2018061. [Google Scholar] [CrossRef] [PubMed]
  242. Lopez, M.; Kainthla, R.; Lazarte, S.; Chen, W.; Nijhawan, A.E.; Knights, S. Outcomes in Kaposi’s sarcoma-associated herpesvirus-associated primary effusion lymphoma and multicentric Castleman’s disease in patients with human immunodeficiency virus (HIV) in a safety-net hospital system. Eur. J. Haematol. 2024, 112, 723–730. [Google Scholar] [CrossRef]
  243. Gonçalves, P.H.; Uldrick, T.S.; Yarchoan, R. HIV-associated Kaposi sarcoma and related diseases. AIDS 2017, 31, 1903–1916. [Google Scholar] [CrossRef]
  244. Cohen, J.I. Herpesvirus latency. J. Clin. Investig. 2020, 130, 3361–3369. [Google Scholar] [CrossRef] [PubMed]
  245. Edelman, D.C. Human herpesvirus 8—A novel human pathogen. Virol. J. 2005, 2, 78. [Google Scholar] [CrossRef]
  246. Andreoni, M.; Sarmati, L.; Nicastri, E.; El Sawaf, G.; El Zalabani, M.; Uccella, I.; Bugarini, R.; Parisi, S.G.; Rezza, G. Primary human herpesvirus 8 infection in immunocompetent children. JAMA 2002, 287, 1295–1300. [Google Scholar] [CrossRef]
  247. Wang, Q.J.; Jenkins, F.J.; Jacobson, L.P.; Kingsley, L.A.; Day, R.D.; Zhang, Z.-W.; Meng, Y.-X.; Pellet, P.E.; Kousoulas, K.G.; Baghian, A.; et al. Primary human herpesvirus 8 infection generates a broadly specific CD8+ T-cell response to viral lytic cycle proteins. Blood 2001, 97, 2366–2373. [Google Scholar] [CrossRef] [PubMed]
  248. Auten, M.; Kim, A.S.; Bradley, K.T.; Rosado, F.G. Human herpesvirus 8-related diseases: Histopathologic diagnosis and disease mechanisms. In Seminars in Diagnostic Pathology; Elsevier: Amsterdam, The Netherlands, 2017. [Google Scholar]
  249. Phipps, W.; Saracino, M.; Selke, S.; Huang, M.-L.; Jaoko, W.; Mandaliya, K.; Wald, A.; Casper, C.; McClelland, R.S. Oral HHV-8 replication among women in Mombasa, Kenya. J. Med. Virol. 2014, 86, 1759–1765. [Google Scholar] [CrossRef]
  250. Taylor, M.M.; Chohan, B.; Lavreys, L.; Hassan, W.; Huang, M.; Corey, L.; Morrow, R.A.; Richardson, B.A.; Mandaliya, K.; Ndinya-Achola, J.; et al. Shedding of Human Herpesvirus 8 in Oral and Genital Secretions from HIV-1-Seropositive and -Seronegative Kenyan Women. J. Infect. Dis. 2004, 190, 484–488. [Google Scholar] [CrossRef] [PubMed]
  251. Zghair, A.N.; Kasim, S.F.; Mohammed, N.S.; Sharma, A.K. Seroprevalence and Biomarkers Detection of Human Herpes Virus (HHV-8) in Patients with Kaposi Sarcoma. J. Tech. 2023, 5, 141–146. [Google Scholar] [CrossRef]
  252. Lawson, J.S.; Glenn, W.K. Evidence for a causal role by human papillomaviruses in prostate cancer—A systematic review. Infect. Agents Cancer 2020, 15, 41. [Google Scholar] [CrossRef] [PubMed]
  253. Frobert, E.; Escuret, V.; Javouhey, E.; Casalegno, J.; Bouscambert-Duchamp, M.; Moulinier, C.; Gillet, Y.; Lina, B.; Floret, D.; Morfin, F. Respiratory viruses in children admitted to hospital intensive care units: Evaluating the CLART® Pneumovir DNA array. J. Med. Virol. 2010, 83, 150–155. [Google Scholar] [CrossRef]
  254. Romero-Gómez, M.P.; González, M.R.; Hierro, S.; Gutiérrez, A. Evaluation of a new immunochromatographic test for detecting adenovirus in respiratory samples from pediatric patients. Enferm. Infecc. Microbiol. Clin. 2008, 26, 598–599. [Google Scholar] [PubMed]
  255. Huang, S.C.M.; Tsao, S.W.; Tsang, C.M. Interplay of viral infection, host cell factors and tumor microenvironment in the pathogenesis of nasopharyngeal carcinoma. Cancers 2018, 10, 106. [Google Scholar] [CrossRef]
  256. Rebolj, M.; Bonde, J.; Preisler, S.; Ejegod, D.; Rygaard, C.; Lynge, E. Human papillomavirus assays and cytology in primary cervical screening of women aged 30 years and above. PLoS ONE 2016, 11, e0147326. [Google Scholar] [CrossRef] [PubMed]
  257. Eklund, C.; Forslund, O.; Wallin, K.-L.; Zhou, T.; Dillner, J. The 2010 global proficiency study of human papillomavirus genotyping in vaccinology. J. Clin. Microbiol. 2012, 50, 2289–2298. [Google Scholar] [CrossRef]
  258. Yang, S.; Rothman, R.E. PCR-based diagnostics for infectious diseases: Uses, limitations, and future applications in acute-care settings. Lancet Infect. Dis. 2004, 4, 337–348. [Google Scholar] [CrossRef] [PubMed]
  259. Saubolle, M.A.; Wojack, B.R.; Wertheimer, A.M.; Fuayagem, A.Z.; Young, S.; Koeneman, B.A. Multicenter clinical validation of a cartridge-based real-time PCR system for detection of Coccidioides spp. in lower respiratory specimens. J. Clin. Microbiol. 2018, 56, e01277-17. [Google Scholar] [CrossRef]
  260. Emmadi, R.; Boonyaratanakornkit, J.B.; Selvarangan, R.; Shyamala, V.; Zimmer, B.L.; Williams, L.; Bryant, B.; Schutzbank, T.; Schoonmaker, M.M.; Wilson, J.A.A.; et al. Molecular methods and platforms for infectious diseases testing: A review of FDA-approved and cleared assays. J. Mol. Diagn. 2011, 13, 583–604. [Google Scholar] [CrossRef]
  261. Mirabile, A.; Sangiorgio, G.; Bonacci, P.G.; Bivona, D.; Nicitra, E.; Bonomo, C.; Bongiorno, D.; Stefani, S.; Musso, N. Advancing Pathogen Identification: The Role of Digital PCR in Enhancing Diagnostic Power in Different Settings. Diagnostics 2024, 14, 1598. [Google Scholar] [CrossRef] [PubMed]
  262. Hindson, C.M.; Chevillet, J.R.; Briggs, H.A.; Gallichotte, E.N.; Ruf, I.K.; Hindson, B.J.; Vessella, R.L.; Tewari, M. Absolute quantification by droplet digital PCR versus analog real-time PCR. Nat. Methods 2013, 10, 1003–1005. [Google Scholar] [CrossRef]
  263. Teh, S.-Y.; Lin, R.; Hung, L.-H.; Lee, A.P. Droplet microfluidics. Lab A Chip 2008, 8, 198–220. [Google Scholar] [CrossRef] [PubMed]
  264. Kwong, J.; Mccallum, N.; Sintchenko, V.; Howden, B. Whole genome sequencing in clinical and public health microbiology. Pathology 2015, 47, 199–210. [Google Scholar] [CrossRef]
  265. Coupland, P.; Chandra, T.; Quail, M.; Reik, W.; Swerdlow, H. Direct sequencing of small genomes on the Pacific Biosciences RS without library preparation. BioTechniques 2012, 53, 365–372. [Google Scholar] [CrossRef]
  266. Rhoads, A.; Au, K.F. Proteomics, and Bioinformatics, PacBio sequencing and its applications. Genom. Proteom. Bioinform. 2015, 13, 278–289. [Google Scholar] [CrossRef] [PubMed]
  267. Shamsipur, M.; Samandari, L.; Taherpour, A.; Pashabadi, A. Sub-femtomolar detection of HIV-1 gene using DNA immobilized on composite platform reinforced by a conductive polymer sandwiched between two nanostructured layers: A solid signal-amplification strategy. Anal. Chim. Acta 2019, 1055, 7–16. [Google Scholar] [CrossRef] [PubMed]
  268. Mukherjee, S.; Strakova, P.; Richtera, L.; Adam, V.; Ashrafi, A. Biosensors-based approaches for other viral infection detection. In Advanced Biosensors for Virus Detection; Elsevier: Amsterdam, The Netherlands, 2022; pp. 391–405. [Google Scholar]
  269. Maddali, H.; Miles, C.E.; Kohn, J.; O’Carroll, D.M. Optical biosensors for virus detection: Prospects for SARS-CoV-2/COVID-19. Chembiochem 2021, 22, 1176–1189. [Google Scholar] [CrossRef]
  270. Castillo-Henríquez, L.; Brenes-Acuña, M.; Castro-Rojas, A.; Cordero-Salmerón, R.; Lopretti-Correa, M.; Vega-Baudrit, J.R. Biosensors for the detection of bacterial and viral clinical pathogens. Sensors 2020, 20, 6926. [Google Scholar] [CrossRef]
  271. Kumar, H.; Kuča, K.; Bhatia, S.K.; Saini, K.; Kaushal, A.; Verma, R.; Bhalla, T.C.; Kumar, D. Applications of Nanotechnology in Sensor-Based Detection of Foodborne Pathogens. Sensors 2020, 20, 1966. [Google Scholar] [CrossRef]
  272. Hua, Y.; Ma, J.; Li, D.; Wang, R. DNA-based biosensors for the biochemical analysis: A review. Biosensors 2022, 12, 183. [Google Scholar] [CrossRef] [PubMed]
  273. Zhang, Z.; Adhikari, B.R.; Sen, P.; Soleymani, L.; Li, Y. Functional nucleic acid-based biosensors for virus detection. Adv. Agrochem. 2023, 2, 246–257. [Google Scholar] [CrossRef]
  274. Collias, D.; Beisel, C.L. CRISPR technologies and the search for the PAM-free nuclease. Nat. Commun. 2021, 12, 555. [Google Scholar] [CrossRef]
  275. Makarova, K.S.; Wolf, Y.I.; Iranzo, J.; Shmakov, S.A.; Alkhnbashi, O.S.; Brouns, S.J.J.; Charpentier, E.; Cheng, D.; Haft, D.H.; Horvath, P.; et al. Evolutionary classification of CRISPR–Cas systems: A burst of class 2 and derived variants. Nat. Rev. Microbiol. 2020, 18, 67–83. [Google Scholar] [CrossRef]
  276. Harrington, L.B.; Burstein, D.; Chen, J.S.; Paez-Espino, D.; Ma, E.; Witte, I.P.; Cofsky, J.C.; Kyrpides, N.C.; Banfield, J.F.; Doudna, J.A. Programmed DNA destruction by miniature CRISPR-Cas14 enzymes. Science 2018, 362, 839–842. [Google Scholar] [CrossRef] [PubMed]
  277. Zhang, F. Development of CRISPR-Cas systems for genome editing and beyond. Q. Rev. Biophys. 2019, 52, e6. [Google Scholar] [CrossRef]
Table 1. Main molecular techniques currently used for viral diagnostics.
Table 1. Main molecular techniques currently used for viral diagnostics.
TechniquePrincipleSamplesAdvantagesDisadvantages
PCRAmplifies specific DNA sequences to detect viral presence.Swabs, CSF, blood, mucosal samplesHigh sensitivity and specificity; rapid resultsSingle-pathogen detection per reaction (conventional PCR)
Real-Time PCR (qPCR)A variant of PCR that quantifies DNA in real time during amplificationSwabs, CSF, blood, mucosal samplesRapid, quantitative; widely applicableLow throughput, single-pathogen detection
Multiplex qPCR Detects multiple pathogens using distinct primersSwabs, CSF, blood, biopsiesSimultaneous detection; cost-effectiveRisk of cross-reactivity, complex setup
Droplet Digital PCR (ddPCR)Absolute quantification of DNA via droplet partitioningSwabs, CSF, blood, salivaHigh precision and sensitivity; ideal for low viral loadsExpensive, prone to contamination, requires specialized equipment
Loop-Mediated Isothermal Amplification (LAMP)Amplifies DNA at a constant temperature, eliminating thermal cyclingSwabs, CSF, blood, ocular fluidsRapid, field-friendly; no complex equipment neededLess specific than PCR; primer design is critical
Microarrays Hybridization-based detection of thousands of DNA fragments simultaneouslyBlood, CSF, tissue samplesHigh throughput; can detect multiple pathogens at onceHigh cost; requires complex data interpretation
BiosensorsConverts DNA-binding events into electrical, optical, or acoustic signalsSaliva, blood, ocular fluidsRapid, portable; cost-effectiveLimited availability; requires advanced transducer technologies
CRISPR
Technology
CRISPR-Cas systems target and detect viral DNA/RNASwabs, CSF, blood, salivaHigh sensitivity and specificity; potential for multiplexingPre-amplification often required; contamination risk
Next-Generation Sequencing (NGS)High-throughput sequencing for comprehensive pathogen analysisBlood, CSF, biopsies, respiratory samplesDetects co-infections and mutations; ideal for unknown pathogensExpensive; requires bioinformatics expertise
Table 3. DNA-based molecular techniques for herpesvirus infection diagnosis described in this review.
Table 3. DNA-based molecular techniques for herpesvirus infection diagnosis described in this review.
VirusMolecular Method of
Viral Genome Detection
Type of SampleClinical ApplicationsReferences
HSV-1qPCRCSF, saliva, blood, swabs from lesions, BALRapid detection of herpes encephalitis; diagnosing oral/genital herpes; outbreak monitoring[65,66]
Multiplex qPCRCSF, feces, urine, saliva, blood, swabs, corneal scrapes, biopsiesDetects multiple viruses; rapid herpes encephalitis diagnosis; early infection detection in transplant patients[67,68]
Droplet Digital PCRCSF, blood, saliva, urine, swabsAccurate monitoring of viral replication and treatment response; detects low-concentration samples[69,70]
NGSBlood, NPAs, lung lavage, stomach fluidDetects unknown or co-infections; suitable for low viral loads[71]
HSV-2qPCRCSF, swabs from genital lesions, bloodDiagnoses genital herpes and HSV-related meningitis; rapid detection of genital herpes[72,73]
Multiplex qPCRCSF, blood, cervical, endocervical, vaginal samplesRapid detection of multiple viruses; comprehensive diagnostics for genital infections[74,75]
Droplet Digital PCRCSF, blood, genital lesion swabsMonitors viral load; co-detection of HSV-1 and HSV-2[76,77]
NGSCSF, blood, lesion swabsHigh sensitivity; useful for early-stage infection or co-infection detection[78]
VZVqPCRVesicle fluids, swabs, crusts, CSF, amniotic fluidDiagnosis of central nervous system infections, prenatal congenital varicella syndrome[74,75]
Nested PCRVesicle fluids, swabs, CSF, amniotic fluidDetects VZV in skin lesions, encephalitis, meningitis[79]
NGSCSFHigh sensitivity; identifies multiple pathogens in CNS samples[80,81]
EBVqPCRCSF, peripheral blood, gingival/salivary swabsMononucleosis diagnosis; monitoring EBV in cancers; oral hairy leukoplakia detection[82,83,84]
NGSBiopsy samplesIdentifies cancer associations (e.g., NPC, Burkitt’s lymphoma)[85]
CMVqPCRBlood, saliva, urine, amniotic fluidDiagnoses congenital infections; monitors immunocompromised patients[86]
NGSAqueous humorDetects ocular infections and antiviral resistance[87]
HHV-6qPCROral samples, CSF, blood, tissuesIdentifies active infection; monitors transplant patients for graft-related complications[88,89]
Multiplex qPCRCSF, oral samples, blood, tissuesRapid encephalitis detection; post-transplant viral monitoring; classification of HHV-6 A and B [90,91]
Droplet Digital PCROral samples, CSF, bloodDetects chromosomally integrated HHV-6 (ciHHV-6); quantifies low-grade infections[92]
Conventional PCRCSF, oral samples, blood, serumDifferentiates between latent and active infections and HHV-6 A and B; standardized methods and rapid results[89]
NGSCSFComprehensive virome analysis; tracks neurotropic impacts[93]
HHV-7qPCRBlood, CSF, salivaRapid detection of reactivations; diagnoses conditions like pityriasis rosea[94]
Multiplex qPCRBlood, CSF, salivaScreens blood donors; detects ocular infections[95,96]
Droplet Digital PCRBlood, CSF, salivaMonitors viral loads in immunocompromised patients[97]
HHV-8qPCRSaliva, blood, semenDetects Kaposi’s sarcoma-associated virus; monitors immune-suppressed patients[98,99]
Nested PCRSaliva, blood, semenIdentifies early lesions; frequent monitoring for disease progression[100]
Droplet Digital PCRSaliva, blood, semenMonitors therapy response; tracks epidemiological transmission patterns[101]
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Maini, G.; Cianci, G.; Ferraresi, M.; Gentili, V.; Bortolotti, D. DNA-Based Technology for Herpesvirus Detection. DNA 2024, 4, 553-581. https://doi.org/10.3390/dna4040037

AMA Style

Maini G, Cianci G, Ferraresi M, Gentili V, Bortolotti D. DNA-Based Technology for Herpesvirus Detection. DNA. 2024; 4(4):553-581. https://doi.org/10.3390/dna4040037

Chicago/Turabian Style

Maini, Gloria, Giorgia Cianci, Matteo Ferraresi, Valentina Gentili, and Daria Bortolotti. 2024. "DNA-Based Technology for Herpesvirus Detection" DNA 4, no. 4: 553-581. https://doi.org/10.3390/dna4040037

APA Style

Maini, G., Cianci, G., Ferraresi, M., Gentili, V., & Bortolotti, D. (2024). DNA-Based Technology for Herpesvirus Detection. DNA, 4(4), 553-581. https://doi.org/10.3390/dna4040037

Article Metrics

Back to TopTop