International Journal of Molecular Medicine is an international journal devoted to molecular mechanisms of human disease.
International Journal of Oncology is an international journal devoted to oncology research and cancer treatment.
Covers molecular medicine topics such as pharmacology, pathology, genetics, neuroscience, infectious diseases, molecular cardiology, and molecular surgery.
Oncology Reports is an international journal devoted to fundamental and applied research in Oncology.
Experimental and Therapeutic Medicine is an international journal devoted to laboratory and clinical medicine.
Oncology Letters is an international journal devoted to Experimental and Clinical Oncology.
Explores a wide range of biological and medical fields, including pharmacology, genetics, microbiology, neuroscience, and molecular cardiology.
International journal addressing all aspects of oncology research, from tumorigenesis and oncogenes to chemotherapy and metastasis.
Multidisciplinary open-access journal spanning biochemistry, genetics, neuroscience, environmental health, and synthetic biology.
Open-access journal combining biochemistry, pharmacology, immunology, and genetics to advance health through functional nutrition.
Publishes open-access research on using epigenetics to advance understanding and treatment of human disease.
An International Open Access Journal Devoted to General Medicine.
Herpes simplex virus (HSV), including types HSV-1 and HSV-2, is the primary agent of genital herpes (GH) and is a globally prevalent virus. HSV-1 and HSV-2 are large double-stranded DNA viruses that belong to the Herpesviridae family, which can be found in several populations, in both urban and remote areas (1,2). HSV-1 commonly causes painful orofacial herpes, as well as life-threatening meningitis, keratitis, encephalitis and neonatal infections (3). GH, caused by HSV-1 or HSV-2, is a prevalent and highly communicable sexually transmitted infection, which is commonly characterized by recurrent, self-limited genital ulcers (4,5). Notably, GH is associated with notable psychological and physiological burdens on affected individuals. In addition to the acute symptoms, GH commonly results in emotional, social, relational and sexual distress. However, evidence regarding the most effective interventions for improving health-related quality of life is still lacking (6). Both HSV-2 and HSV-1 can cause genital ulcers, with HSV-2 being the predominant sexually transmitted type and a major risk factor for increased human immunodeficiency virus 1 (HIV-1) acquisition during co-infection (7–9). Although there is currently no cure, antiviral therapies such as acyclovir can improve symptoms, decrease the frequency of outbreaks, and reduce the risk of transmission to partners (10). Therefore, a comprehensive understanding of the biological mechanisms underlying GH recurrence is essential for improving clinical management and treatment strategies.
Metabolism, encompassing glycolysis, oxidative phosphorylation (OXPHOS) and fatty acid oxidation metabolic pathways, can affect the phenotypes and functions of immune cells. Metabolic regulation of the immune system serves a crucial role in the development and progression of numerous diseases, including cancer, autoimmune disorders and metabolic conditions (11–13). Alterations in metabolite profiles reflect physiological states, and also provide critical insights into cellular functions, tissue metabolism and disease progression (14). Advancements in metabolomics, the comprehensive study of metabolites within biological systems, have enabled the identification of disease-specific pathophysiological characteristics (14–17). Therefore, the present study aimed to investigate serum metabolite changes in patients with recurrent HSV-2 GH, thus offering novel insights into its pathogenesis.
Based on the aforementioned evidence linking metabolism, immunity and disease pathogenesis (10–13), it was hypothesized that metabolic alterations could contribute to GH recurrence via modulating immune cell function. To evaluate this hypothesis, a comprehensive analysis integrating metabolomics, transcriptomics and single-cell RNA-sequencing (scRNA-seq) was carried out to identify metabolic pathways associated with HSV-2 GH recurrence. Through these comprehensive analyses, the current study aimed to uncover the potential molecular mechanisms underlying GH pathogenesis, and to identify novel biomarkers and therapeutic targets.
Blood samples were collected from 14 patients with recurrent HSV-2 GH and 14 healthy volunteers at Sir Run Run Shaw Hospital (Hangzhou, China) between March and June 2024. All individuals provided written informed consent prior to study enrollment. For untargeted metabolomics analysis, serum samples were collected after centrifugation at 12,000 × g and 4°C for 15 min, and were stored at −80°C. Peripheral blood mononuclear cells (PBMCs) were isolated through density gradient centrifugation at 400 × g and 25°C for 30 min, and were utilized for RNA-seq and scRNA-seq. To control for potential hormonal confounding factors, this study exclusively enrolled female participants. The inclusion criteria for patients with GH were as follows: Female participants, aged 18-35 years (mean ± SD, 25.9±4.8 years; median, 25 years); clinically confirmed recurrent GH with HSV-2 PCR positivity and ≥3 annual recurrences; absence of comorbidities; and no prior HSV vaccination. The exclusion criteria were as follows: Pregnant women; treatment with antiviral medications or immunosuppressants within the past 2 weeks; and those with any history of HSV vaccination, diabetes, metabolic syndrome or autoimmune diseases. The inclusion criteria for the participants in the healthy control (HC) group were: Female participants, aged 18-35 years (mean ± SD, 26.6±4.9 years; median, 27 years); no history of GH; negative for HSV infection; and no prior HSV vaccination. The demographic and clinical characteristics of the study participants are listed in Table SI.
Serum samples were stored at −80°C and untargeted metabolomics profiling was performed by LC-Bio Technologies (Hangzhou) Co., Ltd. Samples were randomized based on GH and HC. Following centrifugation at 13,800 × g for 15 min at 4°C, the supernatant was carefully collected into fresh glass vials for subsequent analysis. For quality control (QC) purposes, an aliquot from each biological replicate was pooled to create a representative QC sample.
Chromatographic separation was performed using a Thermo Vanquish Flex UPLC system (Thermo Fisher Scientific, Inc.) equipped with an ACQUITY UPLC T3 column (100×2.1 mm, 1.8 µm; Waters Corporation) maintained at 40°C with a constant flow rate of 0.35 ml/min, using mobile phase A (5 mmol/l ammonium acetate and 5 mmol/l acetic acid in water) and B (LC-MS grade acetonitrile) with the following gradient program: 0.0-0.5 min (99-1% B), 0.5-6.5 min (1-99% B), 6.5-8.0 min (99% B), 8.0-8.5 min (99-1% B) and 8.5-10.0 min (1% B). High-resolution mass spectrometry analysis was conducted on an Orbitrap Exploris 120 instrument (Thermo Fisher Scientific, Inc.) operating in alternating positive (+3,800 V) and negative (−3,400 V) ionization modes with optimized source parameters [sweep gas 1, auxiliary gas 12, sheath gas 50 (arbitrary units), source temperature 350°C]. Data-dependent acquisition was performed with full MS scans (70-1,050 m/z) at 60,000 resolution (200 m/z) using standard automatic gain control (AGC) and automatic maximum injection time, selecting the top four most intense precursor ions (intensity threshold >5,000) for MS/MS fragmentation at 15,000 resolution with custom AGC and injection time settings (6 sec dynamic exclusion). System stability was monitored by analyzing quality control samples every 10 injections, with inter-QC mass deviation used for batch correction.
The analysis was carried out on the Triple TOF 5,600 Plus high-resolution tandem mass spectrometer system (MS/MS; SCIEX) in both positive and negative ion modes. Chromatographic separation was achieved using an ultra-performance liquid chromatography system (SCIEX ExionLC 2.0+ system), and the resulting LC-MS data were preprocessed using XCMS software version 3.8.5 (https://github.com/sneumann/xcms). All data from the untargeted platform (semi-quantitative) were normalized based on the minimum level detected for each metabolite/run and were reported as log2 signal-to-noise ratios. Kyoto Encyclopedia of Genes and Genomes (KEGG) metabolic pathway enrichment analysis were performed using the MetaboAnalyst (https://www.metaboanalyst.ca/) database. The metabolomics data are listed in Table SII.
Blood samples were collected from patients with recurrent HSV-2 GH and healthy volunteers. After density gradient centrifugation, the PBMC were immediately immersed in liquid nitrogen for freezing. The frozen samples were then thoroughly ground using a pre-cooled mortar. Subsequently, cells were lysed with 1 ml TRIzol® reagent (Invitrogen; Thermo Fisher Scientific, Inc.). Following the addition of 200 µl chloroform, the mixture was vigorously shaken for 15 sec, followed by incubation at room temperature for 2 min. The samples were then centrifuged at 12,000 × g for 15 min at 4°C, and the supernatant was transferred to a new tube. To precipitate RNA, each tube was supplemented with 0.5 ml isopropyl alcohol, followed by gently mixing prior to incubation at room temperature for 10 min. After a second centrifugation at 12,000 × g for 10 min at 4°C, the supernatant was discarded. The RNA pellet was then washed with 1 ml 75% ethanol and the supernatant was discarded following centrifugation at 7,500 × g for 5 min at 4°C. After air drying, total RNA was dissolved in DEPC-treated water.
For second-generation sequencing, libraries were prepared as previously described (18). In brief, RNA quantity and integrity were assessed using the Qubit™ 3.0 Fluorometer (cat. no. Q33216; Thermo Fisher Scientific, Inc.) and Agilent 5300 Fragment Analyzer System (cat. no. M5310AA; Agilent Technologies) respectively. RNA samples demonstrating high purity (A260/A280 ratio 1.8-2.2) and RNA integrity numbers >7.0 were selected for subsequent library preparation using Illumina-compatible protocols. mRNA was purified from 2 µg total RNA using mRNA Capture Beads 2.0 (cat. no. 12629ES; Shanghai Yeasen Biotechnology Co., Ltd.) through two rounds of poly(A) selection.
Purified mRNA was fragmented in magnesium-containing fragmentation buffer (cat. no. 12340ES97; Shanghai Yeasen Biotechnology Co., Ltd.) at 94°C for 5 min. First-strand cDNA was synthesized using random hexamer-primed reverse transcription (SuperScript™ IV Reverse Transcriptase; Thermo Fisher Scientific, Inc.). Second-strand cDNA was generated using a dUTP incorporation strategy with E. coli DNA Polymerase I, RNase H and dUTP solution (cat. no. 12340ES97; Shanghai Yeasen Biotechnology Co., Ltd.). Blunt-ended cDNA fragments were adenylated at 3′ ends using Klenow Fragment (3′-5′ exo-) and ligated to Illumina-compatible forked adapters containing T-overhangs. PCR products were size-selected (400±50 bp inserts) using Hieff NGS DNA Selection Beads (cat. no. 12601ES75; Shanghai Yeasen Biotechnology Co., Ltd.). Strand specificity was maintained through dUTP-based strand marking and uracil excision. Libraries were sequenced in 2×150 bp paired-end mode on an Illumina NovaSeq™ X Plus platform (LC-bio Technologies Hangzhou Co. Ltd.) following manufacturer-recommended protocols.
To identify DEGs between patients with GH and healthy volunteers, the ‘DESeq2’ R package (version 4.5; http://bioconductor.org/packages/release/bioc/html/DESeq2.html) in R (version 4.4.3; http://www.R-project.org/) was utilized. Genes showing log2 fold change of ≥1.0 and an adjusted P-value of <0.05 were considered significantly differentially expressed. The RNA-seq data are presented in Table SIII.
KEGG (http://www.genome.jp/kegg/), a comprehensive database for understanding biological systems at the genomic and molecular levels, DEGs of scRNA-seq data was used for pathway enrichment analysis. This resource integrates genomic, chemical and systematic information on biological interactions, reactions and disease pathways.
GSEA was performed based on the detected metabolites using KEGG metabolite sets (19). The analysis aims to identify significant pathways that are associated with the metabolite profiles. The criteria for selecting significant pathways were set as follows: Pathways with an absolute normalized enrichment score (|NES|) >1 and a P-value <0.05 were considered statistically significant. These thresholds help to filter out the most relevant pathways that show a strong association with the metabolic differences observed in the study.
scRNA-seq of PBMCs and neutrophils from GH (n=4) and healthy controls (n=4) was performed using the Chromium Single Cell 3′ Chip and 10× Genomics Chromium Single-Cell 3′ Kit v3 (10× Genomics, LC-Bio Technologies) as previously described (20). The sequencing data were then deposited in the Genome Sequence Archive for human database. Bioinformatics analysis utilized Cell Ranger (v7.1.0; 10× Genomics; http://www.10×genomics.com/support/software/cell-ranger) and Seurat (v4.3.0; Satija Lab; http://satijalab.org/seurat) for data processing, clustering, and differential expression analysis. Nonlinear dimensionality reduction was performed using t-distributed Stochastic Neighbor Embedding implemented in Seurat v4.3.0.
Total RNA was isolated from PBMCs using TRIzol® Reagent (cat. no. 15596018CN; Invitrogen; Thermo Fisher Scientific, Inc.). cDNA was synthesized from 1 µg RNA using oligo dT primer and the PrimeScript RT PCR master mix Kit (cat. no. RR036A; Takara Biotechnology Co., Ltd.) under the following conditions: 37°C for 15 min and 85°C for 5 min. qPCR analysis was carried out using the SYBR Green Master Mix kit (cat. no. 04707516001; Roche Diagnostics) with 95°C for 5 min (initial denaturation) and 40 cycles of 95°C for 5 sec (denaturation) and 60°C for 1 min (annealing and extension). The relative gene expression levels are expressed as fold changes relative to control samples, and were quantified using the 2−ΔΔCq comparative threshold cycle method (21). ACTB, a housekeeping gene, served as an endogenous reference to normalize target gene expression levels across all experimental groups. The primer sequences are listed in Table SIV.
All data were analyzed using SPSS statistics software (version 25.0; IBM Corp.). The results are presented as the mean ± standard error of the mean. The differences were compared using an unpaired Student's t-test. P<0.05 was considered to indicate a statistically significant difference.
To investigate the alterations in metabolites associated with recurrent HSV-2 GH, peripheral blood samples were collected and serum was isolated for comprehensive metabolic assays. The analysis identified a total of 78 upregulated and 100 downregulated metabolites in patients with GH (Fig. 1A). Notably, the top five downregulated metabolites were Deferiprone, ANTHOTHECOL Glabranin, 9(S)-HOTrE and Farrerol; conversely, 2-Ketobutyric acid, Butanal, L-sorbose, PSICOSE and 5-Methoxypsoralen were the five most significantly upregulated metabolites in the serum of patients with recurrent GH (Fig. 1B). To further explore the functional implications of these differentially expressed metabolites, KEGG enrichment analysis on the metabolomics data was performed. The top 20 KEGG-enriched pathways in serum included ‘Glycerophospholipid metabolism’, ‘Choline metabolism in cancer’, ‘Retrograde endocannabinoid signaling’, ‘Cholesterol metabolism’ and ‘Linoleic acid metabolism’ (Fig. 1C). Notably, key metabolites in the Glycerophospholipid metabolism pathway, including O-phosphocholine, PS (18:0/20:4(8Z,11Z,14Z,17Z)) and PE (16:0/18:2), were markedly upregulated in the serum of patients with GH compared with HCs (Fig. 1D). However, the majority of cholesterol-related metabolites were downregulated in patients with GH compared with HCs, except O-phosphocholine (Fig. 1D). Additionally, the levels of metabolites associated with the linoleic acid synthesis pathway, such as 9R,10S-EpOME, PC (18:3/20:1), PC [20:0/14:1(9Z)], 9-KODE and PC [22:0/14:1(9Z)], were significantly reduced in the serum of patients with recurrent GH compared with HCs (Fig. 1D). Overall, these results could provide novel insights into the metabolic alterations associated with GH.
GSEA of the metabolomics data revealed significant differences in the activation of particular metabolic pathways between patients with recurrent HSV-2 GH and HCs. As shown in Fig. 2A, among the top 30 metabolic pathways, 24 were notably enriched in the GH group, while six were enriched in the normal control group. Notably, pathways, such as ‘Biosynthesis of amino acids’, ‘Biosynthesis of phenylpropanoids’, ‘Tropane, piperidine and pyridine alkaloid biosynthesis’, ‘2-Oxocarboxylic acid metabolism’ and ‘Glycine, serine and threonine metabolism’ were significantly enriched in the GH group (Fig. 2A and B). Conversely, the ‘neuroactive ligand-receptor interaction pathway’ was markedly downregulated in patients with recurrent HSV-2 GH compared with HCs (Fig. 2A and C). Metabolomics heatmap data indicated that metabolites associated with neuroactive ligand-receptor interactions, such as palmitoylethanolamide, melatonin and sphingosine-1 phosphate, were all downregulated in the serum samples of patients with recurrent HSV-2 GH compared with HCs (Fig. 2D). Furthermore, the levels of metabolites involved in amino acid biosynthesis, including 4-aminobenzoic acid, 6,7-dihydroxycoumarin, L-tryptophan, bergapten and 5-methoxypsoralen, were notably enhanced in the GH group compared with HCs (Fig. 2D).
To elucidate the potential mechanisms underlying GH-related morbidity and to explore the association between metabolic changes and transcriptional profiles, RNA-seq analysis was conducted on blood cells from healthy individuals and patients with recurrent HSV-2 GH. The results of RNA-seq analysis revealed significant gene expression alterations in patients with recurrent HSV-2 GH, particularly upregulation of genes associated with glycerophospholipid metabolism, cholesterol metabolism and amino acid biosynthesis pathways (Fig. 3A-F). These results were consistent with those obtained in the metabolomics analysis. Several genes associated with the glycerophospholipid pathway, including ETNK1, CRLS1, PLA2G4A, GPD2, PISD, AGPAT5, PLPP3, GPAT3, PLA2G4C and DGKK, were significantly upregulated in the blood cells of patients with recurrent GH compared with HCs (Fig. 3A and B). Similarly, genes associated with cholesterol metabolism, such as SOS1, PLA2G4A, PDGFB, PIK3R3, PDGFC, WASF3, SLC44A1 and PLPP3, were also notably upregulated in the same samples (Fig. 3C and D). In terms of the amino acid biosynthesis pathway, the expression of several genes was also significantly increased in the GH group compared with in the HC group, including that of IDH1, PSAT1, ACO1, RPE, GOT1, GPT2, PRPS2, PSPH and SHMT1 (Fig. 3E and F). RT-qPCR analysis also verified that ETNK1, PLA2G4A, SOS1, GPD2, IDH1 and PSAT1 were significantly upregulated in blood cells from patients with recurrent HSV-2 GH compared with in the HC group (Fig. 3G). These results, combined with the metabolomic data, revealed significant changes in the expression of several metabolites and the activation status of relevant pathways, including those of glycerophospholipid metabolism, cholesterol metabolism and amino acid biosynthesis, in patients with recurrent HSV-2 GH.
To identify the cell population with enhanced expression of differentially expressed metabolism-related genes, the scRNA-seq data in patients with recurrent HSV-2 GH and HC was analyzed, and the clusters of cells in the PBMCs and neutrophils were observed (Fig. 4A). The results demonstrated that IDH1 was highly expressed in monocytes, dendritic cells (DCs) and plasmacytoid DCs (pDCs), with notably elevated expression in monocytes from the GH group (Fig. 4B and C). GOT1 and GOT2 were also markedly upregulated in pDCs and DCs (Fig. 4B and C). Additionally, ETNK1 was highly expressed in B cells, monocytes, DCs and pDCs (Fig. 4B and C). Overall, the scRNA-seq analysis revealed distinct metabolic gene expression patterns across various immune cell subpopulations, including T cells, B cells, monocytes, neutrophils and DCs.
Subsequently, KEGG enrichment analysis of the metabolism-related genes, which were identified by RNA-seq analysis, was carried out. Several significantly enriched pathways were detected, including ‘carbon metabolism’, ‘folate biosynthesis’, ‘valine, leucine and isoleucine degradation’ and ‘alanine, aspartate and glutamate metabolism’ (Fig. 5A). The volcano plot for carbon metabolism revealed a significant number of upregulated genes (n=31) and a few downregulated genes (n=2); among the significantly upregulated genes were IDH1, PSAT1 and RPE, which all have crucial roles in various metabolic processes (22–24) (Fig. 5B). The alanine, aspartate and glutamate metabolism pathway also showed significant changes, with eight upregulated and one downregulated gene. Key genes, such as GOT1 and GPT2, were significantly upregulated in patients with recurrent HSV-2 GH, thus supporting their potential role in the altered metabolic state associated with GH recurrence (Fig. 5C). The folate biosynthesis pathway also exhibited significant changes, with eight upregulated genes and one downregulated gene. Notably, CBR1 and DHFR were significantly altered, suggesting their involvement in the metabolic dysregulation associated with GH recurrence (Fig. 5D). The valine, leucine and isoleucine degradation pathway had 15 upregulated genes, with ACADM and ALDH6A1 being particularly significant (Fig. 5E). The pronounced upregulation of these genes could indicate a shift in amino acid metabolism in patients with recurrent HSV-2 GH. The aforementioned findings underscored the complex metabolic reprogramming associated with GH recurrence, with multiple pathways being significantly affected.
To further analyze the metabolic profiles in different immune cells, the scRNA-seq data were further assessed using KEGG enrichment analysis. All immune cells were highly enriched in the ‘Oxidative phosphorylation’ pathway (Fig. 6A, C, E and G). Monocytes displayed a significant enrichment in the ‘Oxidative phosphorylation’ and ‘Pyrimidine metabolism’ pathways. Furthermore, almost all of the mitochondrial genes, including MT-CYB, MT-ND4 and MT-CO3, were highly expressed in monocytes from patients with recurrent HSV-2 GH, thus indicating robust metabolic activity, particularly within the OXPHOS pathway (Fig. 6A and B). In addition, significant upregulation of mitochondrial genes associated with OXPHOS, such as MT-CYB and MT-ND4, were recorded in T and B cells, and DCs (Fig. 6C-H). These findings suggested that the OXPHOS pathway-associated mitochondrial genes could display functional roles and responses during this viral infection.
By integrating multiple approaches, including transcriptomics, metabolomics and scRNA-seq analyses, significant changes in pathways associated with lipid metabolism, amino acid biosynthesis and cholesterol metabolism were identified in patients with GH. Genes associated with the alanine, aspartate and glutamate, and valine, leucine and isoleucine metabolism pathways were significantly upregulated in patients with recurrent HSV-2 GH. In addition, several metabolism-related genes were upregulated in PBMCs from patients with GH, including IDH1 and ETNK1, the expression of which was notably increased in DCs and monocytes. Additionally, mitochondrial genes involved in OXPHOS, such as MT-CYB and MT-CO3, were significantly elevated across monocytes, DCs, T cells and B cells. Overall, these findings indicated metabolic reprogramming in patients with recurrent HSV-2 GH, thus providing potential biomarkers and therapeutic targets for the development of future treatment strategies.
Glycerophospholipid remodeling serves a critical role in several diseases, including orthoflavivirus infection, brain injury, Pseudomonas infections and cancer (25–27). A previous study demonstrated that viral infections can hijack the host cell glycerophospholipid metabolism to acquire lipid components for viral envelope synthesis, such as phosphatidylserine and phosphatidylinositol. Inhibiting related metabolic enzymes, such as those related to phosphatidylinositol biosynthesis, could markedly reduce viral titers and cytopathic effects (25,28). The present study showed that glycerophospholipid metabolism was upregulated in patients with GH, thus indicating that glycerolipid biosynthesis could represent a conserved host dependency factor exploited by this evolving virus. Dysregulated cholesterol metabolism in arterial macrophages has long been known to be pathogenic in atherosclerosis, and in an ILC2-driven airway inflammation model lipid metabolism dysregulation can also be found (29,30). The present data also found downregulation of cholesterol-related metabolites, which further supported the occurrence of a potential lipid metabolism dysfunction in patients with GH.
Amino acids, the fundamental building blocks of proteins, and related metabolites, derived from both microorganisms and host cells, serve a crucial role in regulating immune cell activation and antibody production, thus markedly affecting several biological processes, such as inflammation and immune responses (31,32). Recent studies have identified key intermediate metabolites in the aromatic amino acid pathways that undergo alterations during sepsis, influenza and severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infections (33–35). Herein, the results showed a significant upregulation of amino acid biosynthesis in patients with recurrent HSV-2 GH, thus suggesting that aberrant amino acid biosynthesis could be involved in the pathophysiology of this disease. A previous Mendelian randomization study revealed that SARS-CoV-2 infection can affect susceptibility to HSV-1 and HSV-2. While SARS-CoV-2 infection could suppress HSV-1 reactivation via inducing a type I interferon response, it could also promote HSV-2 infection via immunosuppression or an inflammatory environment (36). Based on the results of this study, the effects between SARS-CoV-2 and GH could also be mediated by several metabolic pathways.
A previous study reported that serum amino acid metabolism varies at different time points during influenza virus infection (35). The levels of several amino acids, such as valine, proline, citrulline, isoleucine, asparagine and arginine, and their derivatives may be altered following viral infection, and these changes could be associated with disease severity (37). In the present study, the RNA-seq results also revealed that several amino acid metabolic pathways, such as alanine, aspartate and glutamate, and valine, leucine and isoleucine pathways, were enriched in recurrent HSV-2 GH samples. These amino acid metabolism profiles could indicate GH relapse. Furthermore, Zhang et al (38) revealed that SARS-CoV-2 exploits folate and one-carbon metabolism to promote its replication. Consistently, the present study also revealed that folate biosynthesis metabolism was upregulated in patients with recurrent HSV-2 GH. Overall, the aforementioned findings suggested that targeting host metabolic pathways could be a promising therapeutic approach against future GH infection.
OXPHOS is a vital mitochondrial metabolic pathway, which is involved in the generation of adenosine triphosphate through the electron transport chain and chemiosmosis (39). Emerging evidence has suggested that viruses can manipulate host OXPHOS to regulate immune responses, thus enhancing their survival and replication (40). For example, a previous study demonstrated that HIV can preferentially target glycolytic CD4+ T cells by relying on elevated OXPHOS activity regardless of their differentiation and activation status (41). Additionally, liver biopsies from patients with chronic hepatitis C virus have revealed defects in OXPHOS, along with increased expression of oxidative stress-related markers (42). In patients with SARS-CoV-2, the levels of OXPHOS regulators, such as those of mitochondrial nuclear retrograde regulator 1, have been shown to be reduced in heart tissues, potentially contributing to cardiac complications. Furthermore, the expression levels of core mitochondrial genes are also decreased during SARS-CoV-2 infection in both rodent and human hosts (43,44). Herein, the results showed that the expression levels of mitochondrial genes involved in OXPHOS, including MT-CYB, MT-ND4 and MT-CO3 were significantly elevated in monocytes, T cells and B cells derived from patients with GH. Another study revealed that HSV could dysregulate lipid synthesis and induce ferroptosis via enhancing glucose-mediated OXPHOS and glutamine-dependent reductive carboxylation (45). Additionally, a previous study demonstrated that following viral infection, pDCs can retain their ability to secrete type I interferons through lactate dehydrogenase B-dependent metabolic adaptation (46). Overall, these findings suggested that OXPHOS modulation could be viral- and cell type-specific.
Although the current study provided valuable insights, there are several limitations that warrant consideration. Firstly, the sample size was not sufficient to generalize the findings across diverse populations, thus highlighting the need for further research to validate the identified biomarkers. Additionally, although associations between metabolic changes and gene expression were recorded, the underlying causal associations remain to be fully elucidated. Future studies should focus on investigating the functional roles of these metabolites and genes, as well as their interactions within the immune system. Investigating tissue-specific metabolic changes and their effect on immune cell function could yield a more comprehensive understanding of the disease. Furthermore, longitudinal studies should be performed to assess how metabolic alterations evolve over time, and how these changes could be involved in disease progression and recurrence. The current study offers compelling evidence of systemic metabolic reprogramming in recurrent HSV-2 GH and highlights novel potential therapeutic targets. However, the research cohort specifically comprised patients diagnosed with HSV-2-positive GH. Clinically, the recurrence rate of HSV-1 GH is markedly lower than that of HSV-2, primarily due to a reduced propensity for neuronal infection (47). Over the past two decades, HSV-1 has increasingly become the predominant causative agent of first-episode GH in multiple countries, reflecting a shifting epidemiological landscape (48,49). Future studies incorporating rigorous HSV typing are essential to elucidate potential subtype-specific metabolic vulnerabilities.
In conclusion, the current study revealed significant metabolic and transcriptional alterations in patients with recurrent HSV-2 GH, thus providing a foundation for future studies on metabolic interventions for HSV-2 GH recurrence and highlighting potential biomarkers for therapeutic interventions.
Not applicable.
The present study was supported by grants from the Natural Science Foundation of China (grant nos. 82373491 and 82471846) and Peterson's Lab MED Translational Medicine Fund (grant no. 20240131).
The RNA-seq data generated in the present study may be found in the Sequence Read Archive database under accession number PRJNA1267039 or at the following URL: (https://www.ncbi.nlm.nih.gov/bioproject/?term=PRJNA1267039). The metabolomics data generated in the present study may be found in the National Genomics Data Center database under accession number PRJCA040378 or at the following URL: (https://ngdc.cncb.ac.cn/search/specific?db=bioproject&q=PRJCA040378). The scRNA-seq data generated in the present study may be found in the ScienceDB database (https://www.scidb.cn/en/detail?dataSetId=690475ca10ed4424bdc0f69c3b5cd701).
JH, YX, HC and YS conceived and designed the study. JH, CF, JZ and YC performed the experiments. JH, YX and SC analyzed the data. JH, HC and JZ wrote and reviewed the manuscript. YS and HC assume overall responsibility for the manuscript. JH and HC confirm the authenticity of all the raw data. YS and HC were responsible for study supervision and funding acquisition. All authors read and approved the final manuscript.
The present study was approved by the Ethics Committee of Sir Run Run Shaw Hospital, affiliated with Zhejiang University School of Medicine (approval no. 20240106). Written informed consent was obtained from all participants prior to their inclusion in the study.
Not applicable.
The authors declare that they have no competing interests.
|
US Preventive Services Task Force, . Mangione CM, Barry MJ, Nicholson WK, Cabana M, Chelmow D, Coker TR, Davis EM, Donahue KE, Jaén CR, et al: Serologic screening for genital herpes infection: US preventive services task force reaffirmation recommendation statement. JAMA. 329:502–507. 2023. View Article : Google Scholar : PubMed/NCBI | |
|
Havens JL, Calvignac-Spencer S, Merkel K, Burrel S, Boutolleau D and Wertheim JO: Phylogeographic analysis reveals an ancient East African origin of human herpes simplex virus 2 dispersal out-of-Africa. Nat Commun. 13:54772022. View Article : Google Scholar : PubMed/NCBI | |
|
Johnston C, Gottlieb SL and Wald A: Status of vaccine research and development of vaccines for herpes simplex virus. Vaccine. 34:2948–2952. 2016. View Article : Google Scholar : PubMed/NCBI | |
|
Omarova S, Cannon A, Weiss W, Bruccoleri A and Puccio J: Genital herpes simplex virus-an updated review. Adv Pediatr. 69:149–162. 2022. View Article : Google Scholar : PubMed/NCBI | |
|
James C, Harfouche M, Welton NJ, Turner KM, Abu-Raddad LJ, Gottlieb SL and Looker KJ: Herpes simplex virus: global infection prevalence and incidence estimates, 2016. Bull World Health Organ. 98:315–329. 2020. View Article : Google Scholar : PubMed/NCBI | |
|
Bennett C, Rebafka A, Carrier J, Cook S and Edwards D: Impact of primary and recurrent genital herpes on the quality of life of young people and adults: A mixed methods systematic review. JBI Evid Synth. 20:1406–1473. 2022. View Article : Google Scholar : PubMed/NCBI | |
|
Desai DV and Kulkarni SS: Herpes simplex virus: The interplay between HSV, Host, and HIV-1. Viral Immunol. 28:546–555. 2015. View Article : Google Scholar : PubMed/NCBI | |
|
Lafferty WE, Downey L, Celum C and Wald A: Herpes simplex virus type 1 as a cause of genital herpes: Impact on surveillance and prevention. J Infect Dis. 181:1454–1457. 2000. View Article : Google Scholar : PubMed/NCBI | |
|
Masese L, Baeten JM, Richardson BA, Bukusi E, John-Stewart G, Graham SM, Shafi J, Kiarie J, Overbaugh J and McClelland RS: Changes in the contribution of genital tract infections to HIV acquisition among Kenyan high-risk women from 1993 to 2012. AIDS. 29:1077–1085. 2015. View Article : Google Scholar : PubMed/NCBI | |
|
Schalkwijk HH, Snoeck R and Andrei G: Acyclovir resistance in herpes simplex viruses: Prevalence and therapeutic alternatives. Biochem Pharmacol. 206:1153222022. View Article : Google Scholar : PubMed/NCBI | |
|
Ning L, Shishi Z, Bo W and Huiqing L: Targeting immunometabolism against acute lung injury. Clin Immunol. 249:1092892023. View Article : Google Scholar : PubMed/NCBI | |
|
Mockler MB, Conroy MJ and Lysaght J: Targeting T cell immunometabolism for cancer immunotherapy; Understanding the impact of the tumor microenvironment. Front Oncol. 4:1072014. View Article : Google Scholar : PubMed/NCBI | |
|
Fullerton MD, Steinberg GR and Schertzer JD: Immunometabolism of AMPK in insulin resistance and atherosclerosis. Mol Cell Endocrinol. 366:224–234. 2013. View Article : Google Scholar : PubMed/NCBI | |
|
Bone mineral densitometry, . Health and Public Policy Committee, American College of Physicians. Ann Intern Med. 109:8461988.PubMed/NCBI | |
|
Xu H, Zhou S, Tang Q, Xia H and Bi F: Cholesterol metabolism: New functions and therapeutic approaches in cancer. Biochim Biophys Acta Rev Cancer. 1874:1883942020. View Article : Google Scholar : PubMed/NCBI | |
|
Chen D, Zhang X, Li Z and Zhu B: Metabolic regulatory crosstalk between tumor microenvironment and tumor-associated macrophages. Theranostics. 11:1016–1030. 2021. View Article : Google Scholar : PubMed/NCBI | |
|
Di'Narzo AF, Houten SM, Kosoy R, Huang R, Vaz FM, Hou R, Wei G, Wang W, Comella PH, Dodatko T, et al: Integrative analysis of the inflammatory bowel disease serum metabolome improves our understanding of genetic etiology and points to novel putative therapeutic targets. Gastroenterology. 162:828–843. e112022. View Article : Google Scholar : PubMed/NCBI | |
|
Wang Z, Gerstein M and Snyder M: RNA-Seq: A revolutionary tool for transcriptomics. Nat Rev Genet. 10:57–63. 2009. View Article : Google Scholar : PubMed/NCBI | |
|
Canzler S and Hackermuller J: multiGSEA: A GSEA-based pathway enrichment analysis for multi-omics data. BMC Bioinformatics. 21:5612020. View Article : Google Scholar : PubMed/NCBI | |
|
Chen S, Zhu J, Hua C, Feng C, Wu X, Zhou C, Chen X, Zhang B, Xu Y, Ma Z, et al: Single-cell RNA sequencing reveals the diversity of the immunological landscape response to genital herpes. Virol Sin. 39:860–874. 2024. View Article : Google Scholar : PubMed/NCBI | |
|
Livak KJ and Schmittgen TD: Analysis of relative gene expression data using real-time quantitative PCR and the 2(−Delta Delta C(T)) Method. Methods. 25:402–408. 2001. View Article : Google Scholar : PubMed/NCBI | |
|
Zhang C, Yu JJ, Yang C, Yuan ZL, Zeng H, Wang JJ, Shang S, Lv XX, Liu XT, Liu J, et al: Wild-type IDH1 maintains NSCLC stemness and chemoresistance through activation of the serine biosynthetic pathway. Sci Transl Med. 15:eade41132023. View Article : Google Scholar : PubMed/NCBI | |
|
Wang J, Filippakis H, Hougard T, Du H, Ye C, Liu HJ, Zhang L, Hindi K, Bagwe S, Nijmeh J, et al: Interleukin-6 mediates PSAT1 expression and serine metabolism in TSC2-deficient cells. Proc Natl Acad Sci USA. 118:e21012681182021. View Article : Google Scholar : PubMed/NCBI | |
|
Jun S, Datta S, Wang L, Pegany R, Cano M and Handa JT: The impact of lipids, lipid oxidation, and inflammation on AMD, and the potential role of miRNAs on lipid metabolism in the RPE. Exp Eye Res. 181:346–355. 2019. View Article : Google Scholar : PubMed/NCBI | |
|
Hehner J, Schneider L, Woitalla A, Ott B, Vu KCT, Schöbel A, Hain T, Schwudke D and Herker E: Glycerophospholipid remodeling is critical for orthoflavivirus infection. Nat Commun. 15:86832024. View Article : Google Scholar : PubMed/NCBI | |
|
Dolce V, Cappello AR, Lappano R and Maggiolini M: Glycerophospholipid synthesis as a novel drug target against cancer. Curr Mol Pharmacol. 4:167–175. 2011. View Article : Google Scholar : PubMed/NCBI | |
|
Kondakova T, D'Heygere F, Feuilloley MJ, Orange N, Heipieper HJ and Duclairoir Poc C: Glycerophospholipid synthesis and functions in Pseudomonas. Chem Phys Lipids. 190:27–42. 2015. View Article : Google Scholar : PubMed/NCBI | |
|
Farley SE, Kyle JE, Leier HC, Bramer LM, Weinstein JB, Bates TA, Lee JY, Metz TO, Schultz C and Tafesse FG: A global lipid map reveals host dependency factors conserved across SARS-CoV-2 variants. Nat Commun. 13:34872022. View Article : Google Scholar : PubMed/NCBI | |
|
Yan J and Horng T: Lipid metabolism in regulation of macrophage functions. Trends Cell Biol. 30:979–989. 2020. View Article : Google Scholar : PubMed/NCBI | |
|
Karagiannis F, Masouleh SK, Wunderling K, Surendar J, Schmitt V, Kazakov A, Michla M, Hölzel M, Thiele C and Wilhelm C: Lipid-droplet formation drives pathogenic group 2 innate lymphoid cells in airway inflammation. Immunity. 52:620–634. e62020. View Article : Google Scholar : PubMed/NCBI | |
|
Costantini C, Bellet MM, Renga G, Stincardini C, Borghi M, Pariano M, Cellini B, Keller N, Romani L and Zelante T: Tryptophan co-metabolism at the host-pathogen interface. Front Immunol. 11:672020. View Article : Google Scholar : PubMed/NCBI | |
|
Dhankhar R, Gupta V, Kumar S, Kapoor RK and Gulati P: Microbial enzymes for deprivation of amino acid metabolism in malignant cells: Biological strategy for cancer treatment. Appl Microbiol Biotechnol. 104:2857–2869. 2020. View Article : Google Scholar : PubMed/NCBI | |
|
Maeda R, Seki N, Uwamino Y, Wakui M, Nakagama Y, Kido Y, Sasai M, Taira S, Toriu N, Yamamoto M, et al: Amino acid catabolite markers for early prognostication of pneumonia in patients with COVID-19. Nat Commun. 14:84692023. View Article : Google Scholar : PubMed/NCBI | |
|
Al-Shalan HAM, Zhou L, Dong Z, Wang P, Nicholls PK, Boughton B, Stumbles PA, Greene WK and Ma B: Systemic perturbations in amino acids/amino acid derivatives and tryptophan pathway metabolites associated with murine influenza A virus infection. Virol J. 20:2702023. View Article : Google Scholar : PubMed/NCBI | |
|
Chen Q, Liang X, Wu T, Jiang J, Jiang Y, Zhang S, Ruan Y, Zhang H, Zhang C, Chen P, et al: Integrative analysis of metabolomics and proteomics reveals amino acid metabolism disorder in sepsis. J Transl Med. 20:1232022. View Article : Google Scholar : PubMed/NCBI | |
|
Yan M, Xiao LY, Gosau M, Friedrich RE, Smeets R, Fu LL, Feng HC and Burg S: The causal association between COVID-19 and herpes simplex virus: A Mendelian randomization study. Front Immunol. 14:12812922023. View Article : Google Scholar : PubMed/NCBI | |
|
Bourgin M, Durand S and Kroemer G: Diagnostic, prognostic and mechanistic biomarkers of COVID-19 identified by mass spectrometric metabolomics. Metabolites. 13:3422023. View Article : Google Scholar : PubMed/NCBI | |
|
Zhang Y, Guo R, Kim SH, Shah H, Zhang S, Liang JH, Fang Y, Gentili M, Leary CNO, Elledge SJ, et al: SARS-CoV-2 hijacks folate and one-carbon metabolism for viral replication. Nat Commun. 12:16762021. View Article : Google Scholar : PubMed/NCBI | |
|
Ukolova IV and Borovskii GB: OXPHOS organization and activity in mitochondria of plants with different life strategies. Int J Mol Sci. 24:152292023. View Article : Google Scholar : PubMed/NCBI | |
|
Purandare N, Ghosalkar E, Grossman LI and Aras S: Mitochondrial oxidative phosphorylation in viral infections. Viruses. 15:23802023. View Article : Google Scholar : PubMed/NCBI | |
|
Schank M, Zhao J, Moorman JP and Yao ZQ: The Impact of HIV- and ART-induced mitochondrial dysfunction in cellular senescence and aging. Cells. 10:1742021. View Article : Google Scholar : PubMed/NCBI | |
|
Cortelli P, Mandrioli J, Zeviani M, Lodi R, Prata C, Pecorari M, Orlando G and Guaraldi G: Mitochondrial complex III deficiency in a case of HCV related noninflammatory myopathy. J Neurol. 254:1450–1452. 2007. View Article : Google Scholar : PubMed/NCBI | |
|
Guarnieri JW, Dybas JM, Fazelinia H, Kim MS, Frere J, Zhang Y, Soto Albrecht Y, Murdock DG, Angelin A, Singh LN, et al: Core mitochondrial genes are down-regulated during SARS-CoV-2 infection of rodent and human hosts. Sci Transl Med. 15:eabq15332023. View Article : Google Scholar : PubMed/NCBI | |
|
Senthilazhagan K, Sakthimani S, Kallanja D and Venkataraman S: SARS-CoV-2: Analysis of the effects of mutations in non-structural proteins. Arch Virol. 168:1862023. View Article : Google Scholar : PubMed/NCBI | |
|
Sahu U, Mullarkey MP, Murphy SA, Anderson JC, Putluri V, Kamal AHM, Park JH, Lee TJ, Ling AL, Kaipparettu BA, et al: IDH status dictates oHSV mediated metabolic reprogramming affecting anti-tumor immunity. Nat Commun. 16:38742025. View Article : Google Scholar : PubMed/NCBI | |
|
Greene TT, Jo Y, Chiale C, Macal M, Fang Z, Khatri FS, Codrington AL, Kazane KR, Akbulut E, Swaminathan S, et al: Metabolic deficiencies underlie reduced plasmacytoid dendritic cell IFN-I production following viral infection. Nat Commun. 16:14602025. View Article : Google Scholar : PubMed/NCBI | |
|
Lee AG, Scott JM, Fabbrizi MR, Jiang X, Sojka DK, Miller MJ, Baldridge MT, Yokoyama WM and Shin H: T cell response kinetics determines neuroinfection outcomes during murine HSV infection. JCI Insight. 5:e1342582020. View Article : Google Scholar : PubMed/NCBI | |
|
Cairns TM, Ditto NT, Lou H, Brooks BD, Atanasiu D, Eisenberg RJ and Cohen GH: Global sensing of the antigenic structure of herpes simplex virus gD using high-throughput array-based SPR imaging. PLoS Pathog. 13:e10064302017. View Article : Google Scholar : PubMed/NCBI | |
|
Rathbun MM and Szpara ML: A holistic perspective on herpes simplex virus (HSV) ecology and evolution. Adv Virus Res. 110:27–57. 2021. View Article : Google Scholar : PubMed/NCBI |