key: cord-0961308-5r4jnnut authors: Tiwari, Deeksha; Jakhmola, Shweta; Pathak, Devesh K.; Kumar, Rajesh; Jha, Hem Chandra title: Temporal In Vitro Raman Spectroscopy for Monitoring Replication Kinetics of Epstein–Barr Virus Infection in Glial Cells date: 2020-11-04 journal: ACS Omega DOI: 10.1021/acsomega.0c04525 sha: c5f743139ba560c90d1090996292007d1db3b034 doc_id: 961308 cord_uid: 5r4jnnut [Image: see text] Raman spectroscopy can be used as a tool to study virus entry and pathogen-driven manipulation of the host efficiently. To date, Epstein–Barr virus (EBV) entry and altered biochemistry of the glial cell upon infection are elusive. In this study, we detected biomolecular changes in human glial cells, namely, HMC-3 (microglia) and U-87 MG (astrocytes), at two variable cellular locations (nucleus and periphery) by Raman spectroscopy post-EBV infection at different time points. Two possible phenomena, one attributed to the response of the cell to viral attachment and invasion and the other involved in duplication of the virus followed by egress from the host cell, are investigated. These changes corresponded to unique Raman spectra associated with specific biomolecules in the infected and the uninfected cells. The Raman signals from the nucleus and periphery of the cell also varied, indicating differential biochemistry and signaling processes involved in infection progression at these locations. Molecules such as cholesterol, glucose, hyaluronan, phenylalanine, phosphoinositide, etc. are associated with the alterations in the cellular biochemical homeostasis. These molecules are mainly responsible for cellular processes such as lipid transport, cell proliferation, differentiation, and apoptosis in the cells. Raman signatures of these molecules at distinct time points of infection indicated their periodic involvement, depending on the stage of virus infection. Therefore, it is possible to discern the details of variability in EBV infection progression in glial cells at the biomolecular level using time-dependent in vitro Raman scattering. Raman spectroscopy (RS) is a sensitive enough tool to capture the characteristic bond vibrations of molecules and elucidate minute biochemical changes occurring in complex biological systems such as cells, tissue, and other body fluids. 1 Notably, any alteration in the biochemical homeostasis of the body may inevitably be associated with a disease state, mark disease initiation, or represent the aftermath of an illness. 2 For instance, biochemical changes due to manipulation in the carbohydrate metabolism may cause various pathological conditions such as galactosemia, diabetes, diabetic ketoacidosis, hyperglycemia, hereditary fructose intolerance, and glycogen storage disorders. 3 Besides, conditions like phenylketonuria, tyrosinemia, maple syrup urine syndrome, etc. appear due to alterations in the protein expression. 4 Additionally, alterations in lipid metabolism may lead to conditions such as familial hypercholesterolemia, hypertriglyceridemia, and low high-density lipoprotein (HDL). 5 RS is used to analyze human serum samples for various biochemical components such as glucose, cholesterol, lipids, 6 phenylalanine, 7 etc. for the detection of respective metabolic disorders mentioned above. The use of RS has been so far extended as a diabetes management tool to determine the blood glucose level subcutaneously. 8 Thus, the technique presents an immense potential in the field of disease diagnosis. Also, it is widely utilized to understand cancer, infection, and inflammation in the human body. 1 RS can differentiate between invasive and noninvasive breast cancer based on Raman spectral markers. 9 It is also employed to detect inflammation in the colon of patients suffering from inflammatory bowel disease (IBD), 10 in in vitro Tumor necrosis factor or TNF-α (pro-inflammatory cytokine)-treated endothelial cells, 11 and in in vivo experiments on tympanic membrane of a murine model. 12 Signature Raman spectra arising from lipids, collagen, or DNA content of the respective samples identified the disease-associated alterations. Infections with similar clinical symptoms like dengue and malaria can easily be distinguished using RS based on differentially regulated metabolites. 13 Moreover, the technique is used to study various virusassociated modalities of Kaposi's sarcoma-associated herpesvirus (KSHV) infection in BCBL-1, BC-1, and BJAB (B-cell lineage) 14 and rotavirus 15 and human papillomavirus (HPV) infection in primary human keratinocytes (PHKs) and CaSki cells. 9 In the last few decades, the prevalence of ubiquitous herpesviruses has accounted for a major disease burden in the human population worldwide. 16 Human herpesvirus-4 (HHV-4) also known as Epstein−Barr virus (EBV) is an important member of the Herpesviridae family. It infects about 90% of the world population. 17 Primary infection during childhood largely remains asymptomatic; however, infection in adults could cause infectious mononucleosis (IM). 18 Moreover, reactivation of the virus later in life after the initial exposure has been linked to serious conditions such as IM-like disease, hemophagocytic syndrome, chronic active EBV infection, and lymphomas. 19 It is also a recognized oncogenic virus known to infect lymphocytes and cause B-cell lymphomas (Burkitt's and Hodgkin's lymphoma) or lymphoproliferative disorders. EBV can infect epithelial cells resulting in nasopharyngeal carcinoma or EBV-associated gastric cancer. 20 Interestingly, the presence of EBV transcripts and DNA in the cerebrospinal fluid (CSF) of patients with neurodegenerative disorders such as Alzheimer's disease (AD), 21 Parkinson's disease (PD), 22 multiple sclerosis (MS), 23 etc. provides clues toward the possibility of EBV infecting the brain cells (neurons and glial cells). In general, to successfully infect any cell a virus must first attach to its surface and then penetrate through the outer cell membrane to insert its genomic content inside the cell. The entry of the virus inside a cell essentially depends upon the interaction between the virus surface proteins and the host cell receptors. It, therefore, limits the virus infectivity to host cells possessing the receptor and thereby defines its cell tropism. 24 Nonetheless, several recent studies have shown the plausibility of viruses such as SARS-CoV2 25 and Zika 26 virus to infect cells of multiple origins, i.e., exhibiting the property of multitropism. Studies demonstrated that EBV shows the property of multitropism by infecting B-cells, epithelial cells, and possibly neural cells as well. 27 EBV infects B-cells with the help of interaction between gp350, a glycoprotein present on its surface, and CD-21 surface marker expressed on the host cells. 28 However, EBV attaches and enters inside the epithelial cells in a CD-21-independent manner. The predicted cell receptors that may facilitate EBV entry inside the epithelial cells via gHgL (a viral glycoprotein) include integrin and ephrin molecules. 29 Fascinatingly, Jha et al. for the first time characterized in vitro EBV infection in a neuroblastoma cell line (ShSy-5y), teratocarcinoma neurons (Ntera2), and primary human fetal neurons. 29 Further, we successfully demonstrated EBV infection in glial cells, namely, HMC-3 (microglia) and U-87 MG (astroglia). Moreover, the virus may take advantage of diverse pathways in cells of different origins to gain entry and carry out its replication. 30 The type of genetic material (RNA or DNA) of the virus decides further processing, which is accompanied by major alterations in the biochemistry of the host cell. Briefly, these processes involve viral genome replication, transcription, and translation. This disparity in biomolecular pathways adopted by the virus in different cells may alter the kinetics of infection, 31 the duration of time elapsed from attachment to gaining entry, and further carrying out of the replication. 32 For instance, in B-cells, postinfection transcriptional changes are detected within the first 24 h preceding metabolic and phenotypic changes. EBV-initiated transcriptional reprogramming in B-cells is broadly categorized under three stages: RNA synthesis, manipulation of metabolic pathways, followed by cell division that occurs subsequently on the second, third, and fourth days post infection (dpi), respectively. 33 In contrast, de novo (cell-in-cell) EBV infection in epithelial cells is initiated as early as 4 h post infection (hpi), and activation of EBV can be traced by 6 hpi. In SHSy-5y and Ntera2 cells, the infection . The Raman spectra were acquired in the wavenumber range from 400 to 3500 cm −1 for HMC-3 and from 400 to 4000 cm −1 for U-87 MG cells. A 633 nm laser excitation source was used to record the data. could be detected by 1 dpi and proceed to the lytic cycle by 9 dpi. 29 Therefore, the same virus may take different time durations to establish a successful infection in cells of different origins. Therefore, we investigated the biomolecular changes in the glial cells upon EBV infection at different time intervals using RS. We intended to identify the unique Raman signals originating from the EBV invasion and infection in the glial cells, in particular astrocytes and microglia. Also, in our study, we observed differential regulation of various biochemical molecules in the cell body (periphery) and the nucleus. These modulations in the biomolecules also led to alterations in the signaling pathways associated with them. The temporal (infection progression with time) and spatial (infection condition at different locations on the cell) Raman analysis provides a consolidated study on the dynamics of EBV infection. ■ RESULTS Raman Spectra. We observed different peaks for HMC-3 and U-87 MG, pre-and postinfection, on plotting the raw data points (wavenumber vs intensity) of the Raman spectra using Origin-2018b software at various time durations (2, 4, 6, 12, 24, 36 For ease of further processing and validating the number of major peaks, the dimensionality of the raw data was reduced using principal component analysis (PCA). We obtained probable solutions, i.e., the principal components in our data in the range of 3−8, thus validating the number of peaks observed in the graph. Biomolecules' Peak Identification in Glial Cells. The distinct peaks that we obtained from the graph correspond to the signature spectra of unique biomolecules (Table 1) . On comparing the Raman signals of these molecules in the infected samples with those of the uninfected (UI) ones, we observed variations in vibrational intensity at different time points. The positive variation, i.e., upregulation of intensity above the basal level of uninfected cells, represented an increase in the number of molecules probably due to their enhanced anabolic activity. Also, a decline of signal intensity of the molecule below the basal levels depicted a decrease in the number of molecules due to their consumption/catabolism. Specifically, upon EBV infection in the microglial cells, we observed the highest activity of DNA and glycogen (481−495 cm −1 ) molecules at 2 and 12 hpi in the nucleus (Figure 2A The amounts of proline, hydroxyproline, glycogen, and lactic acid molecules (918−930 cm −1 ) 48,49 in the astroglial nucleus were elevated at 4, 24, and 48 hpi ( Figure 2B -1(I)). However, the reserves of these molecules diminished at the periphery at 2 hpi with subsequent restoration at 6 and 36 hpi ( Figure 2B -1(II)). As compared to the uninfected samples, the amounts of phenylalanine, glucose, saccharides, and carbohydrates (1096− 1117 cm −1 ) 36,42,46 were not altered considerably in the nucleus at 2 hpi. However, in the peripheral region, consumption of phenylalanine and saccharide moieties was maximum at 2 hpi, followed by subsequent replenishment at 6 hpi ( Figure 2B -2(II)). The Raman signals generated due to the polynucleotide chain and nucleic acids (1334−1342 cm −1 ) 34,48,45,51 were reduced at 2, 6, and 36 hpi in the nucleus, depicting marked catabolic activity, followed by an intermittent anabolic process at 24 and 48 hpi, which restored the pool ( Figure 2B -3(I)). At the periphery, maximum consumption of the polynucleotide chain and nucleic acids was observed at 2 and 24 hpi with a brief replenishment at 6 and 36 hpi ( Figure 2B For HMC-3 cells, in the interval of 0−2 hpi, we observed an increase in the signal intensities of glucose and lipids ( Figure 3A ) in the nucleus, whereas at the cell periphery, an increase in the intensity of PIP, DNA, and its phosphodiester backbone was obtained ( Figure 3B ). In the time span of 4−6 hpi, the amount of polysaccharide was increased at the cell periphery ( Figure 3B ). Later during 6−12 hpi, the nuclei of microglial cells showed maximum activity with increased levels of DNA and its phosphodiester backbone, glycogen, nucleotides (guanine and cytosine), and amino acids such as proline and tyrosine ( Figure 3A) . However, the peripheral region maintained its status quo without any changes ( Figure 3B ). However, during the last leg of our experimental infection from 24 to 36 hpi, the activity of amide III, nucleotides of DNA (guanine, cytosine, adenine, and thymine), and fatty acids was highest in the nucleus ( Figure 3A ). On the contrary, the amount of cholesterol and polysaccharides was highest in the peripheral region ( Figure 3B ). During the early phase of infection, i.e., from 0 to 2 hpi, purines of DNA and tryptophan showed a considerable increase in the nucleus of U-87 MG ( Figure 3C ). The cell periphery marked an increase in the levels of PIP, cholesterol, lipids, and protein stretch during the same time frame ( Figure 3D ). As the infection progressed from 2 to 4 hpi, the Raman intensities corresponding to lactic acid, proline, and RNA were considerably altered in the nucleus ( Figure 3C ). On the contrary, at the periphery, the amounts of proline molecule and amide III group were enhanced ( Figure 3D ). During the time interval of 4−6 hpi, a variation was accounted for the Raman intensity associated with PIP, cholesterol, polysaccharides, DNA, and glycogen molecules ( Figure 3C ). In the same time span at the periphery, a change was observed in the intensity corresponding to phenylalanine, vibrations due to OH−NH−CH bond stretching, and lipids ( Figure 3D ). Later in the time interval of 6−12 hpi, we documented a change in the levels of lipid-and protein-associated moieties, DNA, and glycogen molecules in the nucleus ( Figure 3C ). At the periphery in the same time interval, the biomolecular activity remained unchanged ( Figure 3D ). Lastly, from 36 to 48 hpi, the nuclear Raman intensity demonstrated an increase in the levels of PIP and cholesterol ( Figure 3C ). Changes due to protein stretch and OH−NH−CH stretching were observed at 36−48 hpi at the cell periphery ( Figure 3D ). Pathway Analysis of Selected Biomolecules. Further, the information regarding the biomolecules was uploaded to Qiagen's IPA system. The global molecular network available in the Ingenuity pathway knowledge base (IPKB) identified canonical pathways and gene networks associated with particular biomolecule-related neuropathologies. We obtained a connectome with the set filters of virus infection and associated diseases including neurodegeneration, suggesting high involvement of cholesterol in various pathways. Prominent molecules obtained on infection of microglia (HMC-3) were cholesterol, cholesterol esters, PIP, 3-nitrotyrosine, lactic acid, lipids, and glucose ( Figure 4A) . Notably, cholesterol trafficking is prominently affected in AD and MS pathologies via ABCA-1 (ATP binding cassette subfamily A member 1) transporter and modulation of apolipoprotein (APOA and APOE) metabolism. 58 Yet another group of molecules involved in cholesterol metabolism and implied in MS, 59 AD, and PD pathologies is that involved in membrane trafficking. These molecules, sterol-O-acyltransferase-1 (SOAT-1), sphingosine-1-phosphate lyase-1 (SGPL-1), and synuclein α (SNCA), are involved in cholesterol transport in and out of the cell and in presynaptic signaling. The connectome indicated various other membrane proteins such as matrix-associated protein-2 (MAP-2), CD44 (a cell surface marker), proteolipid protein-1 (PLP-1), and amyloid precursor protein (APP) directly or indirectly related to cholesterol metabolism in development of neurodegenerative pathologies. These molecules mainly act as cell surface receptors and are responsible for maintaining cell integrity, cell−cell interactions, adhesion, and migration. APP, PLP-1, and MAP-2 are widely recognized in AD, whereas MBP (myelin basic protein) linked with lipid metabolism is a primary marker for neurodegeneration observed in MS and ALS. Cell damage caused by oxidative stress generated due to reactive oxygen species (ROS) is one of the prevailing hypotheses of neurodegenerative pathology. Being rich in peroxidation susceptible substrates and having high oxygen consumption, the brain is more prone to damage caused by ROS. Molecules involved in the oxidative stress hypothesis of neurodegeneration were also interlinked with the ones we found in our study. For example, PSEN-1 (presenilin-1), which is thoroughly studied in MS, 60 AD, and PD pathologies, is the connecting link among PIP, 3nitrotyrosine, lactic acid, cholesterol ester, and cholesterol. Additionally, molecules involved in the oxidative stress hypothesis such as BACE-1 (β site APP cleaving enzyme-1) and AβPP also interlink with the cholesterol metabolism. In the case of astrocytes, we found alterations in pathways implied in sterol biosynthesis, lipid transport, microtubule cytoskeleton constitution, and various endoplasmic reticulum (ER) proteins, along with the molecules involved in transcription regulation, metal ion binding, and various kinases ( Figure 4B ). Cholesterol was found to be related to most of the molecules compared to any other biomolecule under investigation. It is known that cholesterol metabolism is affected via modulation of the enzymes involved in sterol biosynthesis, such as sterol-C5-desaturase (SC5D), 24dehydrocholesterol reductase (DHCR24), squalene monooxygenase (SQLE), and lecithin-cholesterol acyltransferase (LCAT). Dysregulation of sterol biosynthesis is strongly implied in neurodegenerative pathology like AD. 61 Other molecules concerned with AD and MS pathologies are also affected in astroglia on EBV infection such as those involved in lipid transport pathway (involving ABCA-1, APOE, and APOA1) 62−64 and microtubule cytoskeleton proteins (PLP-1, MAP-2, and APP), 65, 66 in addition to PMP-22 (peripheral myelin protein-22) 67 and other membrane proteins (matrix metalo-endopeptidases or MME, BACE-1, and sonic hedgehog or SHH). 68−70 Some other molecules including glutamate ionotropic receptor NMDA type subunit 1 (GRIN1) 71 and sodium voltage-gated channel α subunit 11 (SCN11A) 72 were also reported in this regard. In addition, certain kinases such as protein kinase C ζ (PRKCZ), 73 erb-b2 receptor tyrosine kinase 2 (ERBB2), 74 and neurotrophic receptor tyrosine kinase 2 (NTRK2), 75 also associated to cholesterol metabolism, were observed in astrocytes upon EBV infection. Interestingly, these molecules are reported to be associated with AD, PD, and other neurodegenerative pathologies. Cholesterol metabolism pathways associated with transcription regulator factors such as cAMP-responsive element-binding protein 1 (CREB1), cAMP response element mediator (CREM), MAF (transcription factor), and methyl CpG binding protein (MECP) contemplated in our study were also implied in Huntington's disease (HD) 76 and AD, 76, 77 respectively. Apart from cholesterol, hyaluronic acid (HA) is another molecule modulated on EBV infection. Nonetheless, glycosaminoglycan-binding proteins such as BCAN (brevican core protein), 78 CD44 (CD44 antigen), 79 and HAPLN4 (hyaluronan and proteoglycan link protein 4) 80 related to the HA metabolism are involved in PD pathology. Various other molecules related to HA metabolism such as insulin-like growth factor (IGF1), kinases protein tyrosine kinase 2 (PTK2), matrix metalo-proteinases (MMP14, MMP9), and hyaluronan synthase 3 (HAS3) could be responsible for altered HA metabolism on EBV infection ultimately leading to neurodegeneration. The presence of EBV has been prevalently reported in CSF samples and postmortem brain tissue of patients suffering from neurodegenerative disorders, such as MS, 81 AD, 82 PD, 83 ALS, 81, 84 etc., although how the virus gains entry and propagates inside the cells of the host brain is yet to be elucidated at the biomolecular level. Traditional biomolecular techniques used to detect the virus infection in any cell utilize the expression of viral transcript or proteins in the host. 85 Detection of infection, thereby, becomes organ-and individual-specific, costly, time-consuming, and handlingdependent, along with a low reproducibility rate. RS on the other hand can trace the infection process with minute biomolecular changes occurring in the host cell on virus infection noninvasively and more robustly in lesser time and is also independent of handling constraints. 86 This advantage gives an upper edge to the technique for the early detection of the virus during the infection process. As discussed previously, a virus infection cycle in a mammalian cell follows the steps of attachment, penetration, uncoating, genomic replication, assembly, and finally viral egress. All of these processes are biochemically driven and can be related to changes in the signature Raman spectrum. 87 We observed sequential biomolecular changes occurring in the astrocytes and microglial cell lines post-EBV infection using RS. The information thus obtained provided an insight into the probable virus control and the host cell modulations. Our data suggested differences in the infection pattern of the virus in the glial cell repertoire. Raman signals obtained during initial phases of infection, i.e., in the time duration between causing EBV infection to a healthy cell and 2 hpi, showed increased levels of glucose and lipid molecules in the nucleus and enhanced DNA and PIP activity at the periphery of the microglial cells, whereas Raman bands relating to PIP, lipids (cholesterol and cholesterol esters), and proteins were observed at the periphery of astroglial cells. It is suggested that lipids might be involved in the process of cell membrane modulation to facilitate EBV entry in the cells. In addition, until the elapse of 2 hpi, EBV (a DNA virus) was present at the cell periphery of the glial cells, and alterations in the signaling processes involving PIP were ongoing, implying that signaling molecules such as PIP might help in attachment and entry of EBV inside the glial cells. Many viruses have developed mechanisms to take advantage of phosphatidylinositol (PI)-mediated signaling cascade to gain entry and trigger modulations in the cell microenvironment beforehand to favor the virus's survival. 88 Although PI is the least abundant phospholipid in the cell membrane, it plays a crucial role in modulating the traffic in and out of the cell. It does so by getting differentially phosphorylated and giving rise to various PIP species. 89 Class-I PI-3 kinase (PI-3K) is the most extensively exploited signaling pathway by various viruses during the entry process. For example, HIV-1 is known to employ the PI-3K-mediated micropinocytosis mechanism to modulate the cell cytoskeleton for gaining entry into the cell. 90 Various other studies have also reported the alteration of pathways involving PIP in the successful establishment of virus infections of KSHV, human herpesvirus-8 (HHV-8), 91 HSV-1, 88 HCV, 92 Zaire Ebola virus, 93 and VZV. 94 Thus, our experiments suggested its role in EBV's entry inside the glial cells. Interestingly, PIP is implied in the processing of APP as well and thereby in the development of AD pathologies such as plaque formation or neurofibrillary tangle (NFT) deposition. 95 It also suggested a probable PIP-mediated role of EBV in neurodegeneration. Our experiments also indicated that EBV infection manipulates sterol metabolism in astroglial cells on entry. Interestingly, the brain is the most cholesterol-rich organ, consisting of about 20−25% of the total cholesterol content of the whole body. Sterol metabolism naturally being one of the cornerstones of the energy cycle in the brain plays an important role in numerous neural metabolic pathways. Disruption of sterol homeostasis in the brain may be linked with multiple neurodegenerative pathologies such as MS, AD, PD, HD, and ALS. 96−98 Any disturbance in the enzymatic mechanism of cholesterol synthesis, trafficking, or dissolution would lead to abnormal deposition of various sterol moieties in the intracellular or extracellular matrix. This might cause hindrance in impulse transmission through neurons, leading to atypical pathology of neurodegenerative diseases as AD, 99 PD, or Lewy body dementia. 100 The capacity of EBV to modulate sterol homeostasis in astroglia is observed in our study, and therefore it is suggested that EBV could also mediate neurodegeneration by dysregulation of the cholesterol metabolism. By the end of 2 hpi, the Raman signals obtained from the periphery of microglial cells coincide with those obtained at the nucleus. In addition to the continued glucose and lipid metabolism signals from the nucleus, we observed an increase in the expression of PIP and DNA at both locations. The nuclear signals decline slightly thereafter till 4 hpi. These observations point toward an early start of an increase in the DNA activity inside the nucleus of microglial cells by 2 hpi, accompanied by an enhancement in signaling processes throughout the cell, as indicated by the increased signals for PIP or lipid molecules such as cholesterol. Moreover, by the end of 4 hpi, signals from the periphery showed the presence of polysaccharide, protein-related moieties, and purines of nucleic acids. Most biomolecular signals from the nucleus showed a decline in the period of 4−6 hpi, except for a slight increase in the expression of fatty acids at 6 hpi. However, we noticed a probable increase in cellular traffic at the periphery during 4−6 hpi based on the increased activity of cholesterol, PIP, and amino acids (Phe, Tyr, and Trp) at the periphery of microglia. Contrastingly, we recorded a high activity of astroglial nucleus during 4−6 hpi. We surmise that by the end of 4 hpi the virus must have gained entry into the astroglial cell nucleus. Later, upon reaching the nucleus, it probably utilized cholesterol, polysaccharides, and lipid metabolism to carry out its replication. Besides, few reports suggest the exploitation of cholesterol and saccharide moieties to attach and gain entry into the host cell by the herpesvirus family. 101 We speculated that after entering the nucleus of a microglial cell, during 6−12 hpi, the virus most likely manipulates nuclear metabolism of glycogen 102 and amino acids 103 to facilitate its replication cycle. It is noteworthy to state that abnormal glucose metabolism is often associated with MS disease pathology and various neuro-inflammatory disorders. 104, 105 The microglial nucleus showed maximum activity post 6 hpi till 12 hpi. Enhanced signals for amino acids (Pro and Tyr), glycogen, and DNA metabolism (in the form of signals for DNA, phosphodiester bond, and purines-guanine and cytosine) are observed past 6 h in the nucleus of microglial cells and are maintained till the end of 12 hpi. This coincided with the presence of polysaccharides and amino acids in the nucleus. Nonetheless, in microglia, the activity of most biomolecules at the periphery was downregulated from 6 to 12 hpi, except a slight increase in polysaccharides and amino acids (Pro and Val) signals, whereas in the astroglial cells at 6− 12 hpi, further processing of the virus inside the nucleus is speculated, as implied by the expression of various molecules such as triglycerides, fatty acids, lipids, and proteins. Various viruses like dengue (DENV), 106 human cytomegalovirus (HCMV), 107 herpes simplex virus-1 (HSV-1), 108 and EBV 109 are known to manipulate the glycolytic pathway for their benefit. These viruses could either utilize glycogen for glycogenolysis to sustain increased energy requirements during replication or fuel fatty acid synthesis (HCMV) or enhance pyrimidine production (HSV-1) as required. 110 The manipulation of glycogen metabolism by various viruses to facilitate their replication cycle enabled us to conclude that EBV may modulate glycogen metabolism for aiding its replication cycle in the microglia occurring at 6−12 hpi. Importantly, Pro and Tyr are involved in cell signaling pathways such as those regulating proliferation 111 and ROS scavenging. 112 Modulation of amino acid metabolism similar to that of Tyr and Phe is known to be associated with HIV-1 113 and HBV infections. 114 Then, in the duration of 12−24 hpi, we hypothesized that successive steps of the virus packaging and transport occurred in the astrocytes indicated by the presence of nucleotide molecules at the periphery. However, the virus replication cycle repeated after completion of 24 hpi, as suggested by the repetition of signals obtained for PIP and cholesterol from the nucleus, whereas signals collected from the periphery indicate the presence of nucleotides and proteins, which might denote the viral egress from the cell at later time points (36−48 hpi) . Thereafter, in the microglial cell line, plausibly, the process of viral packaging is initiated (periphery) at 24 hpi and is carried out until 36 hpi. Enhancements in signals produced from amide bond and amino acids (Phe, Tyr, and Trp), nucleotides, and fatty acids corroborate our speculations, as the molecules falling in the broad category of protein and lipids are believed to be utilized by certain viruses such as HCMV to facilitate their survival inside the host. In fact, a wide variety of viruses, such as HCMV, HSV, EBV, DENV, hepatitis B virus (HBV), hepatitis-C virus (HCV), vaccinia virus, and KSHV to name a few, reportedly alter fatty acid metabolism. 114, 115 Lipids and fatty acids being the second preferable energy source after glucose for the brain play a major role in neurophysiology. Thus, any disturbance in the homeostasis of the lipid metabolism in the brain could result in grave consequences. For instance, impaired enzymatic cascades (such as sphingolipid pathway) or oxidative stress resulting in dysregulation of lipid metabolism and lipid rafts are well described to be associated with amyloid plaque and NFT deposition causing AD, 116 PD, or HD. 97 Notably, various lipid molecules are enhanced in the brain of MS patients, thus giving rise to a specific lipidomic profile. 117 Also, a perturbed cholesterol pathway is associated with MS. 96 After 36 hpi, we suspect that many cellular processes simultaneously occur as the Raman signals thus obtained are mixed (pertaining to the obtained signals corresponding to molecules such as phosphodiester, Pro, Tyr, and lipids). These processes may occur to facilitate the initiation of the next virus replication cycle and/or carry on the ongoing cycle. Time-dependent in vitro spatial Raman spectroscopy carried out on different regions of glial cells shows the temporal evolution of EBV infection, enabling one to understand the virus influencing mechanism. In brief, the investigation directed us to believe that EBV enters the glial cells probably in the first 2 hpi by utilizing PIP-dependent signaling pathways, whereas further processing of the virus differs slightly among microglial and astroglial cells. To reach inside and manipulate the nuclear microenvironment of microglial cells, it takes up to 6 hpi and up to 4 hpi in the astroglial cells. Moreover, during its nuclear hijack process from 6 to 12 hpi, the virus manipulates glycogen and amino acid metabolism in the microglial cells. While in the case of the astroglial cells, fatty acids and triglyceride metabolism is affected at 6−12 hpi. Later, during 12−24 hpi, the processes of replication and cellular transport are still carried on in the microglia, whereas EBV showed signs of viral packaging and transport being initiated in astroglial cells. It is only after 24 hpi that viral packaging and egress is initiated in microglial cells. However, in astroglia, from 24 hpi onward we noticed overlapping signals for replication and viral egress. Thus, the study aided us to further our understanding of the involvement of different biomolecules at various stages of EBV infection progression in two different glial cells. With further advances in technology in the future, the application of RS could extend to differentiating the viral infection stages in clinical settings and help in noninvasive and early disease diagnosis. The recent outbreak of SARS-CoV2 has brought forth the importance of such rapid diagnostic tools in the detection of virus infection. 119 The temporal and spatial Raman spectroscopic technique appears to be a step forward toward understanding the viral biology after infection in host cells and also assisting in a comparative analysis of replication kinetics in different cells on infection with multitropic viruses such as EBV. Cell Culture. HMC-3 (SV-40 transformed, immortalized microglial cell lines were obtained from Dr. Anirban Basu's laboratory, National Brain Research Centre, Delhi) and U-87 MG cell lines (glioblastoma cells of epithelial origin from the human brain were purchased from the National Centre for Cell Sciences, Pune) were cultured in high-glucose-containing Dulbecco's modified Eagle's medium (HiMedia) supplemented with 10% fetal bovine serum (Invitrogen), 50 units/ mL penicillin, and 100 μg/mL streptomycin (Invitrogen) over coverslips. The cells were incubated at 37°C with 5% CO 2 . Virus Isolation and Purification. HEK-293T cells (a kind gift from Prof. Erle S. Robertson's laboratory, University of Pennsylvania) transformed with GFP-tagged virus-containing bacmid were used to obtain EBV particles. 118 Briefly, to obtain the virus particles, lytic induction was given to cells with 20 ng/mL tetradecanoyl phorbol acetate and 3 mM butyric acid at 60% cell confluence for 4−5 days. The cell suspension was centrifuged at 775g for 20 min, and the supernatant was then filtered through a 0.45 μm membrane (Millipore). The filtrate was ultracentrifuged at 65 291g for 90 min at 4°C. The concentrated virus pellet was resuspended in a suitable amount of culture media and stored at −80°C until further use. EBV Infection in Glial Cells. An appropriate concentration of EBV for each cell line was investigated. The U-87 MG cells (astrocytes) cultured onto the coverslips were infected with EBV at an MOI (multiplicity of infection) of 2.5 and incubated for the duration of 2, 4, 6, 12, 24, 36, and 48 h. Also, 5.0 MOI of EBV was used for infecting HMC-3 cells (microglia). The microglial cells were incubated with the virus for the duration of 2, 4, 6, 12, 24, and 36 h. After collecting the coverslips at the respective time points, the cells grown on these coverslips were fixed using 4% paraformaldehyde (PFA) for 20 min at room temperature. The coverslips were mounted onto glass slides before visualization on the Raman spectrometer. Raman Spectroscopy. Raman spectroscopy was performed on a LabRAM HR Evolution (Horiba-Jobin Yvon) spectrometer using a He-Ne laser (λ exc = 633 nm, ∼10 mW) excitation source. The sample was focused with the help of a microscope objective to probe the nucleus and the periphery of the cell. The Raman measurement was performed using minimum possible laser power controlled using an ND filter to avoid any laser-related damage to the cells. The Raman study was performed on uninfected (UI) and 2, 4, 6, 12, 24, 36, and 48 hpi of respective cells. The laser was focused onto the nuclei and cell body (periphery) separately as visualized on the microscope to acquire signals from respective places. Data Analysis. Graphical Analysis. All of the raw data of Raman vibrational intensity obtained from the Raman spectroscope within wavenumber ranges of 400−3500 cm −1 (HMC-3) and 400−4000 cm −1 (U-87 MG) was smoothened by 20 points for better visibility. Signals from common cell culture artifacts such as culture medium, phosphate-buffered saline (PBS), PFA, coverslips, and glass slides were subtracted from all of the samples. Thereafter, we selected the most relevant wavenumber peaks from the graph based on intensity. Statistical Analysis. The dimensionality of the raw data was reduced using principal component analysis (PCA) on SPSS software. The total numbers of selected peaks were verified using PCA. Mann−Whitney U test (α = 0.05) was applied to the screened data consisting of wavenumber ranges corresponding to the peaks for comparison of Raman intensities at subsequent time points. Additionally, we also included the odds ratio (OR) analysis to enhance data confidence. Interactome Study. The selected wavenumber peaks were cross-referenced with the available literature to verify the corresponding biomolecules. The biomolecules were then uploaded on Qiagen's Ingenuity Pathway Analysis (IPA) software to obtain an interconnection between them. Thereafter, a global molecular network available on the Ingenuity pathway knowledge base (IPKB) was used as a reference to perform analysis of canonical pathways, diseases, functions, and gene networks. The analysis gave us hits that are most significantly related to the biomolecular changes occurring in viral infection and neurodegenerative diseases. Challenges in application of Raman spectroscopy to biology and materials The biochemical basis of disease Post-Exercise Carbohydrate-Energy Replacement Attenuates Insulin Sensitivity and Glucose Tolerance the Following Morning in Healthy Adults Inherited Metabolic Disorders: Aspects of Chronic Nutrition Management The Treatment of Disorders of Lipid Metabolism Detecting alterations of glucose and lipid components in human serum by near-infrared Raman spectroscopy Surface-Enhanced Raman Scattering (SERS) for Detection of Phenylketonuria for Newborn Screening Direct observation of glucose fingerprint using in vivo Raman spectroscopy Early detection of cervical neoplasia by Raman spectroscopy Clinical characterization of in vivo inflammatory bowel disease with Raman spectroscopy Unsaturated lipid bodies as a hallmark of inflammation studied by Raman 2D and 3D microscopy Identification of early inflammatory changes in the tympanic membrane with Raman spectroscopy Rapid Discrimination of Malaria-and Dengue-Infected Patients Sera Using Raman Spectroscopy Spectroscopic analysis of Kaposi's sarcoma-associated herpesvirus infected cells by Raman tweezers Rapid and sensitive detection of rotavirus molecular signatures using surface enhanced Raman spectroscopy Herpesviruses: Harmonious Pathogens but Relevant Cofactors in Other Diseases? Front A reliable Epstein-Barr Virus classification based on phylogenomic and population analyses Clinical characteristics of primary and reactivated Epstein-Barr virus infection in children Virus: Diseases Linked to Infection and Transformation Herpes virus in Alzheimer's disease: relation to progression of the disease The association between infectious burden and Parkinson's disease: A case-control study EBV and MS: Major cause, minor contribution or red-herring? Cellular receptors for viruses: links to tropism and pathogenesis Epstein Barr virus entry; kissing and conjugation Identification of an epitope in the major envelope protein of Epstein-Barr virus that mediates viral binding to the B lymphocyte EBV receptor (CR2) Alternate replication in B cells and epithelial cells switches tropism of Epstein−Barr virus Comparative study of influenza virus replication in MDCK cells and in primary cells derived from adenoids and airway epithelium Comparative tropism, replication kinetics, and cell damage profiling of SARS-CoV-2 and SARS-CoV with implications for clinical manifestations, transmissibility, and laboratory studies of COVID-19: an observational study Epstein−Barr virus reprograms human B lymphocytes immediately in the prelatent phase of infection Nearinfrared Raman spectroscopy for the classification of epithelial precancers and cancers Near-Infrared Micro-Raman Spectroscopy for in Vitro Detection of Cervical Cancer Near infrared Raman spectra of human brain lipids Evaluation of Raman probe for oesophageal cancer diagnostics Characterization by Raman spectroscopy of conformational changes on guanine−cytosine and adenine−thymine oligonucleotides induced by aminooxy analogues of spermidine Raman spectroscopy: elucidation of biochemical changes in carcinogenesis of oesophagus Diagnosis of Basal Cell Carcinoma by Raman Spectroscopy Raman spectroscopy for optical diagnosis in the larynx: preliminary findings Structural evaluation of human and sheep bone and comparison with synthetic hydroxyapatite by FT-Raman spectroscopy Correlation between nearinfrared Raman spectroscopy and the histopathological analysis of atherosclerosis in human coronary arteries Near-infrared Raman spectroscopy for optical diagnosis of lung cancer Tissue Raman Spectroscopy for the Study of Radiation Damage: Brain Irradiation of Mice Effect of substrate choice and tissue type on tissue preparation for spectral histopathology by Raman microspectroscopy Raman spectroscopy for identification of epithelial cancers NIR-FT Raman and FT-IR spectral studies and ab initio calculations of the anti-cancer drug combretastatin-A4 Micro-Raman spectroscopy used to identify and grade human skin pilomatrixoma Raman spectroscopy of biological tissues Magnificent Milestones and Emerging Opportunities in Medical Engineering Prospects for in vivo Raman spectroscopy Understanding the molecular information contained in principal component analysis of vibrational spectra of biological systems Detection of Skin Cancer by Classification of Raman Spectra Micro-Raman spectroscopy detects individual neoplastic and normal hematopoietic cells Detection of meningioma in dura mater by Raman spectroscopy Deficiency of ABCA1 impairs apolipoprotein E metabolism in brain The first sphingosine 1-phosphate lyase inhibitors against multiple sclerosis: a successful drug discovery tale Presenilin1 regulates Th1 and Th17 effector responses but is not required for experimental autoimmune encephalomyelitis Importance of apolipoprotein AI in multiple sclerosis Opposing effects of Apoe/Apoa1 double deletion on amyloid-β pathology and cognitive performance in APP mice Involvement of Lipids in Alzheimer's Disease Pathology and Potential Therapies Cytoskeletal pathologies of Alzheimer disease PLP1 mutations in patients with multiple sclerosis: Identification of a new mutation and potential pathogenicity of the mutations Oligodendrocytes and Schwann Cells Cleavage of membraneassociated ICAM-1 from astrocytes: involvement of a metalloprotease Beta-Secretase Cleavage of Alzheimer's Amyloid Precursor Protein by the Transmembrane Aspartic Protease BACE Sonic hedgehog pathway activation increases mitochondrial abundance and activity in hippocampal neurons Progressive neuroanatomical changes caused by Grin1 loss-of-function mutation Sodium Channel Cleavage Is Associated with Aberrant Neuronal Activity and Cognitive Deficits in a Mouse Model of Alzheimer's Disease Atypical protein kinase C in neurodegenerative disease I: PKMζ aggregates with limbic neurofibrillary tangles and AMPA receptors in Alzheimer disease ErbB2 regulates autophagic flux to modulate the proteostasis of APP-CTFs in Alzheimer's disease Genetic Association of Neurotrophic Tyrosine Kinase Receptor Type 2 (NTRK2) With Alzheimer's Disease Disruption of CREB function in brain leads to neurodegeneration Methyl-CpG Binding Protein 2 in Alzheimer Dementia Glycoproteomics in neurodegenerative diseases Hyaluronan Synthesis, Catabolism, and Signaling in Neurodegenerative Diseases Elevated Hapln2 Expression Contributes to Protein Aggregation and Neurodegeneration in an Animal Model of Parkinson's Disease. Front Complete Epstein-Barr virus seropositivity in a large cohort of patients with early multiple sclerosis Human herpesvirus 4 and adaptive immunity in Alzheimer's disease Human serum antibodies against EBV latent membrane protein 1 cross-react with α-synuclein Epstein-Barr virus antibodies in serum and cerebrospinal fluid from Multiple sclerosis, Chronic Inflammatory Demyelinating Polyradiculoneuropathy and Amyotrophic Lateral Sclerosis Laboratory Diagnosis of Viral Infection Raman Spectroscopy as a New Biochemical Diagnostic Tool Raman spectroscopy detects biochemical changes due to proliferation in mammalian cell cultures Epidermal Growth Factor Receptor-PI3K Signaling Controls Cofilin Activity To Facilitate Herpes Simplex Virus 1 Entry into Neuronal Cells PIP2: choreographer of actin-adaptor proteins in the HIV-1 dance AKTivation of PI3K/AKT/mTOR signaling pathway by KSHV Roles for endocytic trafficking and phosphatidylinositol 4-kinase III alpha in hepatitis C virus replication Integrin 1 Mediates Vaccinia Virus Entry through Activation of PI3K/Akt Signaling Phosphatidylinositol-3-phosphate regulates sorting and processing of amyloid precursor protein through the endosomal system Highdensity lipoprotein cholesterol is associated with multiple sclerosis fatigue: A fatigue-metabolism nexus? Lipid Metabolism in Neurodegenerative Diseases Cholesterol Metabolism in the Brain and Its Association with Parkinson's Disease Cellular cholesterol homeostasis and Alzheimer's disease Dysregulated Lipid Metabolism and Its Role in α-Synucleinopathy in Parkinson's Disease Molecular requirement for sterols in herpes simplex virus entry and infectivity Rhinovirus induces an anabolic reprogramming in host cell metabolism essential for viral replication Glutaminolysis and Glycolysis Are Essential for Optimal Replication of Marek's Disease Virus Perturbed glucose metabolism: insights into multiple sclerosis pathogenesis Glycogen synthase kinase-3 (GSK3): inflammation, diseases, and therapeutics Dengue Virus Induces and Requires Glycolysis for Optimal Replication Dynamics of the Cellular Metabolome during Human Cytomegalovirus Infection Herpes simplex type 1 activates glycolysis through engagement of the enzyme 6-phosphofructo-1-kinase (PFK-1) Enhanced aerobic glycolysis of nasopharyngeal carcinoma cells by Epstein-Barr virus latent membrane protein 1 Divergent effects of human cytomegalovirus and herpes simplex virus-1 on cellular metabolism Oncogenic human herpesvirus hijacks proline metabolism for tumorigenesis Comparative metabolomics unveils molecular changes and metabolic networks of syringin against hepatitis B mice by untargeted mass spectrometry Disturbed Amino Acid Metabolism in HIV: Association with Neuropsychiatric Symptoms. Front Multiomics analyses reveal metabolic alterations regulated by hepatitis B virus core protein in hepatocellular carcinoma cells Chewing the Fat: The Conserved Ability of DNA Viruses to Hijack Cellular Lipid Metabolism Lipid Integration in Neurodegeneration: An Overview of Alzheimer's Disease Lipid profile of cerebrospinal fluid in multiple sclerosis patients: a potential tool for diagnosis Rapid Detection of COVID-19 Causative Virus (SARS-CoV-2) in Human Nasopharyngeal Swab Specimens Using Field-Effect Transistor-Based Biosensor Targeting Epstein−Barr virus oncoprotein LMP1-mediated glycolysis sensitizes nasopharyngeal carcinoma to radiation therapy A nuclear receptor involved in cholesterol metabolism regulates herpesvirus latency and reactivation Cholesterol is critical for Epstein-Barr virus latent membrane protein 2A trafficking and protein stability Viral MicroRNAs Repress the Cholesterol Pathway, and 25-Hydroxycholesterol Inhibits Infection Hepatitis B virus X protein regulates hepatic glucose homeostasis via activation of inducible nitric oxide synthase Conserved Tryptophan Motifs in the Large Tegument Protein pUL36 Are Required for Efficient Secondary Envelopment of Herpes Simplex Virus Capsids Role of indoleamine-2,3-dioxygenase in alpha/beta and gamma interferon-mediated antiviral effects against herpes simplex virus infections Inhibition of the phosphatidylinositol 3-kinase-Akt pathway enhances gamma-2 herpesvirus lytic replication and facilitates reactivation from latency Human cytomegalovirus infection induces adipocyte-like lipogenesis through activation of sterol regulatory element binding protein 1 Global Metabolic Profiling of Infection by an Oncogenic Virus: KSHV Induces and Requires Lipogenesis for Survival of Latent Infection Glutamine metabolism is essential for human cytomegalovirus infection Effect of infection on lipid profile: focus on Epstein−Barr virus Dengue virus nonstructural protein 3 redistributes fatty acid synthase to sites of viral replication and increases cellular fatty acid synthesis De novo Fatty Acid Biosynthesis Contributes Significantly to Establishment of a Bioenergetically Favorable Environment for Vaccinia Virus Infection Activation of ATP citrate lyase by mTOR signal induces disturbed lipid metabolism in hepatitis B virus pre-S2 mutant tumorigenesis Hepatitis C virus core protein inhibits microsomal triglyceride transfer protein activity and very low density lipoprotein secretion: a model of viral-related steatosis Author Contributions ∥ D.T. and S.J. contributed equally to this work. We appreciate our laboratory colleagues (from Infection-bioengineering group), in particular Omkar Indari, for insightful discussions and advice and Buddhadev Baral for his help in performing the experiments. We gratefully acknowledge the Indian Institute of Technology Indore for providing facilities and support.