Pipes Feed Preview: Feed not found & ScienceDirect Publication: The Spine Journal & ScienceDirect Publication: World Neurosurgery & Neurosurgery : nature.com subject feeds & jns - News & Surgical Neurology International & Feed not found & Feed not found & Feed not found & Feed not found & Feed not found & Feed not found & Feed not found & jns - News & Journal of Neurosurgery & Journal of Neurosurgery: Spine & Journal of Neurosurgery: Pediatrics & Neurosurgical Focus & Journal of Neurology, Neurosurgery & Psychiatry current issue & AANS Neurosurgeon & Neurosurgery & tandf: British Journal of Neurosurgery: Table of Contents & The New England Journal of Medicine: Search Results in Neurology/Neurosurgery

  1. The preclinical and clinical trials of mesenchymal stem cell’s secretome in traumatic brain injury: Review of basic science

    Fri, 13 Jun 2025 17:11:59 -0000

    The preclinical and clinical trials of mesenchymal stem cell’s secretome in traumatic brain injury: Review of basic science Category: Article Type: I Gde Anom Ananta Yudha1, Andi Asadul Islam2, Mochammad Hatta3, Firdaus Hamid3Doctoral Program of Medicine, Faculty of Medicine, Hasanuddin University, Sulawesi Selatan, IndonesiaDepartment of Neurosurgery, Universitas Hasanuddin, Sulawesi Selatan, IndonesiaDepartment of Microbiology, Faculty of … Continue reading The preclinical and clinical trials of mesenchymal stem cell’s secretome in traumatic brain injury: Review of basic science
    <div><!DOCTYPE html PUBLIC "-//W3C//DTD HTML 4.0 Transitional//EN" "http://www.w3.org/TR/REC-html40/loose.dtd"> <html><head><meta http-equiv="content-type" content="text/html; charset=utf-8"></head><body><div class="row"><div class="col-lg-9 col-sm-8 col-xs-12"><div class="media-body details-body"> <a href="https://surgicalneurologyint.com/?post_type=surgicalint_articles&p=13633"><h2 class="media-heading"><h2 class="media-heading">The preclinical and clinical trials of mesenchymal stem cell’s secretome in traumatic brain injury: Review of basic science</h2></h2></a> </div><div class="disp_categories"> <p><label>Category: </label><span></span></p> <p><label>Article Type: </label><span></span></p> </div><a href="mailto:anomanantayudha@gmail.com" target="_top">I Gde Anom Ananta Yudha</a><sup>1</sup>, <a href="mailto:andiasadul@yahoo.com" target="_top">Andi Asadul Islam</a><sup>2</sup>, <a href="mailto:hattaram@yahoo.com" target="_top">Mochammad Hatta</a><sup>3</sup>, <a href="mailto:firdaus.hamid@gmail.com" target="_top">Firdaus Hamid</a><sup>3</sup><ol class="smalllist"><li>Doctoral Program of Medicine, Faculty of Medicine, Hasanuddin University, Sulawesi Selatan, Indonesia</li><li>Department of Neurosurgery, Universitas Hasanuddin, Sulawesi Selatan, Indonesia</li><li>Department of Microbiology, Faculty of Medicine, Hasanuddin University, Makassar, Indonesia</li></ol><p><strong>Correspondence Address:</strong><br>I Gde Anom Ananta Yudha, Doctoral Program of Medicine, Faculty of Medicine, Hasanuddin University, Makassar, Indonesia.<br></p><p><strong>DOI:</strong>10.25259/SNI_1025_2024</p>Copyright: © 2025 Surgical Neurology International This is an open-access article distributed under the terms of the Creative Commons Attribution-Non Commercial-Share Alike 4.0 License, which allows others to remix, transform, and build upon the work non-commercially, as long as the author is credited and the new creations are licensed under the identical terms.<div class="parablock"><p><strong>How to cite this article: </strong>I Gde Anom Ananta Yudha1, Andi Asadul Islam2, Mochammad Hatta3, Firdaus Hamid3. The preclinical and clinical trials of mesenchymal stem cell’s secretome in traumatic brain injury: Review of basic science. 13-Jun-2025;16:235</p></div><div class="parablock"><p><strong>How to cite this URL: </strong>I Gde Anom Ananta Yudha1, Andi Asadul Islam2, Mochammad Hatta3, Firdaus Hamid3. The preclinical and clinical trials of mesenchymal stem cell’s secretome in traumatic brain injury: Review of basic science. 13-Jun-2025;16:235. Available from: <a href="https://surgicalneurologyint.com/?post_type=surgicalint_articles&p=13633">https://surgicalneurologyint.com/?post_type=surgicalint_articles&p=13633</a></p></div> </div> <div class="col-lg-3 col-sm-4 col-xs-12"><div class="article-detail-sidebar"><div class="icon sidebar-icon clearfix add-readinglist-icon"><button id="bookmark-article" class="add-reading-list-article">Add to Reading List</button><button id="bookmark-remove-article" class="remove-reading-list-article">Remove from Reading List</button></div><div class="icon sidebar-icon clearfix"><a class="btn btn-link" target="_blank" type="button" id="OpenPdf" href="https://surgicalneurologyint.com/wp-content/uploads/2025/06/13633/SNI-16-235.pdf"><img decoding="async" src="https://i1.wp.com/surgicalneurologyint.com/wp-content/themes/surgicalint/images/pdf-icon.png?w=604&#038;ssl=1" class="no-popup" data-recalc-dims="1"></a><a target="_blank" href="javascript:void(0);" onclick="return PrintArticle();"><img decoding="async" src="https://i0.wp.com/surgicalneurologyint.com/wp-content/themes/surgicalint/images/file-icon.png?w=604&#038;ssl=1" class="no-popup" data-recalc-dims="1"></a><a class="btn btn-link" type="button" id="EmaiLPDF"><img decoding="async" src="https://i1.wp.com/surgicalneurologyint.com/wp-content/themes/surgicalint/images/mail-icon.png?w=604&#038;ssl=1" class="no-popup" data-recalc-dims="1"></a></div><div class="date"> <p>Date of Submission<br><span class="darkgray">01-Dec-2024</span></p> <p>Date of Acceptance<br><span class="darkgray">09-Apr-2025</span></p> <p>Date of Web Publication<br><span class="darkgray">13-Jun-2025</span></p> </div> </div></div> </div> <!--.row --><div class="row"> <div class="blogparagraph col-lg-9 col-sm-8 col-xs-12"> <h3 class="blogheading pull-left Main-Title"><a href="javascript:void(0);" name="Abstract">Abstract</a></h3> <div class="clearfix"></div> <div class="hline"></div> <p><strong>Background: </strong>Traumatic brain injury (TBI) presents with associated neurologic and vascular damage triggers a chain of events that lead to a secondary brain injury. Proper prevention may limit undesirable outcomes. Mesenchymal stem cells (MSCs) and their secretome are promising therapeutic agents for a variety of neurological injuries, including TBI, due to their neuroprotective effects. This paper offers a concise overview of the use of MSCs and secretomes to prevent secondary brain injury and improve functional outcomes in TBI patients.</p><p><strong>Methods: </strong>An electronic database search on PubMed, Cochrane, Scopus, and clinicaltrials.gov was performed to include all relevant studies. Our framework incorporates an analysis of preclinical and clinical studies investigating the effects of MSCs and secretome on clinically relevant neurological and histopathological outcomes.</p><p><strong>Results: </strong>Immunomodulation by molecular factors secreted by MSCs is considered to be a key mechanism involved in their multi-potential therapeutic effects. Regulated neuroinflammation is required for healthy remodeling of the central nervous system during development and adulthood. Moreover, immune cells and their secreted factors can also contribute to tissue repair and neurological recovery following acute brain injury. The use of secretome has key advantages over cell-based therapies, such as lower immunogenicity and easy production, handling, and storage.</p><p><strong>Conclusion: </strong>Compared with traditional therapies, MSC and secretome treatment can directly improve TBI-induced pathological changes and promote recovery of neurological function. MSCs and their secretome hold great promise to bridge this gap in translation for TBI. Further clinical trials are needed to confirm its efficacy and safety.</p><p><strong>Keywords: </strong>Mesenchymal stem cell, Neuroprotection, Secretome, Traumatic brain injury</p><p></p></div> </div></body></html> </div><div><div class="row"> <div class="blogparagraph col-lg-9 col-sm-8 col-xs-12"><p></p><p><inline-graphic xmlns:xlink="http://www.w3.org/1999/xlink" xlink:href="SNI-16-235-inline001.tif"/></p><p></p><h3 class="blogheading Main-Title"><a href="javascript:void(0);" name="INTRODUCTION">INTRODUCTION</a></h3><div class="clearfix"></div><div class="hline"></div><p>More than 27 million people were diagnosed with and treated for traumatic brain injury (TBI) in 2016, with an age-adjusted incidence of 369/100,000 persons around the world, according to the largest study to date estimating the global incidence of TBI. More than 55 million people around the world are thought to be living with a TBI, according to this study.[<xref ref-type="bibr" rid="ref19"> <a href='#ref19'>19</a> </xref>] This represents roughly 0.7% of the global population. Low- and middle-income countries (e.g., Indonesia) have nearly 3 times as many TBI cases as high-income countries due to a higher prevalence of risk factors for TBI causes (e.g., motor vehicle crashes and falls from height) and differences in health-care systems that allow patients to seek medical care and address associated health effects. The estimated cost of non-fatal TBI-related health care in the United States in 2016 was $40.6 billion, while global hospitalization expenses for severe TBI ranged between $2,130 and $401,008. Developing countries also face these problems.[<xref ref-type="bibr" rid="ref18"> <a href='#ref18'>18</a> </xref>]</p><p>Multiple mechanisms, including excitotoxicity, mitochondrial dysfunction, oxidative stress, lipid peroxidation, neuroinflammation, axon degeneration, and apoptotic cell death, contribute to secondary injuries. Recent advances in neuroprotection have acknowledged this intricate structure and interplay, placing greater emphasis on therapeutic measures that encourage the recovery and optimal performance of nonneuronal cells as well as more directly obstructing neuronal cell death pathways . Moleac901 promotes neurogenesis.[<xref ref-type="bibr" rid="ref19"> <a href='#ref19'>19</a> </xref>] Previous studies showed the role of erythropoietin, high mobility group box 1, in potential treatment in TBI.[<xref ref-type="bibr" rid="ref19"> <a href='#ref19'>19</a> </xref>,<xref ref-type="bibr" rid="ref23"> <a href='#ref23'>23</a> </xref>] Mild hypothermia therapy also affects matrix metalloproteinase 9 (MMP-9) protein level, MMP-9 messenger RNA (mRNA), 1562 C/T Polymorphism, and Marshall computed tomography (CT) score in high-risk TBI.[<xref ref-type="bibr" rid="ref12"> <a href='#ref12'>12</a> </xref>,<xref ref-type="bibr" rid="ref13"> <a href='#ref13'>13</a> </xref>,<xref ref-type="bibr" rid="ref16"> <a href='#ref16'>16</a> </xref>] Mesenchymal stem cells (MSCs) also show tremendous promise in terms of neuroprotection in experimental TBI. Not only MSCs but also their released bioactive substances, known as the secretome, can provide immunomodulation for tissue regeneration and neurological recovery after TBI.[<xref ref-type="bibr" rid="ref23"> <a href='#ref23'>23</a> </xref>] Some previous research studied the potential therapeutic rat model of TBI. The model of rat TBI was also reported with a modification of the Marmarou model.[<xref ref-type="bibr" rid="ref15"> <a href='#ref15'>15</a> </xref>] Caffeic acid phenethyl ester can reduce brain edema in TBI subjects.[<xref ref-type="bibr" rid="ref14"> <a href='#ref14'>14</a> </xref>,<xref ref-type="bibr" rid="ref16"> <a href='#ref16'>16</a> </xref>]</p><p>In 2016, the Mesenchymal and Tissue Stem Cell Committee of the International Society for Cellular Therapy defined MSCs as meeting three criteria: (1) <i>In vitro</i> multipotent differentiation potential to osteoblasts, adipocytes, and chondroblasts; (2) must express specific surface markers of CD105, CD73, and CD90, and lack expression of CD45, CD34 or CD14 or CD11b, CD79 alpha or CD19, and human leukocyte antigen - DR(HLA-DR); and (3) must be plastic-adherent when maintained in standard culture conditions. MSCs have been extensively investigated in the treatment of numerous human diseases, including type 1 diabetes, neurodegenerative diseases, such as Parkinson’s disease and Alzheimer’s disease, spinal cord injuries, and tumors. Constant interaction between MSCs and the immune system is essential for maintaining tissue homeostasis and regulating inflammatory responses. MSCs can inhibit CD4+ (helper) and CD8+ (cytotoxic) T cells, affect B-cell functions through cell-to-cell contact, suppress the proliferation and cytotoxicity of natural killer (NK) cells, and increase regulatory T-cell (Treg) generation <i>in vitro</i> and <i>in vivo</i> through cell communication and soluble factors.[<xref ref-type="bibr" rid="ref7"> <a href='#ref7'>7</a> </xref>] Many studies indicate that many of the therapeutic effects of MSCs may be due to the paracrine substances secreted by extracellular vesicles rather than cellular engraftment and response to the site of injury. Exosomes derived from mesenchymal stem cells have been found to be effective in a growing number of animal models for the treatment of liver fibrosis, liver injury, hypoxic pulmonary hypertension, acute lung injury, acute renal injury, and cardiovascular diseases. This hypothesis is supported by preclinical studies demonstrating comparable or even enhanced organ function following infusion of mesenchymal stem cell conditioned medium(MSC-CM) as opposed to MSC transplantation. Furthermore, it has been demonstrated that MSC-derived exosomes are as effective as MSCs in enhancing functional outcomes, further demonstrating the significance of the secretome in promoting stroke repair. In TBI, exosomes from bone marrow mesenchymal stem cells (BMMSCs) could reduce neuroinflammation by regulating the phenotypes of microglia and aiding in the healing of nerve damage.</p><h3 class="blogheading Main-Title"><a href="javascript:void(0);" name="MATERIALS AND METHODS">MATERIALS AND METHODS</a></h3><div class="clearfix"></div><div class="hline"></div><h3 class = "title3">Literature search</h3><p>This methods section provides a structured approach for conducting a thorough and systematic review of the existing literature on MSC secretome in TBI, ensuring transparency, reproducibility, and rigor in the evaluation of preclinical and clinical evidence.</p><p>A comprehensive and systematic literature search was conducted to identify relevant studies on the preclinical and clinical trials of mesenchymal stem cell (MSC) secretome in the context of TBI. The search was performed using the following electronic databases up to June 2024: PubMed/MEDLINE, EMBASE, Web of Science, Scopus, and Cochrane Library.</p><p>The search strategy incorporated a combination of keywords and Medical Subject Headings terms related to MSC secretome and TBI. The primary search terms included: “Mesenchymal Stem Cells,” “MSC Secretome,” Traumatic Brain Injury,” “Preclinical Trials,” “Clinical Trials,” “Therapeutic Efficacy,” and “Neuroprotection.” Boolean operators (AND, OR) were used to refine the search. In addition, reference lists of identified articles and relevant reviews were manually searched to capture any studies not retrieved through the database search.</p><h3 class = "title3">Study selection</h3><p>The study inclusion and exclusion criteria were as follows: <list list-type="bullet"> <list-item><p>Inclusion criteria</p> <p><list list-type="order"> <list-item><p>Study Type: Both preclinical (<i>in vitro</i> and <i>in vivo</i>) and clinical studies evaluating the therapeutic potential of MSC secretome in TBI</p></list-item> <list-item><p>Population: Preclinical studies involving animal models of TBI and clinical studies involving human subjects with TBI</p></list-item> <list-item><p>Intervention: Administration of MSC-derived secretome</p></list-item> <list-item><p>Outcomes: Studies reporting on neuroprotective effects, functional recovery, molecular mechanisms, safety, and efficacy of MSC secretome in TBI</p></list-item> <list-item><p>Language: Articles published in English.</p></list-item> </list></p></list-item> <list-item><p>Exclusion criteria</p> <p><list list-type="order"> <list-item><p>Non-relevant Studies: Studies not focused on MSC secretome or not related to TBI</p></list-item> <list-item><p>Reviews and Meta-analyses: Articles that are reviews, editorials, commentaries, or letters without original data</p></list-item> <list-item><p>Duplicate Publications: Duplicate studies or multiple publications reporting the same data</p></list-item> <list-item><p>Incomplete Data: Studies lacking sufficient methodological details or outcome measures.</p></list-item> </list></p></list-item> </list></p><h3 class = "title3">Data extraction</h3><p>A standardized data extraction form was developed to collect pertinent information from each included study. The following data were extracted: general information, study design, intervention details, outcomes measured, and results.</p><h3 class = "title3">Data synthesis and quality assessment</h3><p>The extracted data were synthesized through a qualitative and quantitative approach to evaluate the therapeutic efficacy of MSC secretome in TBI. Preclinical and clinical studies were categorized based on their study design, intervention type, and outcome measures. The synthesis focused on neuroprotective effects, functional recovery, molecular mechanisms, safety and adverse events, and efficacy in clinical trials. A narrative synthesis was conducted to integrate findings across studies.</p><h3 class = "title3">Institutional Review Board (IRB)</h3><p>This article has been exempted from IRB review ref. No 9886/UN4.6.8/PJ.00.01/2024 from Faculty of Medicine, Hasanuddin University.</p><h3 class="blogheading Main-Title"><a href="javascript:void(0);" name="RESULTS">RESULTS</a></h3><div class="clearfix"></div><div class="hline"></div><p>Based on the systematic multi-database analysis, clinical trials of stem cells have been performed all around the world, with a total of 131 consisting of 101 observational and 30 interventional studies performed predominantly in the USA and China. In conclusion, these results suggested that only a small number of clinical trials focused on the transplantation of stem cells into patients with a relatively restricted range of diagnoses. This study was also supported by other reviews in which only five <i>in vitro</i> studies and nine <i>in vivo</i> studies until 2022 have been published to support data on the use of MSCs secretome in TBI.</p><p>Several clinical studies have evaluated the safety and efficacy of mesenchymal stem cell (MSC) therapy for patients with TBI, demonstrating potential benefits in neurological function, motor recovery, and inflammation reduction [<xref ref-type="fig" rid="F1"> <a href='#F1'>Figure 1</a> </xref>].</p><div class="row"> <div class="col-xs-12 content-figure col-wrap"> <div class="col-xs-2 figure-body col"><a href='javascript:void(0);' name='F1'></a> <br /><img src='https://i0.wp.com/surgicalneurologyint.com/wp-content/uploads/2025/06/13633/SNI-16-235-g001.png?w=604&#038;ssl=1' data-recalc-dims="1" /></div><div class="col-xs-10 col"> <div class="figure-content"><h3>Figure 1:</h3><p>Preferred Reporting Items for Systematic Reviews and Meta-Analyses flow chart. n: Number of articles.</p></div> </div> </div> </div><div class="clearfix">&nbsp;</div><h3 class = "title3">Motor function improvement in chronic TBI</h3><p>The Stem Cell Therapy for Traumatic Brain Injury( STEMTRA) trial (NCT02416492, Phase 2, <i>n</i> = 63) assessed the efficacy of allogeneic SB623 cell transplantation in chronic TBI patients with motor deficits. The study found a significant improvement in Fugl-Meyer Motor Scale scores at 24 weeks (<i>P</i> = 0.040), with no dose-limiting toxicities or deaths. However, secondary outcomes did not reach statistical significance.</p><h3 class = "title3">Safety and inflammatory response in acute TBI</h3><p>A Phase 1/2 study (NCT01575470, <i>n</i> = 25) on autologous bone marrow mononuclear cell (BMMNC) transplantation in severe TBI patients within 36 h post-injury reported no severe adverse events. In addition, structural preservation of brain tissue and a significant reduction in inflammatory markers (Interleukin-1beta [IL-1β], interferon-gamma [IFN-γ], and tumor necrosis factor-alpha [TNF-α]) were observed, suggesting a neuroprotective role of stem cell therapy.</p><h3 class = "title3">Neurological and functional recovery with umbilical cord MSCs</h3><p>A Phase 2 study (<i>n</i> = 40) investigating umbilical cord mesenchymal stem cell (UCMSC) transplantation in TBI patients with sequelae found significant improvements in neurological function and self-care ability based on Fugl-Meyer Assessments and Functional Independence Measures at 6 months post-transplantation.</p><h3 class = "title3">Reduction in intracranial pressure (ICP) and neurointensive care duration</h3><p>A Phase 1 study (<i>n</i> = 29) on autologous BMMNC therapy in pediatric TBI patients reported a significant reduction in Pediatric Intensity Level of Therapy scores within 24 h post-treatment, persisting for 1 week (<i>P</i> < 0.05). In addition, the therapy reduced the need for prolonged neurointensive care, as reflected in a shorter duration of ICP monitoring compared to controls (8.2 ± 1.3 vs. 15.6 ± 3.5 days, <i>P</i> = 0.03).</p><h3 class = "title3">Brain function and motor recovery in subacute TBI</h3><p>A Phase 1 study (<i>n</i> = 97) evaluating autologous BMMSC therapy through lumbar puncture reported functional improvements in 39.2% (38/97) of patients (<i>P</i> = 0.007). Among patients in a persistent vegetative state, 45.8% (11/24) showed increased consciousness (<i>P</i> = 0.024), while 37.0% (27/73) with motor disorders exhibited motor function recovery (<i>P</i> = 0.025). The age of patients and time from injury to therapy significantly influenced outcomes (<i>P</i> < 0.05), whereas the number of cell injections did not correlate with improvements (<i>P</i> > 0.05).</p><h3 class = "title3">Safety and feasibility in pediatric TBI</h3><p>A Phase 1 study (NCT00254722, <i>n</i> = 10) confirmed the safety and feasibility of autologous bone marrow progenitor cell infusion in pediatric TBI patients within 24 h of injury. The study reported no severe adverse effects and suggested potential functional improvements.</p><h3 class = "title3">Long-term safety and efficacy in chronic TBI</h3><p>An open-label Phase 1 study (NCT02028104, <i>n</i> = 50) on autologous BMMNC therapy in chronic TBI patients (1–65 years) demonstrated long-term safety and efficacy, with reported improvements in common TBI symptoms over follow-up periods.</p><h3 class = "title3">Preclinical studies on MSC secretome in TBI</h3><p>Preclinical research has further highlighted the therapeutic potential of MSC-derived secretome in modulating inflammation, reducing oxidative stress, and promoting neuroprotection in TBI models.</p><h3 class = "title3" style="color:green;">In vitro studies</h3><p><list list-type="bullet"> <list-item><p>Human adipose-derived MSCs (ASC) increased wound closure and reduced oxidative stress in injured astrocytes (Torrente <i>et al</i>., 2014)[<xref ref-type="bibr" rid="ref24"> <a href='#ref24'> 24 </a> </xref>]</p></list-item> <list-item><p>ASC-conditioned media promoted cell survival, reduced IL-1β expression, and decreased apoptosis in human neuroblastoma cells (Kappy <i>et al</i>., 2018)[<xref ref-type="bibr" rid="ref8"> <a href='#ref8'> 8 </a> </xref>]</p></list-item> <list-item><p>Mitochondrial neuroglobin (Ngb) was identified as a key mediator in the neuroprotective effects of ASC-conditioned medium, leading to reduced inflammation and enhanced mitochondrial function (Baez-Jurado <i>et al</i>., 2019).[<xref ref-type="bibr" rid="ref2"> <a href='#ref2'> 2 </a> </xref>]</p></list-item> </list></p><h3 class = "title3" style="color:green;">In vivo studies</h3><p><list list-type="bullet"> <list-item><p>Intravenous infusion of ASC secretome improved sensorimotor and cognitive functions, decreased neuroinflammation, and reduced apoptosis in a TBI rat model (Xu <i>et al</i>., 2020)[<xref ref-type="bibr" rid="ref28"> <a href='#ref28'> 28 </a> </xref>]</p></list-item> <list-item><p>Adipose-derived MSC transplants enhanced motor and cognitive function while reducing neuronal apoptosis and contusion volume, particularly in young rats (Tajiri <i>et al</i>., 2014).[<xref ref-type="bibr" rid="ref21"> <a href='#ref21'> 21 </a> </xref>]</p></list-item> </list></p><p>MSC therapy, including bone marrow-derived and umbilical cord-derived stem cells, has demonstrated safety and potential efficacy in improving neurological function, motor recovery, and inflammatory regulation in both clinical and preclinical TBI studies. While early-phase clinical trials suggest promising benefits, further large-scale randomized controlled trials are needed to confirm the long-term therapeutic effects and optimize treatment protocols.</p><h3 class="blogheading Main-Title"><a href="javascript:void(0);" name="DISCUSSION">DISCUSSION</a></h3><div class="clearfix"></div><div class="hline"></div><h3 class = "title3">Molecular pathophysiology of TBI</h3><p>Brain trauma can be categorized into primary and secondary brain injuries, which encompass the injury process. The occurrence of primary brain damage is attributed to the direct application of force on nerve cells, which might manifest as contact or inertial forces. These pathways ultimately result in neuronal apoptosis, synaptic plasticity, tissue injury, and cerebral shrinkage.[<xref ref-type="bibr" rid="ref26"> <a href='#ref26'> 26 </a> </xref>] Secondary injury mechanisms are also triggered immediately after the traumatic incident and continue as a sequence. Several biochemical changes contribute to secondary injury, including disruptions in cellular calcium balance, glutamate excitotoxicity, mitochondrial dysfunction, heightened production of free radicals, inflammation, increased lipid peroxidation, apoptosis, diffuse axonal injury, and breakdown of the blood-brain barrier (BBB). This inflammation has been implicated in both the initial and long-term aspects of neuropathology resulting from TBI.</p><p>Excitotoxicity is the main cause of various events that occur after TBI. Glutamate levels have been found to be at their peak immediately following TBI and remain elevated for a period of 24–48 h. This is mainly attributed to the mechanical disruption of the BBB. Some studies correlate the excessive glutamate levels linked to cerebral ischemia, seizures, and increased ICP.[<xref ref-type="bibr" rid="ref29"> <a href='#ref29'> 29 </a> </xref>] Elevated levels of calcium within cells trigger the activation of many destructive enzymes, such as phospholipases that harm cell and mitochondrial membranes, proteases that damage the cell’s cytoskeleton, and endonucleases that induce deoxyribonucleic acid (DNA) breakage. This leads to both apoptosis and necrosis.</p><p>Inflammation is a protective response that occurs when tissue homeostasis is disrupted by pathogens, physical agents, toxins, vascular changes, tissue necrosis, or immunological reactions. The immune cells present in the brain parenchyma, both those that originate from outside the brain and those that reside within it, release various substances that promote inflammation. These substances include damage-associated molecular patterns( DAMPs), cytokines, chemokines, reactive oxygen species(ROS), prostaglandins, and complement factors. Several studies have documented an increase in the expression of IL-1β, TNF-α, IL-6, C-C motif chemokine ligand 2(CCL2), CCL3, C-X-C motif chemokine ligand (CXCL1), CXCL2, CXCL8/IL-8, CXCL10, C-C chemokine receptor (CCR)2, CCR5, C-X-C chemokine receptor (CXCR) 4, and CX3CR1 within 6 h of TBI.[<xref ref-type="bibr" rid="ref25"> <a href='#ref25'> 25 </a> </xref>] The post-TBI triggers the release of inflammatory mediators that not only modify the resident cells of the central nervous system but also attract peripheral cells to enter the brain. Signals from the surrounding tissue microenvironment influence the polarization of these immune cells toward pro-inflammatory or anti-inflammatory phenotypes. After experiencing a TBI, astrocytes rapidly become active and undergo substantial structural alterations, including the shortening of their processes and swelling of their cell bodies, resulting in a hypertrophic form.[<xref ref-type="bibr" rid="ref1"> <a href='#ref1'> 1 </a> </xref>]</p><p>Astrocytes, when activated, perform the functions of engulfing debris and producing various substances such as cytokines, chemokines, and inflammatory mediators such as TNF-α, cyclooxygenase-2, and MMP-9 to support the ongoing inflammatory process. The activation of microglia during a TBI leads to an increase in the inflammatory response since it triggers the production and release of TNF-α and interleukins such as IL-1β and IL-6. Cytokine interactions lead to the movement of monocytes toward the location of the injury. At the site of the injury, cytokines, including IFN-γ and monocyte chemoattractant protein-1, continue to stimulate the macrophages, causing them to gather at the site of inflammation.[<xref ref-type="bibr" rid="ref11"> <a href='#ref11'> 11 </a> </xref>] The macrophages further contribute to the worsening of the inflammatory process by continuously producing TNF-α and IL-1 [<xref ref-type="fig" rid="F2"> <a href='#F2'> Figure 2 </a> </xref>].</p><div class="row"> <div class="col-xs-12 content-figure col-wrap"> <div class="col-xs-2 figure-body col"><a href='javascript:void(0);' name='F2'></a> <br /><img src='https://i0.wp.com/surgicalneurologyint.com/wp-content/uploads/2025/06/13633/SNI-16-235-g002.png?w=604&#038;ssl=1' data-recalc-dims="1" /></div><div class="col-xs-10 col"> <div class="figure-content"><h3>Figure 2:</h3><p>Pathophysiology of traumatic brain injury. APC: Antigen-presenting cell, ATP: Adenosine triphosphate, BBB: Blood-brain barrier, CREB: cAMP response element-binding protein, NMDA: N-methyl-D-aspartate, NOS: Nitric oxide synthase, OH: Hydroxyl radical, PKC: Protein kinase C, PLC: Phospholipase C, PN: Peroxynitrite, ROS: Reactive oxygen species, SDF: Stromal cell-derived factor, TNF: Tumor necrosis factor.</p></div> </div> </div> </div><div class="clearfix">&nbsp;</div><h3 class = "title3">Biomarkers following TBI</h3><p>Biomarkers associated with TBI are commonly quantified in bodily fluids. The majority of the data currently accessible were acquired through measures conducted in cerebrospinal fluid (CSF), blood (serum or plasma), or saliva. The glial protein S100B and neuron-specific enolase (NSE) were formerly thought to have a direct correlation with the severity of brain damage following an injury. The presence of markers in blood or other biological fluids may occur through BBB disruption or release, regardless of BBB integrity, or through passage through the recently found glymphatic system. The integrity of the BBB can be evaluated by measuring the ratio of albumin levels in the CSF to those in the serum. This assessment can help determine if the BBB has been compromised due to trauma or other pathological factors. In cases of severe TBI, the disruption of the BBB leads to an increase in the ratio of CSF to serum albumin. In cases of mild TBI, the ratio of CSF to serum albumin remains mostly within the normal range. Indicators of sudden damage to astroglial cells consist of increased levels of S100B and elevated levels of glial fibrillary acidic protein(GFAP), which can be identified in both CSF and peripheral blood.[<xref ref-type="bibr" rid="ref5"> <a href='#ref5'> 5 </a> </xref>]</p><p>NSE, α-II spectrin, and ubiquitin carboxyl-terminal hydrolase L1(UCH_L1) are biomarkers that indicate acute neuronal injury in TBI. Elevated levels of NSE were observed in both the CSF in the brain’s ventricles and the blood serum in investigations of severe TBI. The extent of this elevation was directly related to greater mortality rates and more severe scores on the Glasgow Coma Scale (GCS) for both adults and children. The increased levels of spectrin breakdown products and UCH-L1, along with GFAP, have been found to be directly connected to the severity of trauma. These biomarkers also enhance the predicted accuracy of the IMPACT outcome calculator for patients with severe TBI.[<xref ref-type="bibr" rid="ref6"> <a href='#ref6'> 6 </a> </xref>] Fluid indicators of acute axonal damage comprise tau proteins, neurofilament light peptide, and phosphorylated neurofilament heavy peptide.[<xref ref-type="bibr" rid="ref13"> <a href='#ref13'> 13 </a> </xref>,<xref ref-type="bibr" rid="ref18"> <a href='#ref18'> 18 </a> </xref>]</p><h3 class = "title3">Role of MSCs and secretome in TBI</h3><p>The medical community faces a significant problem in optimizing the therapy and prevention of subsequent damage after TBI. Animal investigations of brain vascular injuries have demonstrated that the administration of molecular agents that enhance the expression of transcription factors responsible for regulating cytoprotective proteins can provide neuroprotection and reduce the breakdown of the BBB.</p><p>The process of neurogenesis in the brain of a young adult can be broken down into a series of distinct developmental steps, each of which can be studied separately. These steps include the proliferation of precursor cells, the survival of newly born cells, their migration, and their differentiation into mature, functional neurons. Precursor cells can be stem cells, which have a slow-dividing cell cycle, long-term self-renewal potential, and multipotentiality, or progenitors, which have an increased rate of turnover and diminished self-renewal capabilities.</p><h3 class = "title3">Animal trial of MSCs in TBI</h3><p>According to a collection of laboratory studies conducted on TBI, the secretome was found to enhance the healing of wounds by promoting cell survival and growth, reversing structural changes, and increasing the movement and alignment of cells. These effects were observed to be dependent on the dosage of the secretome. The majority of the studies have demonstrated a significant improvement in mitochondrial function. This is achieved through the decrease in harmful free radicals (O<sub>2</sub><sup>−</sup>), the maintenance of the mitochondrial membrane potential, and the increase in the production of mitochondrial antioxidant enzymes (superoxide dismutase 2(SOD2), glutathione peroxidase 1(GPX-1), and catalase). In addition, there is a reduction in oxidative stress, decreased DNA damage and nuclear fragmentation, and modulation of inflammatory cytokines. Specifically, there is a decrease in the expression of IL-6, TNF-α, and granulocyte-macrophage colony-stimulating factor(GM-CSF) and an increase in the expression of IL-2 and IL-8. These findings indicate that the secretome treatment has a targeted effect in reducing apoptosis.</p><h3 class = "title3">Clinical trial of stem cell and cell therapy in TBI</h3><p>Several pioneer studies have shown the harmlessness and usefulness of cell therapy in treating pathological TBI. Based on an interim analysis of the STEMTRA trial, which included 63 TBI patients given allogeneic modified bone marrow-derived MSCs, they showed SB623 cell implantation appeared to be safe and well tolerated, and patients implanted with SB623 experienced significant improvement from baseline motor status at 6 months compared to controls.[<xref ref-type="bibr" rid="ref9"> <a href='#ref9'> 9 </a> </xref>] Sharma <i>et al</i>. studied to study the effects of intrathecal transplantation of autologous BMMNCs in 50 patients with chronic TBI. The results showed that 92% of patients experienced improvements in various symptoms and functional abilities. Factors such as age, severity of injury, and time since injury were found to affect the outcome of the transplantation. The study suggests that cell transplantation, combined with neurorehabilitation, can enhance functional recovery and improve the quality of life in patients with chronic TBI, particularly in younger patients with milder injuries.[<xref ref-type="bibr" rid="ref20"> <a href='#ref20'> 20 </a> </xref>]</p><p>Wang <i>et al</i>., in 2013, investigated the effects of UCMSC transplantation on patients with sequelae of TBI. The results showed that stem cell transplantation significantly improved neurological function, including upper and lower extremity motor function, sensation, balance, self-care, mobility, communication, and sphincter control. These findings suggest that stem cell transplantation with UCMSCs may be a safe and effective treatment for TBI sequelae. The study also highlighted the potential mechanisms through which stem cell transplantation may improve neurological function, including cell replacement, trophic support, and stimulation of endogenous neural repair and regeneration.[<xref ref-type="bibr" rid="ref27"> <a href='#ref27'> 27 </a> </xref>] Another study included a total of 97 patients with severe traumatic brain injury (sTBI) (24 with persistent vegetative state and 73 with disturbance in motor activity) who received autologous BMMSC therapy. The study showed that 38/97 patients (39.2%) improved in the function of the brain after transplant; 11/24 patients (45.8%) with persistent vegetative state showed improvements in consciousness; and 20/73 patients (37.0%) with a motor disorder showed improvements in motor functions.[<xref ref-type="bibr" rid="ref22"> <a href='#ref22'> 22 </a> </xref>]</p><p>Cox <i>et al</i>., in 2017, studied 25 patients with sTBI given the low dose, medium dose, and high dose of autologous BMMNCs to receive target doses of 6 × 10<sup>6</sup> BMMNC/kg, 9 × 10<sup>6</sup> BMMNC/kg, and 12 × 10<sup>6</sup> BMMNC/kg, respectively, for every group. Functional and neurocognitive outcomes were measured and correlated with imaging data. They concluded that treatment of BMMNC was safe and appeared to preserve critical regions of interest that correlated with functional outcomes.[<xref ref-type="bibr" rid="ref4"> <a href='#ref4'> 4 </a> </xref>] In a study regarding the use of BMMNC in pediatric patients, they studied 10 sTBI children treated with 6 × 10<sup>6</sup> autologous BMMNCs/kg body weight delivered intravenously within 48 h after TBI. The study concluded that GCS in 6 months showed 70% with good outcomes and 30% with moderate to severe disability. They also concluded the use of BMMNC in children is feasible and safe.[<xref ref-type="bibr" rid="ref3"> <a href='#ref3'> 3 </a> </xref>] Another study conducted in the pediatric population was conducted in 2015 by Liao <i>et al</i>. They included 29 sTBI patients which treated with intravenous autologous BMMNCs in 19 patients. The study showed that intravenous autologous BMMNC therapy was associated with lower treatment intensity required to manage ICP, associated severity of organ injury, and duration of neurointensive care following severe TBI.[<xref ref-type="bibr" rid="ref10"> <a href='#ref10'> 10 </a> </xref>] All studies are concluded in <xref ref-type="table" rid="T1"> <a href='#T1'> Table 1 </a> </xref>.</p><div class="row"> <div class="col-xs-12 content-figure col-wrap"> <div class="col-xs-2 figure-body col"><a href='javascript:void(0);' name='T1'></a> <br /><img src='https://i2.wp.com/surgicalneurologyint.com/wp-content/uploads/2025/06/13633/SNI-16-235-t001.png?w=604&#038;ssl=1' data-recalc-dims="1" /></div><div class="col-xs-10 col"> <div class="figure-content"><h3>Table 1:</h3><p>Clinical study for MSC in TBI.</p></div> </div> </div> </div><div class="clearfix">&nbsp;</div><h3 class = "title3">Preclinical study of the role of the secretome in TBI</h3><p>In general, the secretome is generated using a variety of protocols. The most frequently employed MSC source is adipose tissue (ASC). <xref ref-type="table" rid="T2"> <a href='#T2'> Table 2 </a> </xref> summarizes the findings of <i>in vitro</i> TBI studies that investigate the effects of the secretome and potential mechanisms of action. The same research group conducts the majority of studies and utilizes the human astrocyte-cell line (T98G) that has been exposed to scratch injury (which simulates mechanical injury) in conjunction with glucose deprivation (which induces metabolic impairment). The reduction of free radicals (O<sub>2</sub><sup>−</sup>) has been demonstrated to be a distinct benefit to mitochondrial function among secretome-induced mechanisms of action.[<xref ref-type="bibr" rid="ref24"> <a href='#ref24'> 24 </a> </xref>] The application of ASC-derived secretome treatment to scratch-injured neurons resulted in a reduction in cell mortality, diminished mitochondrial dysfunction, and a decrease in the expression of the pro-inflammatory cytokine IL-1β.[<xref ref-type="bibr" rid="ref8"> <a href='#ref8'> 8 </a> </xref>]</p><div class="row"> <div class="col-xs-12 content-figure col-wrap"> <div class="col-xs-2 figure-body col"><a href='javascript:void(0);' name='T2'></a> <br /><img src='https://i0.wp.com/surgicalneurologyint.com/wp-content/uploads/2025/06/13633/SNI-16-235-t002.png?w=604&#038;ssl=1' data-recalc-dims="1" /></div><div class="col-xs-10 col"> <div class="figure-content"><h3>Table 2:</h3><p>Preclinical study of MSC secretome.</p></div> </div> </div> </div><div class="clearfix">&nbsp;</div><p>Additional mechanisms of action included the decrease in the expression of IL-6, TNF-α, and GM-CSF, as well as the increase in the expression of IL-2 and IL-8 and the modulation of inflammatory cytokines. Intriguingly, the secretome treatment resulted in an increase in the production of Ngb in astrocytes. The secretome’s protective effects on mitochondrial injury in astrocytes were significantly diminished when Ngb was suppressed by small interfering RNA(siRNA).[<xref ref-type="bibr" rid="ref2"> <a href='#ref2'> 2 </a> </xref>] In rodent models of TBI, the secretome enhances functional recovery and improves neurological outcomes. Enhancements in cognitive function, including spatial learning and memory, and sensorimotor deficits. Secretome also possesses potent immunomodulatory properties, which result in a reduction in the expression of pro-inflammatory cytokines (e.g., IL-6 and TNF-α) and a concurrent increase in anti-inflammatory cytokines (e.g., Transforming growth factor-beta) in injured brain tissue. This shift in microglia/macrophage activation toward protective phenotypes is characterized by a decrease in Iba<sub>1</sub> +/NOS<sub>2</sub> + and an increase in Iba<sub>1</sub> +/Arg<sub>1</sub> + cells.[<xref ref-type="bibr" rid="ref28"> <a href='#ref28'> 28 </a> </xref>]</p><p>The efficacy of MSC or secretome in TBI in geriatric animals is inadequately investigated, with only a small amount of studies available. Tajiri <i>et al</i>. demonstrated the efficacy of intravenous ASCs transplantation or their secretome in young TBI rats (6 months of age). However, these interventions had diminished or no benefit in aged TBI rats (20 months of age). In particular, secretome treatment enhanced neurological recovery and diminished anatomical injury in young TBI rodents; however, it was ineffective in aged TBI rats. As an aside, the treatment with the cellular counterpart ASC resulted in an improvement in sensorimotor and cognitive functions, as well as reduced anatomical damage, in young rats. Conversely, it partially improved cognitive function recovery and anatomical damage in aged TBI rats, suggesting that the responsiveness to MSC or secretome treatment is age-dependent. The observation that secretome treatment is protective in young TBI animals but not in aged TBI animals may be attributed to the aged brain’s diminished capacity to respond to trophic/regenerative stimuli and/or a dysregulated immune response in the aged brain that is less responsive to secretome immunomodulatory activity.[<xref ref-type="bibr" rid="ref21"> <a href='#ref21'> 21 </a> </xref>]</p><p>The phenotypes and functions of immune cells have been identified as potently regulated by factors released by MSCs. Each time a particular factor is inhibited, the efficacy of MSCs is diminished, suggesting that this factor plays a critical role in comparison to the others. However, the majority of these observations have been made using <i>in vitro</i> systems that are overly simplified, as they only permit binary interactions between MSCs and immune cells. There is a possibility that a single compound will not be sufficient to accomplish protection <i>in vivo</i>, and the synergistic effects of multiple mediators will be required. This translation gap for complex neurological disorders, such as TBI, is of tremendous potential to be bridged by MSCs and their secretome. Nevertheless, the present challenge is to identify the optimal combination of bioactive factors released by MSCs that will enhance functional recovery and provide sustained neuroprotection.[<xref ref-type="bibr" rid="ref17"> <a href='#ref17'> 17 </a> </xref>,<xref ref-type="bibr" rid="ref21"> <a href='#ref21'> 21 </a> </xref>]</p><h3 class = "title3">Future consideration</h3><p>Significant data from preclinical research demonstrate that the secretome produced from mesenchymal stem cells (MSCs) is a promising biotherapeutic product that may effectively alleviate the pathological alterations associated with TBI. Although the secretome has several therapeutic advantages in treating TBI, it is important to acknowledge and tackle the limitations associated with its use in order to ensure its wider acceptance in clinical settings. Preclinical experiments have demonstrated the safety of secretome for the treatment of TBI. However, to achieve the creation of clinically quality secretome, it is crucial to standardize the manufacturing process. This is because variations in growth conditions and the source of MSCs might result in varying quantities of these secreted proteins. Similarly, it is necessary to standardize the characterization procedure to determine the precise quantities of different bioactive compounds. This will enable the selection of a suitable therapeutic dosage for administration. In addition to the challenges in production, it is crucial to take into account the mechanism of action of secretome for its successful use in regenerative medicine.</p><h3 class="blogheading Main-Title"><a href="javascript:void(0);" name="CONCLUSION">CONCLUSION</a></h3><div class="clearfix"></div><div class="hline"></div><p>MSCs have proven thorough examination in both preclinical and clinical studies. Multiple studies provide evidence supporting the safety and little danger of using MSCs as a cell-based therapy for treating TBI. The therapeutic effectiveness of secretome, independent of the cells, indicates that the paracrine mechanism, rather than differentiation, is one of the primary ways in which MSCs provide therapeutic advantages. Utilizing the released substances of MSCs for TBI therapy might have significant advantages in improving the physiological implications of neurological impairment in TBI.</p><p>The secretome provides therapeutic benefits through several pathways, including angiogenesis, neurogenesis, anti-inflammatory responses, and immunomodulatory properties. While there has been some progress in using secretome as a therapeutic approach for TBI, further work is required to address the obstacles identified in this study to apply these therapeutic strategies in a clinical setting successfully.</p><p></p><h3 class="blogheading Main-Title"><a href="javascript:void(0);" name="Ethical approval: ">Ethical approval: </a></h3><div class="clearfix"></div><div class="hline"></div><p>The Institutional Review Board approval is not required.</p><h3 class="blogheading Main-Title"><a href="javascript:void(0);" name="Declaration of patient consent: ">Declaration of patient consent: </a></h3><div class="clearfix"></div><div class="hline"></div><p>Patient’s consent was not required as there are no patients in this study.</p><h3 class="blogheading Main-Title"><a href="javascript:void(0);" name="Financial support and sponsorship: ">Financial support and sponsorship: </a></h3><div class="clearfix"></div><div class="hline"></div><p>Nil.</p><h3 class="blogheading Main-Title"><a href="javascript:void(0);" name="Conflicts of interest: ">Conflicts of interest: </a></h3><div class="clearfix"></div><div class="hline"></div><p>There are no conflicts of interest.</p><h3 class="blogheading Main-Title"><a href="javascript:void(0);" name="Use of artificial intelligence (AI)-assisted technology for manuscript preparation: ">Use of artificial intelligence (AI)-assisted technology for manuscript preparation: </a></h3><div class="clearfix"></div><div class="hline"></div><p>The authors confirm that there was no use of artificial intelligence (AI)-assisted technology for assisting in the writing or editing of the manuscript and no images were manipulated using AI.</p><h3 class="blogheading Main-Title"><a href="javascript:void(0);" name="Disclaimer">Disclaimer</a></h3><div class="clearfix"></div><div class="hline"></div><p>The views and opinions expressed in this article are those of the authors and do not necessarily reflect the official policy or position of the Journal or its management. The information contained in this article should not be considered to be medical advice; patients should consult their own physicians for advice as to their specific medical needs.</p></div> </div></div><div><div class="row"> <div class="blogparagraph col-lg-9 col-sm-8 col-xs-12"></div> </div><div class="row"> <div class="blogparagraph col-lg-9 col-sm-8 col-xs-12"> <h3 class="blogheading pull-left Main-Title"><a name="References" href="javascript:void(0);">References</a></h3> <div class="clearfix"></div> <div class="hline"></div> <p><a href='javascript:void(0);' name='ref1' style='text-decoration: none;'>1.</a> Abbott NJ, Rönnbäck L, Hansson E. Astrocyte-endothelial interactions at the blood-brain barrier. Nat Rev Neurosci. 2006. 7: 41-53</p><p><a href='javascript:void(0);' name='ref2' style='text-decoration: none;'>2.</a> Baez-Jurado E, Guio-Vega G, Hidalgo-Lanussa O, González J, Echeverria V, Ashraf G. Mitochondrial neuroglobin is necessary for protection induced by conditioned medium from human adipose-derived mesenchymal stem cells in astrocytic cells subjected to scratch and metabolic injury. Mol Neurobiol. 2019. 56: 5167-87</p><p><a href='javascript:void(0);' name='ref3' style='text-decoration: none;'>3.</a> Cox CS, Baumgartner JE, Harting MT, Worth LL, Walker PA, Shah SK. Autologous bone marrow mononuclear cell therapy for severe traumatic brain injury in children. Neurosurgery. 2011. 68: 588-600</p><p><a href='javascript:void(0);' name='ref4' style='text-decoration: none;'>4.</a> Cox CS, Hetz RA, Liao GP, Aertker BM, Ewing-Cobbs L, Juranek J. Treatment of severe adult traumatic brain injury using bone marrow mononuclear cells. Stem Cells. 2017. 35: 1065-79</p><p><a href='javascript:void(0);' name='ref5' style='text-decoration: none;'>5.</a> Czeiter E, Mondello S, Kovacs N, Sandor J, Gabrielli A, Schmid K. Brain injury biomarkers may improve the predictive power of the IMPACT outcome calculator. J Neurotrauma. 2012. 29: 1770-8</p><p><a href='javascript:void(0);' name='ref6' style='text-decoration: none;'>6.</a> GBD 2016 Traumatic Brain Injury and Spinal Cord Injury Collaborato. Global, regional, and national burden of traumatic brain injury and spinal cord injury 1990-2016: A systematic analysis for the global burden of disease study 2016. Lancet Neurol. 2019. 18: 56-87</p><p><a href='javascript:void(0);' name='ref7' style='text-decoration: none;'>7.</a> Jovic D, Yu Y, Wang D, Wang K, Li H, Xu F. A brief overview of global trends in MSC-based cell therapy. Stem Cell Rev Rep. 2022. 18: 1525-45</p><p><a href='javascript:void(0);' name='ref8' style='text-decoration: none;'>8.</a> Kappy NS, Chang S, Harris WM, Plastini M, Ortiz T, Zhang P. Human adipose-derived stem cell treatment modulates cellular protection in both in vitro and in vivo traumatic brain injury models. J Trauma Acute Care Surg. 2018. 84: 745-51</p><p><a href='javascript:void(0);' name='ref9' style='text-decoration: none;'>9.</a> Kawabori M, Weintraub AH, Imai H, Zinkevych I, McAllister P, Steinberg GK. Cell therapy for chronic TBI: Interim analysis of the randomized controlled stemtra trial. Neurology. 2021. 96: e1202-14</p><p><a href='javascript:void(0);' name='ref10' style='text-decoration: none;'>10.</a> Liao GP, Harting MT, Hetz RA, Walker PA, Shah SK, Corkins CJ. Autologous bone marrow mononuclear cells reduce therapeutic intensity for severe traumatic brain injury in children. Pediatr Crit Care Med. 2015. 16: 245-55</p><p><a href='javascript:void(0);' name='ref11' style='text-decoration: none;'>11.</a> Lucas SM, Rothwell NJ, Gibson RM. The role of inflammation in CNS injury and disease. Br J Pharmacol. 2006. 147: S232-40</p><p><a href='javascript:void(0);' name='ref12' style='text-decoration: none;'>12.</a> Muhammad SA. Mesenchymal stromal cell secretome as a therapeutic strategy for traumatic brain injury. Biofactors. 2019. 45: 880-91</p><p><a href='javascript:void(0);' name='ref13' style='text-decoration: none;'>13.</a> Nasution RA, Islam AA, Hatta M, Prihantono , Kaelan C, Poniman J. Modification of the marmarou model in developing countries. Ann Med Surg (Lond). 2020. 57: 109-13</p><p><a href='javascript:void(0);' name='ref14' style='text-decoration: none;'>14.</a> Nasution RA, Islam AA, Hatta M, Prihantono , Warsinggih , Ludong DH. Effects of caffeic acid phenethyl ester in reducing cerebral edema in rat subjects experiencing brain injury: An in vivo study. Ann Med Surg (Lond). 2020. 57: 328-33</p><p><a href='javascript:void(0);' name='ref15' style='text-decoration: none;'>15.</a> Prasetyo E, Islam AA, Hatta M, Widodo D, Pattelongi I. The effect of mild hypothermia therapy in the level of mmp-9 protein and the marshall ct score in high risk traumatic brain injury. Crit Care Shock. 2018. 21: 25-32</p><p><a href='javascript:void(0);' name='ref16' style='text-decoration: none;'>16.</a> Prasetyo E, Islam AA, Hatta M, Widodo D, Pattelongi I. The profile of MMP-9, MMP-9 mRNA expression,-1562 C/T polymorphism and outcome in high-risk traumatic brain injury: The effect of therapeutic mild hypothermia. Neurol Med Chir (Tokyo). 2017. 57: 612-9</p><p><a href='javascript:void(0);' name='ref17' style='text-decoration: none;'>17.</a> Rosyidi RM, Priyanto B, Islam AA, Hatta M, Bukhari A, Prihastomo KT. Role of MLC901 in increasing neurogenesis in rats with traumatic brain injury. Ann Med Surg (Lond). 2020. 60: 36-40</p><p><a href='javascript:void(0);' name='ref18' style='text-decoration: none;'>18.</a> Rosyidi RM, Priyanto B, Laraswati NK, Islam AA, Hatta M, Bukhari A. Characteristics and clinical outcome of traumatic brain injury in Lombok, Indonesia. Interdiscip Neurosurg. 2019. 18: 100470</p><p><a href='javascript:void(0);' name='ref19' style='text-decoration: none;'>19.</a> Said MF, Islam AA, Massi MN, Prihantono , Hatta M, Patellongi IJ. Effect of erythropoietin administration on expression of mRNA brain-derived neurotrophic factor, levels of stromal cell-derived factor-1, and neuron specific enolase in brain injury model Sprague Dawle y. Ann Med Surg. 2021. 70: 102877</p><p><a href='javascript:void(0);' name='ref20' style='text-decoration: none;'>20.</a> Sharma AK, Sane HM, Kulkarni PP, Gokulchandran N, Biju H, Badhe PB. Autologous bone marrow mononuclear cell transplantation in patients with chronic traumatic brain injury-a clinical study. Cell Regen. 2020. 9: 3</p><p><a href='javascript:void(0);' name='ref21' style='text-decoration: none;'>21.</a> Tajiri N, Acosta SA, Shahaduzzaman , Ishikawa H, Shinozuka K, Pabon M. Intravenous transplants of human adipose-derived stem cell protect the brain from traumatic brain injury-induced neurodegeneration and motor and cognitive impairments: Cell graft biodistribution and soluble factors in young and aged rats. J Neurosci. 2014. 34: 313-26</p><p><a href='javascript:void(0);' name='ref22' style='text-decoration: none;'>22.</a> Tian C, Wang X, Wang X, Wang L, Wang X, Wu S. Autologous bone marrow mesenchymal stem cell therapy in the subacute stage of traumatic brain injury by lumbar puncture. Exp Clin Transplant. 2013. 11: 176-81</p><p><a href='javascript:void(0);' name='ref23' style='text-decoration: none;'>23.</a> Tommy T, Islam AA, Hatta M, Bukhari A. Immunomodulatory properties of high mobility group box 1 and its potential role in brain injury: Review article. Ann Med Surg (Lond)2020;. 59: 106-9</p><p><a href='javascript:void(0);' name='ref24' style='text-decoration: none;'>24.</a> Torrente D, Avila MF, Cabezas R, Morales L, Gonzalez J, Samudio I. Paracrine factors of human mesenchymal stem cells increase wound closure and reduce reactive oxygen species production in a traumatic brain injury in vitro model. Hum Exp Toxicol. 2014. 33: 673-84</p><p><a href='javascript:void(0);' name='ref25' style='text-decoration: none;'>25.</a> Tweedie D, Karnati HK, Mullins R, Pick CG, Hoffer BJ, Goetzl EJ. Time-dependent cytokine and chemokine changes in mouse cerebral cortex following a mild traumatic brain injury. eLife. 2020. 9: e55827</p><p><a href='javascript:void(0);' name='ref26' style='text-decoration: none;'>26.</a> Veenith T, Goon SS, Burnstein RM. Molecular mechanisms of traumatic brain injury: The missing link in management. World J Emerg Surg. 2009. 4: 7</p><p><a href='javascript:void(0);' name='ref27' style='text-decoration: none;'>27.</a> Wang S, Cheng H, Dai G, Wang X, Hua R, Liu X. Umbilical cord mesenchymal stem cell transplantation significantly improves neurological function in patients with sequelae of traumatic brain injury. Brain Res. 2013. 1532: 76-84</p><p><a href='javascript:void(0);' name='ref28' style='text-decoration: none;'>28.</a> Xu C, Diao YF, Wang J, Liang J, Xu HH, Zhao ML. Intravenously infusing the secretome of adipose-derived mesenchymal stem cells ameliorates neuroinflammation and neurological functioning after traumatic brain injury. Stem Cells Dev. 2020. 29: 222-34</p><p><a href='javascript:void(0);' name='ref29' style='text-decoration: none;'>29.</a> Zauner A, Bullock R, Kuta AJ, Woodward J, Young HF. Glutamate release and cerebral blood flow after severe human head injury. Acta Neurochir Suppl. 1996. 67: 40-4</p></div> </div></div>
  2. Abscess formation within brain metastasis

    Fri, 13 Jun 2025 17:09:53 -0000

    Abscess formation within brain metastasis Category: Article Type: Emilio González Martínez, Giancarlo Daniel Mattos PiaggioDepartment of Neurological Surgery, University Hospital of León, León, SpainCorrespondence Address:Emilio González Martínez, Department of Neurological Surgery, University Hospital of León, León, Spain.DOI:10.25259/SNI_62_2025Copyright: © 2025 Surgical Neurology International This is an open-access article distributed under the terms of the Creative Commons … Continue reading Abscess formation within brain metastasis
    <div><!DOCTYPE html PUBLIC "-//W3C//DTD HTML 4.0 Transitional//EN" "http://www.w3.org/TR/REC-html40/loose.dtd"> <html><head><meta http-equiv="content-type" content="text/html; charset=utf-8"></head><body><div class="row"><div class="col-lg-9 col-sm-8 col-xs-12"><div class="media-body details-body"> <a href="https://surgicalneurologyint.com/?post_type=surgicalint_articles&p=13632"><h2 class="media-heading"><h2 class="media-heading">Abscess formation within brain metastasis</h2></h2></a> </div><div class="disp_categories"> <p><label>Category: </label><span></span></p> <p><label>Article Type: </label><span></span></p> </div><a href="mailto:egonzalezmart@saludcastillayleon.es" target="_top">Emilio González Martínez</a>, <a href="mailto:gmattospiaggio@gmail.com" target="_top">Giancarlo Daniel Mattos Piaggio</a><ol class="smalllist"><li>Department of Neurological Surgery, University Hospital of León, León, Spain</li></ol><p><strong>Correspondence Address:</strong><br>Emilio González Martínez, Department of Neurological Surgery, University Hospital of León, León, Spain.<br></p><p><strong>DOI:</strong>10.25259/SNI_62_2025</p>Copyright: © 2025 Surgical Neurology International This is an open-access article distributed under the terms of the Creative Commons Attribution-Non Commercial-Share Alike 4.0 License, which allows others to remix, transform, and build upon the work non-commercially, as long as the author is credited and the new creations are licensed under the identical terms.<div class="parablock"><p><strong>How to cite this article: </strong>Emilio González Martínez, Giancarlo Daniel Mattos Piaggio. Abscess formation within brain metastasis. 13-Jun-2025;16:236</p></div><div class="parablock"><p><strong>How to cite this URL: </strong>Emilio González Martínez, Giancarlo Daniel Mattos Piaggio. Abscess formation within brain metastasis. 13-Jun-2025;16:236. Available from: <a href="https://surgicalneurologyint.com/?post_type=surgicalint_articles&p=13632">https://surgicalneurologyint.com/?post_type=surgicalint_articles&p=13632</a></p></div> </div> <div class="col-lg-3 col-sm-4 col-xs-12"><div class="article-detail-sidebar"><div class="icon sidebar-icon clearfix add-readinglist-icon"><button id="bookmark-article" class="add-reading-list-article">Add to Reading List</button><button id="bookmark-remove-article" class="remove-reading-list-article">Remove from Reading List</button></div><div class="icon sidebar-icon clearfix"><a class="btn btn-link" target="_blank" type="button" id="OpenPdf" href="https://surgicalneurologyint.com/wp-content/uploads/2025/06/13632/SNI-16-236.pdf"><img decoding="async" src="https://i1.wp.com/surgicalneurologyint.com/wp-content/themes/surgicalint/images/pdf-icon.png?w=604&#038;ssl=1" class="no-popup" data-recalc-dims="1"></a><a target="_blank" href="javascript:void(0);" onclick="return PrintArticle();"><img decoding="async" src="https://i0.wp.com/surgicalneurologyint.com/wp-content/themes/surgicalint/images/file-icon.png?w=604&#038;ssl=1" class="no-popup" data-recalc-dims="1"></a><a class="btn btn-link" type="button" id="EmaiLPDF"><img decoding="async" src="https://i1.wp.com/surgicalneurologyint.com/wp-content/themes/surgicalint/images/mail-icon.png?w=604&#038;ssl=1" class="no-popup" data-recalc-dims="1"></a></div><div class="date"> <p>Date of Submission<br><span class="darkgray">19-Jan-2025</span></p> <p>Date of Acceptance<br><span class="darkgray">06-May-2025</span></p> <p>Date of Web Publication<br><span class="darkgray">13-Jun-2025</span></p> </div> </div></div> </div> <!--.row --><div class="row"> <div class="blogparagraph col-lg-9 col-sm-8 col-xs-12"> <h3 class="blogheading pull-left Main-Title"><a href="javascript:void(0);" name="Abstract">Abstract</a></h3> <div class="clearfix"></div> <div class="hline"></div> <p><strong>Background: </strong>Brain abscesses are a potentially curable but life-threatening medical condition. Their occurrence within brain metastases has been exceptionally reported. In this study, we describe in detail this rare entity.</p><p><strong>Case Description: </strong>A previously healthy 64-year-old female was referred to our neurosurgical department with right-sided deviation of the oral commissure and a 7-day history of holocranial headache. Magnetic resonance imaging revealed a tumor in the right frontal lobe, exhibiting an iso-intense signal on T1-weighted images with heterogeneous enhancement. On diffusion-weighted imaging, the lesion displayed a hyperintense signal. A body computed tomography scan identified a lung tumor in the left superior lobe and a potential metastasis in the right adrenal gland. Intraoperative findings and histopathological examination revealed metastasis from lung adenocarcinoma. In addition, purulent content was noted within the abscess, and cultures identified <i>Staphylococcus epidermidis</i> and <i>Streptococcus mitis</i>.</p><p><strong>Conclusion: </strong>Abscess formation within metastases is rarely diagnosed preoperatively, primarily because these lesions do not present with characteristic clinical or radiological features. Early recognition of this entity is crucial for establishing an appropriate treatment plan.</p><p><strong>Keywords: </strong>Abscess formation, Lung cancer, Metastasis, Neuro-oncology</p><p></p></div> </div></body></html> </div><div><div class="row"> <div class="blogparagraph col-lg-9 col-sm-8 col-xs-12"><p></p><p><inline-graphic xmlns:xlink="http://www.w3.org/1999/xlink" xlink:href="SNI-16-236-inline001.tif"/></p><p></p><h3 class="blogheading Main-Title"><a href="javascript:void(0);" name="INTRODUCTION">INTRODUCTION</a></h3><div class="clearfix"></div><div class="hline"></div><p>The formation of an abscess within brain metastasis is an event infrequently reported in the literature. The lack of awareness about this entity leads to misdiagnosis. The development of different sequences of magnetic resonance imaging (MRI), such as diffusion-weighted imaging (DWI) or apparent diffusion coefficient (ADC), and spectroscopy may help to diagnose.[<xref ref-type="bibr" rid="ref4"> <a href='#ref4'>4</a> </xref>]</p><p>We report a case of brain metastasis from lung adenocarcinoma with a concomitant abscess formation. This study aims to review the main characteristics of this rare phenomenon.</p><h3 class="blogheading Main-Title"><a href="javascript:void(0);" name="CASE DESCRIPTION">CASE DESCRIPTION</a></h3><div class="clearfix"></div><div class="hline"></div><p>A previously healthy 64-year-old female was admitted to our Neurosurgical Department due to the discovery of a cerebral tumor on a computed tomography (CT) scan. She had smoked approximately 20 cigarettes/day for 15 years before admission. No other relevant medical conditions were noted, although she had multiple missing teeth. On admission, she presented right-sided deviation of the oral commissure and a 7-day holocranial headache. Fever or neck stiffness was not detected. Physical examination only showed a left flattened nasolabial fold.</p><p>A chest X-ray showed a left upper-lobe lung mass. MRI revealed a 25 mm rounded lesion in the right-lobar frontal tumor with an associated hemispheric vasogenic edema and 5 mm displacement of the midline. On T1-weighted images, the lesion presented an iso-intense signal with a heterogeneous center and annular enhancement after gadolinium administration and a heterogeneous signal on T2-weighted images. On DWI, it showed a central slight hyper-intense signal and a halo of hypo-intense signal [<xref ref-type="fig" rid="F1"> <a href='#F1'>Figure 1</a> </xref>]. A body CT scan confirmed the presence of the lung mass in the left superior lobe, along with mediastinal and perihilar bilateral adenopathies and bronchogenic seed, as well as a lesion in the right adrenal gland consistent with metastasis. Blood test revealed no significant inflammatory parameters, but carcinoembryonic antigen, CA 15-3, and CA 125 were elevated.</p><div class="row"> <div class="col-xs-12 content-figure col-wrap"> <div class="col-xs-2 figure-body col"><a href='javascript:void(0);' name='F1'></a> <br /><img src='https://i0.wp.com/surgicalneurologyint.com/wp-content/uploads/2025/06/13632/SNI-16-236-g001.png?w=604&#038;ssl=1' data-recalc-dims="1" /></div><div class="col-xs-10 col"> <div class="figure-content"><h3>Figure 1:</h3><p>Magnetic resonance imaging reveals a mass lesion in the right frontal lobe. (a) T1-weighted images reveal a mass lesion in the right frontal lobe that appears iso-intense. (b) Gadolinium-enhanced sequences show irregular enhancement with an annular ring. (c) T2-weighted images demonstrate a heterogeneous signal. (d) Diffusion-weighted imaging (DWI) shows central areas of high signal intensity (restriction) interspersed with peripheral regions of low signal intensity (no restriction).</p></div> </div> </div> </div><div class="clearfix">&nbsp;</div><p>Due to the extension of the primary tumor, thoracic surgery was declined, but cranial surgery was performed. The tumor was approached through a right frontal craniotomy and a transsulcal approach. During the procedure, the lesion was punctured, and unexpectedly, purulent material flowed from inside and, consequently, was collected. Finally, the tumor was completely removed. Five days after surgery, the patient was discharged without complications. Cultured from the pus isolated <i>Staphylococcus epidermidis</i> and <i>Streptococcus mitis</i>. Pathology revealed epithelial cells with mitotic activity and mucoid cytoplasmic inclusions forming a glandular but irregular pattern, consistent with metastasis from lung adenocarcinoma [<xref ref-type="fig" rid="F2"> <a href='#F2'>Figure 2</a> </xref>]. The patient completed a 6-week regimen of ceftriaxone and was followed by whole-brain radiotherapy and systemic chemotherapy. After 8 months of follow-up, the patient remains stable.</p><div class="row"> <div class="col-xs-12 content-figure col-wrap"> <div class="col-xs-2 figure-body col"><a href='javascript:void(0);' name='F2'></a> <br /><img src='https://i1.wp.com/surgicalneurologyint.com/wp-content/uploads/2025/06/13632/SNI-16-236-g002.png?w=604&#038;ssl=1' data-recalc-dims="1" /></div><div class="col-xs-10 col"> <div class="figure-content"><h3>Figure 2:</h3><p>(a) Hematoxylin and eosin stain of the brain lesion (×10). (b) Periodic acid–Schiff stain (×40). Epithelial cells and mucoid cytoplasmic inclusions – arrows – which determine their glandular origin.</p></div> </div> </div> </div><div class="clearfix">&nbsp;</div><h3 class="blogheading Main-Title"><a href="javascript:void(0);" name="DISCUSSION">DISCUSSION</a></h3><div class="clearfix"></div><div class="hline"></div><p>Approximately 40% of cancer patients develop brain metastases during their disease. Melanoma has the highest propensity to metastasize into the brain, followed by lung and breast carcinomas.[<xref ref-type="bibr" rid="ref3"> <a href='#ref3'>3</a> </xref>] Overall, 50% of metastases arise from lung carcinoma, followed by breast carcinoma.[<xref ref-type="bibr" rid="ref5"> <a href='#ref5'>5</a> </xref>,<xref ref-type="bibr" rid="ref18"> <a href='#ref18'>18</a> </xref>,<xref ref-type="bibr" rid="ref26"> <a href='#ref26'>26</a> </xref>] Brain abscesses, on the other hand, are a well-known process and relatively common in daily neurosurgical practice.</p><p>Although both brain metastases and brain abscesses are common, their coexistence is rare. This phenomenon has been reported mainly in pituitary adenomas, meningiomas, gliomas, and rarely in craniopharyngiomas, Rathke’s cleft cyst, and primary central nervous system lymphoma.[<xref ref-type="bibr" rid="ref8"> <a href='#ref8'>8</a> </xref>-<xref ref-type="bibr" rid="ref10"> <a href='#ref10'>10</a> </xref>,<xref ref-type="bibr" rid="ref12"> <a href='#ref12'>12</a> </xref>-<xref ref-type="bibr" rid="ref14"> <a href='#ref14'>14</a> </xref>,<xref ref-type="bibr" rid="ref24"> <a href='#ref24'>24</a> </xref>] To our knowledge, abscess formation within metastases has been documented in only eight patients.[<xref ref-type="bibr" rid="ref1"> <a href='#ref1'>1</a> </xref>,<xref ref-type="bibr" rid="ref4"> <a href='#ref4'>4</a> </xref>,<xref ref-type="bibr" rid="ref16"> <a href='#ref16'>16</a> </xref>,<xref ref-type="bibr" rid="ref19"> <a href='#ref19'>19</a> </xref>,<xref ref-type="bibr" rid="ref21"> <a href='#ref21'>21</a> </xref>,<xref ref-type="bibr" rid="ref23"> <a href='#ref23'>23</a> </xref>,<xref ref-type="bibr" rid="ref27"> <a href='#ref27'>27</a> </xref>]</p><h3 class = "title3">Pathophysiology of abscess formation in metastases</h3><p>It is remarkable how some cancers metastasize to the brain, despite the blood–brain barrier (BBB) being the tightest barrier in the body. The current studies have shown that non-small cell lung carcinomas overexpress proteins involved in interactions between metastatic cells and endothelial cells, as well as in angiogenesis and reprogramming of glial cells toward an immunosuppressive state.[<xref ref-type="bibr" rid="ref30"> <a href='#ref30'> 30 </a> </xref>] The process of abscess formation involves several mechanisms, including hematogenous co-metastasis of bacteria and tumoral cells, over-infection from distant infection, or a single coincidental event. Metastatic cells passing through the BBB induce endothelial cell impairment, which makes it easier for the penetration and proliferation of bacterial agents.[<xref ref-type="bibr" rid="ref1"> <a href='#ref1'> 1 </a> </xref>] This process may be further facilitated by the optimal environment provided by tumoral necrosis and immunosuppressive conditions, such as diabetes mellitus, steroid use, chronic diseases, or other acquired immunodeficiencies.[<xref ref-type="bibr" rid="ref1"> <a href='#ref1'> 1 </a> </xref>,<xref ref-type="bibr" rid="ref27"> <a href='#ref27'> 27 </a> </xref>] No known predisposing factors were identified in our patient, although her immunological status was not assessed.</p><h3 class = "title3">Bacteriology</h3><p>Up to 50% of cultures of abscess material in brain tumors are negative. Gram-positive bacteria are more common in metastatic tumors, though cases with <i>Salmonella</i>, diphtheroids, or Acinetobacter have also been reported.[<xref ref-type="bibr" rid="ref11"> <a href='#ref11'> 11 </a> </xref>,<xref ref-type="bibr" rid="ref16"> <a href='#ref16'> 16 </a> </xref>,<xref ref-type="bibr" rid="ref19"> <a href='#ref19'> 19 </a> </xref>,<xref ref-type="bibr" rid="ref23"> <a href='#ref23'> 23 </a> </xref>] In our patient, <i>S. epidermidis</i> and <i>S. mitis</i> were isolated from the pus. <i>S. mitis</i> is a member of the <i>Streptococcus viridans</i> group, which is involved in almost two-thirds of brain abscesses.[<xref ref-type="bibr" rid="ref15"> <a href='#ref15'> 15 </a> </xref>] Both <i>S. mitis</i> and <i>S. epidermidis</i> are oral commensals that can cause polymicrobial brain abscesses.[<xref ref-type="bibr" rid="ref20"> <a href='#ref20'> 20 </a> </xref>,<xref ref-type="bibr" rid="ref22"> <a href='#ref22'> 22 </a> </xref>] The disruption of the tooth-tissue interface in our patient may have facilitated their entry into the bloodstream, leading to abscess formation.</p><p>In pathological studies, although bacteria are not usually observed, some inflammatory features, such as pus or leukocytes, may be seen.[<xref ref-type="bibr" rid="ref14"> <a href='#ref14'> 14 </a> </xref>,<xref ref-type="bibr" rid="ref17"> <a href='#ref17'> 17 </a> </xref>]</p><h3 class = "title3">Clinical presentation and blood test findings</h3><p>Abscess formation within tumors rarely causes the classic triad of abscess symptoms, which includes fever, meningeal signs, and altered levels of consciousness. Clinical features vary depending on the location of the metastasis. Intracranial hypertension symptoms and symptoms secondary to mass effect, such as headache, focal neurologic deficit, or ataxia and gait disturbance in metastasis located in the posterior fossa, are common.[<xref ref-type="bibr" rid="ref4"> <a href='#ref4'> 4 </a> </xref>,<xref ref-type="bibr" rid="ref11"> <a href='#ref11'> 11 </a> </xref>,<xref ref-type="bibr" rid="ref19"> <a href='#ref19'> 19 </a> </xref>]</p><p>Particularly, pyrexia is as frequent as focal deficit in abscess formation in meningiomas, while sellar tumors often with chronic headache, visual impairment, and hypopituitarism symptoms.[<xref ref-type="bibr" rid="ref6"> <a href='#ref6'> 6 </a> </xref>,<xref ref-type="bibr" rid="ref14"> <a href='#ref14'> 14 </a> </xref>]</p><p>In cases of abscess formation, infectious signs such as fever or meningismus, as well as inflammatory parameters in blood tests, may be absent. These features may be missing even in the presence of a brain abscess due to the lack of overt systemic infection, which complicates diagnosis.[<xref ref-type="bibr" rid="ref8"> <a href='#ref8'> 8 </a> </xref>,<xref ref-type="bibr" rid="ref28"> <a href='#ref28'> 28 </a> </xref>]</p><h3 class = "title3">Radiological features</h3><p>Metastases, gliomas, and abscesses are frequently confused on conventional MRI.[<xref ref-type="bibr" rid="ref2"> <a href='#ref2'> 2 </a> </xref>] These lesions generally present with hypo-intense signals on T1-weighted, resulting from intratumoral necrosis, with peripheral enhancement after contrast administration. On T2-weighted images, they display heterogeneous or hyperintense signal. All of these lesions may exhibit a ring-enhancing pattern, although this is more typical in brain abscesses.</p><p>Recent advances in MRI, including DWI and ADC techniques, have greatly improved the diagnosis of a brain abscess. On DWI, brain abscesses show homogenously hyper-intense signals because of restricted diffusion, along with a significantly reduced ADC, reflecting their high viscosity and cellularity. In contrast, metastases and gliomas commonly exhibit unrestricted diffusion within the lesion and restricted diffusion in their borders on DWI, with low signal on ADC. In the present case, the lesion exhibited mixed features on DWI, which have previously been associated with irregular densities of pus, tumor cells, necrosis areas, or hemorrhage.[<xref ref-type="bibr" rid="ref14"> <a href='#ref14'> 14 </a> </xref>,<xref ref-type="bibr" rid="ref29"> <a href='#ref29'> 29 </a> </xref>]</p><p>In brain spectroscopy, brain abscess shows a high level of lactate, alanine, cytosolic acid, and acetate, which originate from the enhanced glycolysis and fermentation of the infecting micro-organisms. However, these findings are inconsistent. A high choline/creatine ratio is often observed in metastatic tumors, whereas this ratio tends to be lower in cerebral abscesses.[<xref ref-type="bibr" rid="ref2"> <a href='#ref2'> 2 </a> </xref>,<xref ref-type="bibr" rid="ref7"> <a href='#ref7'> 7 </a> </xref>] However, the mixed signal pattern in cases of brain metastases with abscess formation can create overlapping features, making the diagnosis challenging.[<xref ref-type="bibr" rid="ref14"> <a href='#ref14'> 14 </a> </xref>]</p><h3 class = "title3">Treatment and outcome</h3><p>Without surgical intervention, abscess formation within metastases may lead to acute neurologic deterioration and death. Unexpected abscess formation may be overlooked during surgical excision if the purulent content is not collected and cultured, often being mistaken for a necrotic liquefied cyst.[<xref ref-type="bibr" rid="ref4"> <a href='#ref4'> 4 </a> </xref>] Therefore, total tumor resection followed by specific antibiotic therapy is essential.</p><p>The overall survival rate has significantly improved with recent advances in immunotherapy and chemotherapy. The presence of brain metastases remains a critical factor for poor prognosis, being the median overall survival in these cases being approximately 12 months.[<xref ref-type="bibr" rid="ref25"> <a href='#ref25'> 25 </a> </xref>] Nevertheless, the concurrence of abscess formation within brain metastases does not significantly impact prognosis.[<xref ref-type="bibr" rid="ref11"> <a href='#ref11'> 11 </a> </xref>,<xref ref-type="bibr" rid="ref19"> <a href='#ref19'> 19 </a> </xref>]</p><h3 class="blogheading Main-Title"><a href="javascript:void(0);" name="CONCLUSION">CONCLUSION</a></h3><div class="clearfix"></div><div class="hline"></div><p>Abscess formation within metastases may be easily misdiagnosed, as it does not present characteristic clinical and radiological features. Therefore, it is often an unexpected intraoperative finding. Despite being a rare event, this entity should be recognized to optimize disease management.</p><p></p><h3 class="blogheading Main-Title"><a href="javascript:void(0);" name="Ethical approval: ">Ethical approval: </a></h3><div class="clearfix"></div><div class="hline"></div><p>Institutional Review Board approval is not required.</p><h3 class="blogheading Main-Title"><a href="javascript:void(0);" name="Declaration of patient consent: ">Declaration of patient consent: </a></h3><div class="clearfix"></div><div class="hline"></div><p>Patient’s consent not required as patient’s identity is not disclosed or compromised.</p><h3 class="blogheading Main-Title"><a href="javascript:void(0);" name="Financial support and sponsorship: ">Financial support and sponsorship: </a></h3><div class="clearfix"></div><div class="hline"></div><p>Nil.</p><h3 class="blogheading Main-Title"><a href="javascript:void(0);" name="Conflicts of interest: ">Conflicts of interest: </a></h3><div class="clearfix"></div><div class="hline"></div><p>There are no conflicts of interest.</p><h3 class="blogheading Main-Title"><a href="javascript:void(0);" name="Use of artificial intelligence (AI)-assisted technology for manuscript preparation: ">Use of artificial intelligence (AI)-assisted technology for manuscript preparation: </a></h3><div class="clearfix"></div><div class="hline"></div><p>The authors confirm that there was no use of artificial intelligence (AI)-assisted technology for assisting in the writing or editing of the manuscript and no images were manipulated using AI.</p><h3 class="blogheading Main-Title"><a href="javascript:void(0);" name="Disclaimer">Disclaimer</a></h3><div class="clearfix"></div><div class="hline"></div><p>The views and opinions expressed in this article are those of the authors and do not necessarily reflect the official policy or position of the Journal or its management. The information contained in this article should not be considered to be medical advice; patients should consult their own physicians for advice as to their specific medical needs.</p></div> </div></div><div><div class="row"> <div class="blogparagraph col-lg-9 col-sm-8 col-xs-12"></div> </div><div class="row"> <div class="blogparagraph col-lg-9 col-sm-8 col-xs-12"> <h3 class="blogheading pull-left Main-Title"><a name="References" href="javascript:void(0);">References</a></h3> <div class="clearfix"></div> <div class="hline"></div> <p><a href='javascript:void(0);' name='ref1' style='text-decoration: none;'>1.</a> Ahmed H, Khan A, Abdul Rauf S, Somro J, Saleem SE, Parvez J. Cerebellar abscess secondary to metastatic lung adenocarcinoma: A case report. J Med Case Rep. 2024. 18: 389</p><p><a href='javascript:void(0);' name='ref2' style='text-decoration: none;'>2.</a> Chiang IC, Hsieh TJ, Chiu ML, Liu GC, Kuo YT, Lin WC. Distinction between pyogenic brain abscess and necrotic brain tumour using 3-tesla MR spectroscopy, diffusion and perfusion imaging. Br J Radiol. 2009. 82: 813-20</p><p><a href='javascript:void(0);' name='ref3' style='text-decoration: none;'>3.</a> Gavrilovic IT, Posner JB. Brain metastases: Epidemiology and pathophysiology. J Neurooncol. 2005. 75: 5-14</p><p><a href='javascript:void(0);' name='ref4' style='text-decoration: none;'>4.</a> Goto Y, Ebisu T, Mineura K. Abscess formation within a cerebellar metastasis: Case report and literature review. Int J Surg Case Rep. 2015. 10: 59-64</p><p><a href='javascript:void(0);' name='ref5' style='text-decoration: none;'>5.</a> Grinberg-Rashi H, Ofek E, Perelman M, Skarda J, Yaron P, Hajdúch M. The expression of three genes in primary non-small cell lung cancer is associated with metastatic spread to the brain. Clin Cancer Res. 2009. 15: 1755-61</p><p><a href='javascript:void(0);' name='ref6' style='text-decoration: none;'>6.</a> Hatiboglu MA, Iplikcioglu AC, Ozcan D. Abscess formation within invasive pituitary adenoma. J Clin Neurosci. 2006. 13: 774-7</p><p><a href='javascript:void(0);' name='ref7' style='text-decoration: none;'>7.</a> Hwang YF, Huang TY, Hwang SL, Kwan AL, Howng SL. Differentiation among metastatic brain tumors, radiation necroses, and brain abscesses using proton magnetic resonance spectroscopy. Kaohsiung J Med Sci. 2004. 20: 437-42</p><p><a href='javascript:void(0);' name='ref8' style='text-decoration: none;'>8.</a> Ichikawa M, Shimizu Y, Sato M, Imataka K, Masuda A, Hara Y. Abscess within a glioblastoma multiforme--case report. Neurol Med Chir (Tokyo). 1992. 32: 829-33</p><p><a href='javascript:void(0);' name='ref9' style='text-decoration: none;'>9.</a> Israel ZH, Yacoub M, Gomori JM, Dotan S, Fellig S, Shoshan Y. Rathke’s cleft cyst abscess. Pediatr Neurosurg. 2000. 33: 159-61</p><p><a href='javascript:void(0);' name='ref10' style='text-decoration: none;'>10.</a> Kano M, Watanabe M, Maeda M. Primary CNS lymphoma associated with streptococcal abscess: An autopsy case. Brain Tumor Pathol. 1999. 16: 92-7</p><p><a href='javascript:void(0);' name='ref11' style='text-decoration: none;'>11.</a> Kovacic S, Bunc G, Krajnc I. Abscess formation within cerebellar metastatic carcinoma--report of two cases and review of the literature. Wien Klin Wochenschr. 2004. 116: 60-3</p><p><a href='javascript:void(0);' name='ref12' style='text-decoration: none;'>12.</a> Kroppenstedt SN, Liebig T, Mueller W, Gräf KJ, Lanksch WR, Unterberg AW. Secondary abscess formation in pituitary adenoma after tooth extraction. Case report. J Neurosurg. 2001. 94: 335-8</p><p><a href='javascript:void(0);' name='ref13' style='text-decoration: none;'>13.</a> Kuge A, Sato S, Takemura S, Sakurada K, Kondo R, Kayama T. Abscess formation associated with pituitary adenoma: A case report: Changes in the MRI appearance of pituitary adenoma before and after abscess formation. Surg Neurol Int. 2011. 2: 3</p><p><a href='javascript:void(0);' name='ref14' style='text-decoration: none;'>14.</a> Lo WB, Cahill J, Carey M, Mehta H, Shad A. Infected intracranial meningiomas. World Neurosurg. 2014. 81: 651.e9-13</p><p><a href='javascript:void(0);' name='ref15' style='text-decoration: none;'>15.</a> Lu CH, Chang WN, Lin YC, Tsai NW, Liliang PC TM. Bacterial brain abscess: Microbiological features, epidemiological trends and therapeuPc outcomes. QJM. 2002. 95: 501-9</p><p><a href='javascript:void(0);' name='ref16' style='text-decoration: none;'>16.</a> Moiyadi A, Shetty P. Abscess in a metastasis. J Neurosurg. 2010. 112: 474-5</p><p><a href='javascript:void(0);' name='ref17' style='text-decoration: none;'>17.</a> Nassar SI, Haddad FS, Hanbali FS, Kanaan NV. Abscess superimposed on brain tumor: Two case reports and review of the literature. Surg Neurol. 1997. 47: 484-8</p><p><a href='javascript:void(0);' name='ref18' style='text-decoration: none;'>18.</a> Nathoo N, Chahlavi A, Barnett GH, Toms SA. Pathobiology of brain metastases. J Clin Pathol. 2005. 58: 237-42</p><p><a href='javascript:void(0);' name='ref19' style='text-decoration: none;'>19.</a> Ng WP, Lozano A. Abscess within a brain metastasis. Can J Neurol Sci. 1996. 23: 300-2</p><p><a href='javascript:void(0);' name='ref20' style='text-decoration: none;'>20.</a> Ohara-Nemoto Y, Haraga H, Kimura S, Nemoto TK. Occurrence of staphylococci in the oral cavities of healthy adults and nasal oral trafficking of the bacteria. J Med Microbiol. 2008. 57: 95-9</p><p><a href='javascript:void(0);' name='ref21' style='text-decoration: none;'>21.</a> Oommen A, Johny M, Pratap T, Paul RS, Chinnan JP, Jalal MJ. Listeria monocytogenes brain abscess within a metastatic intracerebellar space-occupying lesion in a patient with carcinoma lung: First case report from India. Indian J Neurosurg. 2017. 6: 129-34</p><p><a href='javascript:void(0);' name='ref22' style='text-decoration: none;'>22.</a> Robertson D, Smith AJ. The microbiology of the acute dental abscess. J Med Microbiol. 2009. 58: 155-62</p><p><a href='javascript:void(0);' name='ref23' style='text-decoration: none;'>23.</a> Rodriguez RE, Valero V, Watanakunakorn C. Salmonella focal intracranial infections: Review of the world literature (1884-1984) and report of an unusual case. Rev Infect Dis. 1986. 8: 31-41</p><p><a href='javascript:void(0);' name='ref24' style='text-decoration: none;'>24.</a> Shimomura T, Hori S, Kasai N, Tsuruta K, Okada H. Meningioma associated with intratumoral abscess formation--case report. Neurol Med Chir (Tokyo). 1994. 34: 440-3</p><p><a href='javascript:void(0);' name='ref25' style='text-decoration: none;'>25.</a> Sperduto PW, Yang TJ, Beal K, Pan H, Brown PD, Bangdiwala A. Estimating survival in patients with lung cancer and brain metastases: An update of the graded prognostic assessment for lung cancer using molecular markers (Lung-molGPA). JAMA Oncol. 2017. 3: 827-31</p><p><a href='javascript:void(0);' name='ref26' style='text-decoration: none;'>26.</a> Subramanian A, Harris A, Piggott K, Shieff C, Bradford R. Metastasis to and from the central nervous system--the’relatively protected site’. Lancet Oncol. 2002. 3: 498-507</p><p><a href='javascript:void(0);' name='ref27' style='text-decoration: none;'>27.</a> Takayasu T, Yamasaki F, Shishido T, Takano M, Maruyama H, Sugiyama K. Abscess formation in metastatic brain tumor with history of immune checkpoint inhibitor: A case report. NMC Case Rep J. 2019. 6: 11-5</p><p><a href='javascript:void(0);' name='ref28' style='text-decoration: none;'>28.</a> Tsai TH, Hwang YF, Hwang SL, Hung CH, Chu CW, Lua BK. Low-grade astrocytoma associated with abscess formation: Case report and literature review. Kaohsiung J Med Sci. 2008. 24: 262-9</p><p><a href='javascript:void(0);' name='ref29' style='text-decoration: none;'>29.</a> Tsugu A, Osada T, Nishiyama J, Matsumae M. Glioblastoma associated with intratumoral abscess formation. Case report. Neurol Med Chir (Tokyo). 2012. 52: 99-102</p><p><a href='javascript:void(0);' name='ref30' style='text-decoration: none;'>30.</a> Wang Y, Chen R, Wa Y, Ding S, Yang Y, Liao J. Tumor immune microenvironment and immunotherapy in brain metastasis from non-small cell lung cancer. Front Immunol. 2022. 13: 829451</p></div> </div></div>
  3. Viral immunological complications in neurological surgery: A comprehensive review of homeostatic disturbances and cognitive impairments

    Fri, 13 Jun 2025 16:54:25 -0000

    Viral immunological complications in neurological surgery: A comprehensive review of homeostatic disturbances and cognitive impairments Category: Article Type: Maral Moafi1, Rasa Zafari1, Kamyab Rabiee2, Mohammad Javad Ebrahimi1, Homa Seyedmirzaei3, Alireza Soltani Khaboushan1Department of Neurosurgery, Tehran University of Medical Sciences, Tehran, IranDepartment of Neurology, Shahroud University of Medical Sciences, Shahroud University of Medical Sciences and Health … Continue reading Viral immunological complications in neurological surgery: A comprehensive review of homeostatic disturbances and cognitive impairments
    <div><!DOCTYPE html PUBLIC "-//W3C//DTD HTML 4.0 Transitional//EN" "http://www.w3.org/TR/REC-html40/loose.dtd"> <html><head><meta http-equiv="content-type" content="text/html; charset=utf-8"></head><body><div class="row"><div class="col-lg-9 col-sm-8 col-xs-12"><div class="media-body details-body"> <a href="https://surgicalneurologyint.com/?post_type=surgicalint_articles&p=13627"><h2 class="media-heading"><h2 class="media-heading">Viral immunological complications in neurological surgery: A comprehensive review of homeostatic disturbances and cognitive impairments</h2></h2></a> </div><div class="disp_categories"> <p><label>Category: </label><span></span></p> <p><label>Article Type: </label><span></span></p> </div><a href="mailto:maral.moafy@yahoo.com" target="_top">Maral Moafi</a><sup>1</sup>, <a href="mailto:rasazafari22@gmail.com" target="_top">Rasa Zafari</a><sup>1</sup>, <a href="mailto:rabiee.kamyab@gmail.com" target="_top">Kamyab Rabiee</a><sup>2</sup>, <a href="mailto:ebrahimi137377@gmail.com" target="_top">Mohammad Javad Ebrahimi</a><sup>1</sup>, <a href="mailto:homa_sdmr@yahoo.com" target="_top">Homa Seyedmirzaei</a><sup>3</sup>, <a href="mailto:alirezasoltanykhaboshan@gmail.com" target="_top">Alireza Soltani Khaboushan</a><sup>1</sup><ol class="smalllist"><li>Department of Neurosurgery, Tehran University of Medical Sciences, Tehran, Iran</li><li>Department of Neurology, Shahroud University of Medical Sciences, Shahroud University of Medical Sciences and Health Services, Shahroud, Iran</li><li>Sports Medicine Research Center, Neuroscience Institute, Tehran, Iran</li></ol><p><strong>Correspondence Address:</strong><br>Alireza Soltani Khaboushan, Department of Neurosurgery, Tehran University of Medical Sciences, Tehran, Iran.<br></p><p><strong>DOI:</strong>10.25259/SNI_337_2025</p>Copyright: © 2025 Surgical Neurology International This is an open-access article distributed under the terms of the Creative Commons Attribution-Non Commercial-Share Alike 4.0 License, which allows others to remix, transform, and build upon the work non-commercially, as long as the author is credited and the new creations are licensed under the identical terms.<div class="parablock"><p><strong>How to cite this article: </strong>Maral Moafi1, Rasa Zafari1, Kamyab Rabiee2, Mohammad Javad Ebrahimi1, Homa Seyedmirzaei3, Alireza Soltani Khaboushan1. Viral immunological complications in neurological surgery: A comprehensive review of homeostatic disturbances and cognitive impairments. 13-Jun-2025;16:241</p></div><div class="parablock"><p><strong>How to cite this URL: </strong>Maral Moafi1, Rasa Zafari1, Kamyab Rabiee2, Mohammad Javad Ebrahimi1, Homa Seyedmirzaei3, Alireza Soltani Khaboushan1. Viral immunological complications in neurological surgery: A comprehensive review of homeostatic disturbances and cognitive impairments. 13-Jun-2025;16:241. Available from: <a href="https://surgicalneurologyint.com/?post_type=surgicalint_articles&p=13627">https://surgicalneurologyint.com/?post_type=surgicalint_articles&p=13627</a></p></div> </div> <div class="col-lg-3 col-sm-4 col-xs-12"><div class="article-detail-sidebar"><div class="icon sidebar-icon clearfix add-readinglist-icon"><button id="bookmark-article" class="add-reading-list-article">Add to Reading List</button><button id="bookmark-remove-article" class="remove-reading-list-article">Remove from Reading List</button></div><div class="icon sidebar-icon clearfix"><a class="btn btn-link" target="_blank" type="button" id="OpenPdf" href="https://surgicalneurologyint.com/wp-content/uploads/2025/06/13627/SNI-16-241.pdf"><img decoding="async" src="https://i1.wp.com/surgicalneurologyint.com/wp-content/themes/surgicalint/images/pdf-icon.png?w=604&#038;ssl=1" class="no-popup" data-recalc-dims="1"></a><a target="_blank" href="javascript:void(0);" onclick="return PrintArticle();"><img decoding="async" src="https://i0.wp.com/surgicalneurologyint.com/wp-content/themes/surgicalint/images/file-icon.png?w=604&#038;ssl=1" class="no-popup" data-recalc-dims="1"></a><a class="btn btn-link" type="button" id="EmaiLPDF"><img decoding="async" src="https://i1.wp.com/surgicalneurologyint.com/wp-content/themes/surgicalint/images/mail-icon.png?w=604&#038;ssl=1" class="no-popup" data-recalc-dims="1"></a></div><div class="date"> <p>Date of Submission<br><span class="darkgray">03-Apr-2025</span></p> <p>Date of Acceptance<br><span class="darkgray">13-May-2025</span></p> <p>Date of Web Publication<br><span class="darkgray">13-Jun-2025</span></p> </div> </div></div> </div> <!--.row --><div class="row"> <div class="blogparagraph col-lg-9 col-sm-8 col-xs-12"> <h3 class="blogheading pull-left Main-Title"><a href="javascript:void(0);" name="Abstract">Abstract</a></h3> <div class="clearfix"></div> <div class="hline"></div> <p><strong>Background: </strong>Neurosurgical procedures are essential for treating various brain and spinal conditions, but they also carry the risk of infections, including viral infections. These infections can disrupt brain homeostasis, leading to cognitive impairments. During surgery, protective barriers like the blood-brain barrier (BBB) can be compromised, and cerebrospinal fluid may be exposed to pathogens. This makes the brain more susceptible to viral infections, which can trigger inflammation. Over time, this inflammation can have lasting effects on cognitive function, impacting the brain’s ability to maintain neural integrity.</p><p><strong>Methods: </strong>A review of the literature was performed using PubMed, Google Scholar, Scopus, and Web of Science from inception to January 2025. We focus on the impact of viral infections after neurosurgical procedures and how these infections lead to neuroinflammation.</p><p><strong>Results: </strong>Viral infections after neurosurgery activate neuroinflammatory responses, with microglia and astrocytes playing a key role. The release of cytokines such as tumor necrosis factor-alpha and interleukin-1 causes significant neuronal damage, impairing synaptic function and connectivity. This inflammatory process, combined with BBB disruption, leads to cognitive dysfunction both in the immediate postoperative period and in the long-term. Understanding these processes is essential for addressing cognitive decline in patients who have undergone neurosurgery.</p><p><strong>Conclusion: </strong>Viral infections following neurosurgery are a significant risk factor for cognitive decline. Neuroinflammation, especially when coupled with BBB disruption, contributes to both short-term and long-term cognitive impairments. This review highlights the need for targeted interventions to control inflammation and protect the BBB in the perioperative period. Future research focused on neuroprotective therapies, including anti-inflammatory agents and strategies to preserve BBB integrity, is critical for improving cognitive outcomes in neurosurgical patients.</p><p><strong>Keywords: </strong>Brain homeostasis, Cognitive decline, Neuroinflammation, Neurological surgery, Viral infection</p><p></p></div> </div></body></html> </div><div><div class="row"> <div class="blogparagraph col-lg-9 col-sm-8 col-xs-12"><p></p><p><inline-graphic xmlns:xlink="http://www.w3.org/1999/xlink" xlink:href="SNI-16-241-inline001.tif"/></p><p></p><h3 class="blogheading Main-Title"><a href="javascript:void(0);" name="INTRODUCTION">INTRODUCTION</a></h3><div class="clearfix"></div><div class="hline"></div><p>Neurosurgical interventions employed to address neurological disorders and urgent situations often encompass the creation of a burr hole, performing a craniotomy to gain access to the brain, doing a laminectomy to access the spinal cord, and implanting a cerebral shunt.[<xref ref-type="bibr" rid="ref42"> <a href='#ref42'>42</a> </xref>,<xref ref-type="bibr" rid="ref66"> <a href='#ref66'>66</a> </xref>] These procedures have the possibility of postoperative infections, which can substantially impact patient outcomes. The average occurrence of cerebral infection is about 6% following surgery, and it is a notable complication that occurs after craniotomies, especially in oncological patients who are already immunocompromised due to the disease and treatment. Postoperative cerebral infections are influenced by various risk factors, such as the length of the surgery, the form of incision, cerebrospinal fluid leaks, and secondary surgeries.[<xref ref-type="bibr" rid="ref111"> <a href='#ref111'>111</a> </xref>] The risk exists even with minimally invasive neurosurgical procedures, including deep brain stimulation, laser interstitial thermal therapy, and stereotactic electroencephalography. Moreover, factors such as usage of bevacizumab, cirrhosis, foreign body implantation, previous radiotherapy, and previous surgeries increase the likelihood of surgical site infections (SSIs) following craniotomy for neuro-oncologic disorders.[<xref ref-type="bibr" rid="ref52"> <a href='#ref52'>52</a> </xref>] Severe intraoperative hyperglycemia is linked to an increased likelihood of postoperative infections in patients who are having elective brain neurosurgical procedures.[<xref ref-type="bibr" rid="ref34"> <a href='#ref34'>34</a> </xref>,<xref ref-type="bibr" rid="ref47"> <a href='#ref47'>47</a> </xref>] These infections can result in inflammation of the nervous system, disruption of the blood-brain barrier (BBB), and disturbances in homeostasis, highlighting the significance of taking preventive measures and closely monitoring patients to achieve improved results after surgery.[<xref ref-type="bibr" rid="ref88"> <a href='#ref88'>88</a> </xref>,<xref ref-type="bibr" rid="ref101"> <a href='#ref101'>101</a> </xref>]</p><p>The central nervous system (CNS) is characterized as an immune privilege, which results in a usually severe condition when infected. Factors leading to the danger of the host defense play a significant role in establishing neurosurgical infections. CNS infections possess unique features that differentiate them from infections impacting other organs.[<xref ref-type="bibr" rid="ref29"> <a href='#ref29'>29</a> </xref>] First, CNS has a BBB that selectively allows certain drugs, such as antibiotics, to pass through. Second, the subarachnoid space within the CNS is uniform, enabling infections to spread continuously through the cerebrospinal fluid. Third, circulatory disturbances in the CNS can elevate intracranial pressure, resulting in reduced blood flow and potential damage to brain tissue due to venous and arterial infarctions. Last, cerebral edema caused by infection can increase intracranial pressure, leading to brain damage as the limited space within the skull cannot accommodate the increased volume.[<xref ref-type="bibr" rid="ref111"> <a href='#ref111'>111</a> </xref>]</p><p>A CNS infection can pose a significant risk to individuals with compromised immune systems, potentially leading to life-threatening consequences. For instance, CNS infections in cancer patients result in prolonged administration of antibiotics, supplementary surgical interventions, increased treatment expenses, and inferior treatment results. Moreover, the treatment of the primary ailment may be prolonged or postponed due to the continuous infection.[<xref ref-type="bibr" rid="ref111"> <a href='#ref111'>111</a> </xref>] Herein, we comprehensively review post-neurosurgical viral infections and their effect on brain homeostasis and cognitive function.</p><h3 class="blogheading Main-Title"><a href="javascript:void(0);" name="VIRAL CNS INFECTION">VIRAL CNS INFECTION</a></h3><div class="clearfix"></div><div class="hline"></div><p>The etiologies of CNS infections include bacteria, viruses, parasites, and fungi. Viral CNS infections are rare and typically lead to mild, self-limiting illness. However, these infections are highly significant due to their capacity to cause fatalities and neurological harm. Neural tissues are highly susceptible to disruptions in metabolic processes, often with incomplete recovery.[<xref ref-type="bibr" rid="ref29"> <a href='#ref29'>29</a> </xref>] Viruses can move from the original infection site to the CNS through the bloodstream (viremia) or the peripheral nervous system. Viruses can move through the bloodstream by attaching themselves to cells, such as monocytes, T cells, and B cells, or by existing freely in plasma.[<xref ref-type="bibr" rid="ref7"> <a href='#ref7'>7</a> </xref>,<xref ref-type="bibr" rid="ref56"> <a href='#ref56'>56</a> </xref>,<xref ref-type="bibr" rid="ref86"> <a href='#ref86'>86</a> </xref>]</p><p>For CNS protection, the innate immune system uses pattern recognition receptors (PRRs), located either in the cytosol or on the surface of different cells, and induces intracellular pathways. The induction of these signaling pathways mediates an antiviral response mainly regulated by type I interferon (IFN).[<xref ref-type="bibr" rid="ref43"> <a href='#ref43'>43</a> </xref>,<xref ref-type="bibr" rid="ref106"> <a href='#ref106'>106</a> </xref>,<xref ref-type="bibr" rid="ref123"> <a href='#ref123'>123</a> </xref>] Toll-like receptors (TLRs), retinoic acid-inducible gene I (RIG-1), NOD-like receptors (NLRs), and various cytokines, including interleukin (IL)-10 and transforming growth factor-beta (TGF-b), are also involved in CNS antiviral responses.[<xref ref-type="bibr" rid="ref35"> <a href='#ref35'>35</a> </xref>,<xref ref-type="bibr" rid="ref48"> <a href='#ref48'>48</a> </xref>,<xref ref-type="bibr" rid="ref86"> <a href='#ref86'>86</a> </xref>,<xref ref-type="bibr" rid="ref115"> <a href='#ref115'>115</a> </xref>]</p><p>Viruses can spread from the bloodstream to the CNS via infected T cells or monocytes, infection, and replication in the brain’s capillary endothelial cells, transcytosis without replication, increased BBB permeability, or cerebrospinal fluid (CSF). Viruses in the peripheral nervous system infect nerve fiber dendrites and axons, reproducing in neuron cytoplasm or nuclei. They spread through synapses, and the strong immune responses from microglia and astrocytes in the CNS are crucial for limiting infection and minimizing tissue damage.[<xref ref-type="bibr" rid="ref40"> <a href='#ref40'>40</a> </xref>,<xref ref-type="bibr" rid="ref43"> <a href='#ref43'>43</a> </xref>,<xref ref-type="bibr" rid="ref56"> <a href='#ref56'>56</a> </xref>,<xref ref-type="bibr" rid="ref75"> <a href='#ref75'>75</a> </xref>,<xref ref-type="bibr" rid="ref86"> <a href='#ref86'>86</a> </xref>] Early detection and intervention are crucial for improved outcomes. Gaining a better understanding of CNS viral infections and the immune system’s role will aid in developing effective treatments, particularly for chronic infections.[<xref ref-type="bibr" rid="ref40"> <a href='#ref40'>40</a> </xref>,<xref ref-type="bibr" rid="ref106"> <a href='#ref106'>106</a> </xref>]</p><h3 class="blogheading Main-Title"><a href="javascript:void(0);" name="POST-SURGICAL CNS INFECTIONS">POST-SURGICAL CNS INFECTIONS</a></h3><div class="clearfix"></div><div class="hline"></div><h3 class = "title3">Viral infection due to surgical procedure</h3><p>One major contributing cause to the development of postoperative cognitive decline (POCD) is the body’s inflammatory reaction, which can be triggered by a surgical procedure. Of course, it might be challenging to determine the source and the result of the interaction between inflammation and injury to brain tissue. Any tissue damage is usually followed by an inflammatory response, which aids in healing but can cause harm if excessively activated. Experimental research in the rat brain revealed that following subarachnoid hemorrhage[<xref ref-type="bibr" rid="ref20"> <a href='#ref20'> 20 </a> </xref>] and experimental ischemia in the anterior region of the brain,[<xref ref-type="bibr" rid="ref18"> <a href='#ref18'> 18 </a> </xref>] there was also an elevation of cell pathways linked to inflammation, including those embodied by the nuclear transcription factor kB (NF-kB).</p><p>Cerebral inflammation and localized inflammatory responses can lead to localized ischemia. Pre-existing brain diseases or surgical interventions can trigger systemic inflammatory reactions in the nervous system. Such inflammation can damage brain tissue and impair cognitive performance. This can result in a variety of clinical events, such as septic encephalopathy and delirium, with symptoms ranging from mild cognitive impairment to coma due to the suppression of electroencephalogram activity.[<xref ref-type="bibr" rid="ref123"> <a href='#ref123'> 123 </a> </xref>] Histological analysis of septic mice showed secondary neuronal degeneration, perivascular edema, and significant BBB disruption.[<xref ref-type="bibr" rid="ref20"> <a href='#ref20'> 20 </a> </xref>]</p><p>Endothelial swelling is the first response to acute hypoxia in endothelial cells, which may occur in neurosurgical procedures and anesthesia. Reperfusion and reoxygenation lead to the generation of active metabolites that mobilize neutrophils, causing them to aggregate with endothelial cells. This process results in microthrombi formation and the release of potent pressor chemicals, leading to capillary blockage and blood flow obstruction, even when major vessels have recovered, known as the no-reflow phenomenon.[<xref ref-type="bibr" rid="ref49"> <a href='#ref49'> 49 </a> </xref>,<xref ref-type="bibr" rid="ref59"> <a href='#ref59'> 59 </a> </xref>,<xref ref-type="bibr" rid="ref87"> <a href='#ref87'> 87 </a> </xref>] Neutrophils adhere to the capillary endothelium, causing tissue damage by releasing proteolytic enzymes, leukotrienes, cytokines, and free oxygen radicals. These substances activate or are cytotoxic to platelets, arterial walls, and polymorphonuclear cells. During the preoperative phase, these mechanisms contribute to general nervous system damage.[<xref ref-type="bibr" rid="ref1"> <a href='#ref1'> 1 </a> </xref>] Long-term postoperative CNS dysfunction or delayed recovery can result from the CNS reacting to systemic inflammatory mediators. Even a mild response, like fever, may lead to diminished nervous system function and cognitive impairment.[<xref ref-type="bibr" rid="ref74"> <a href='#ref74'> 74 </a> </xref>]</p><p>Ninety percent of healthy, non-inflammatory CSF cells are T cells, primarily consisting of helper CD4 cells, regulatory CD4 cells, and CD8 cytotoxic T cells.[<xref ref-type="bibr" rid="ref44"> <a href='#ref44'> 44 </a> </xref>] T cells can enter the brain parenchyma in a pathogenic state. T cells have been implicated in viral and autoimmune neurological disorders, including herpes simplex encephalitis (HSVE) and multiple sclerosis.[<xref ref-type="bibr" rid="ref57"> <a href='#ref57'> 57 </a> </xref>] HSVE often results from a recurrence of a previous infection rather than a new infection during surgery. This reactivation can occur in two ways: the virus may reactivate in the trigeminal ganglion and travel to the CNS, or it may reactivate directly within the CNS where it has remained latent. Viral DNA can be found in the brains of individuals without neurological diseases, which may explain some HSVE cases without a clear prior history.[<xref ref-type="bibr" rid="ref21"> <a href='#ref21'> 21 </a> </xref>,<xref ref-type="bibr" rid="ref33"> <a href='#ref33'> 33 </a> </xref>] T-cell functions in neurodegenerative illnesses, including Parkinson’s Disease, have drawn a lot of attention lately.[<xref ref-type="bibr" rid="ref8"> <a href='#ref8'> 8 </a> </xref>] There is no concrete evidence linking T cells to the pathogenic phase of POCD. One study found that after surgery-induced cognitive impairment in mice, interleukin (IL)-17 was upregulated while IL-10 was downregulated in T helper cells and regulatory T cells, respectively.[<xref ref-type="bibr" rid="ref107"> <a href='#ref107'> 107 </a> </xref>] An imbalance in T-cell subtypes may exacerbate POCD. More research is required to understand how T cells contribute to POCD.[<xref ref-type="bibr" rid="ref60"> <a href='#ref60'> 60 </a> </xref>] <xref ref-type="table" rid="T1"> <a href='#T1'> Table 1 </a> </xref> is a detailed about common post-neurosurgical viral infections.</p><div class="row"> <div class="col-xs-12 content-figure col-wrap"> <div class="col-xs-2 figure-body col"><a href='javascript:void(0);' name='T1'></a> <br /><img src='https://i0.wp.com/surgicalneurologyint.com/wp-content/uploads/2025/06/13627/SNI-16-241-t001.png?w=604&#038;ssl=1' data-recalc-dims="1" /></div><div class="col-xs-10 col"> <div class="figure-content"><h3>Table 1:</h3><p>Overview of common post-neurosurgical viral infections.</p></div> </div> </div> </div><div class="clearfix">&nbsp;</div><h3 class = "title3">BBB breakdown</h3><p>The glial membrane, which comprised capillary endothelial cells, capillary basement membrane, and the terminal foot of astrocytes, generally comprised most of the BBB.[<xref ref-type="bibr" rid="ref84"> <a href='#ref84'> 84 </a> </xref>] Due to this tight structure, only gases, water, and tiny fat-soluble molecules could passively spread across the BBB. Proinflammatory cytokines such as tumor necrosis factor-alpha (TNF-a) and IL-1, however, can increase cyclooxygenase-2 in neurovascular endothelial cells, enhancing local prostaglandin synthesis and increasing BBB permeability.[<xref ref-type="bibr" rid="ref24"> <a href='#ref24'> 24 </a> </xref>] In addition, TNF-a increased matrix metalloproteinase (MMP) transcription, particularly MMP-9, further breaking down the BBB and extracellular matrix proteins.[<xref ref-type="bibr" rid="ref84"> <a href='#ref84'> 84 </a> </xref>] In response to surgical trauma, MMP-9 deletion mice exhibited better cognitive performance under fear-related conditions than wild-type mice. In these mice, Monocyte chemoattractant protein-1 (MCP-1) attracted brain monocyte-derived macrophages (BMDMs) to the CNS during inflammation and BBB disruption. On CNS entry, BMDMs increased NF-kB transcription, releasing proinflammatory cytokines[<xref ref-type="bibr" rid="ref91"> <a href='#ref91'> 91 </a> </xref>] and triggering microglia cells to intensify the neuroinflammatory response. Preoperative BMDM depletion decreased the incidence of POCD in mice models.[<xref ref-type="bibr" rid="ref22"> <a href='#ref22'> 22 </a> </xref>] The migration of BMDM may significantly contribute to POCD. Once the BBB is compromised, cytokines can more easily enter the CNS, promoting BMDM transport into neural tissues and triggering immune dysregulation. This connection between the peripheral and CNS’s immune systems can worsen neuroinflammation, harm brain tissue, and lead to POCD.[<xref ref-type="bibr" rid="ref105"> <a href='#ref105'> 105 </a> </xref>]</p><h3 class = "title3">The meningitis</h3><p>It is well established that HSV-2 is the primary cause of aseptic meningitis, while HSV-1 is associated with encephalitis. Notably, over 15% of individuals with HSV-2 infections in the CNS present with encephalitis instead of meningitis due to a significant overlap between the two conditions.[<xref ref-type="bibr" rid="ref70"> <a href='#ref70'> 70 </a> </xref>] The potential for aseptic inflammatory meningitis to arise from surgery can complicate the interpretation of CSF results in a postoperative setting. Although brain biopsy was previously considered the most effective method for definitively diagnosing HSVE, testing for HSV-1 or HSV-2 using polymerase chain reaction (PCR) on CSF is now recognized as more effective, boasting a sensitivity of over 95% and a specificity of more than 99%.[<xref ref-type="bibr" rid="ref108"> <a href='#ref108'> 108 </a> </xref>]</p><h3 class = "title3">Brain and subdural abscesses</h3><p>Brain abscesses are regarded as infections that could be fatal. In the 1980s, reports stated that the death rate from brain abscesses might reach 40%. The death rate for patients with brain abscesses has decreased as a result of improvements in radiographic scanning, the creation of innovative surgical procedures, and the accessibility of more recent antibiotics.[<xref ref-type="bibr" rid="ref11"> <a href='#ref11'> 11 </a> </xref>]</p><p>The signs and symptoms of a growing infection can be challenging to identify, particularly in the case of postoperative brain abscesses due to the neurologic changes brought on by the underlying disease and the rehabilitation process following brain surgery. Headaches, seizures, cranial nerve palsies, and behavioral abnormalities are the most common clinical signs of brain abscesses (primary and postoperative). Less frequently, fever and altered consciousness occur.[<xref ref-type="bibr" rid="ref51"> <a href='#ref51'> 51 </a> </xref>]</p><h3 class="blogheading Main-Title"><a href="javascript:void(0);" name="IMMUNE RESPONSE IN THE CNS">IMMUNE RESPONSE IN THE CNS</a></h3><div class="clearfix"></div><div class="hline"></div><h3 class = "title3">Mechanisms of viral entry and neuroinvasion</h3><p>Viruses use several methods to enter brain tissue. One such method is passive diffusion and endothelial cell infection, which enables viruses to breach the BBB by disrupting endothelial impermeability and releasing viral proteins into the bloodstream, as seen in flaviviruses.[<xref ref-type="bibr" rid="ref4"> <a href='#ref4'> 4 </a> </xref>] The other way of virus neuroinvasion is virus transcytosis, through which viral particles are absorbed by endothelial cells and transported to the tissue side of the brain.[<xref ref-type="bibr" rid="ref4"> <a href='#ref4'> 4 </a> </xref>] It is reported that the virus transcytosis method is seen in dengue virus infection.[<xref ref-type="bibr" rid="ref13"> <a href='#ref13'> 13 </a> </xref>] The last entry mechanism of viruses into the brain tissue is cell-associated virus transport. In this method, viruses are carried by blood cells and undergo blood-to-tissue transmigration by endothelial cells. This means of neuroinvasion is shown to be used by the Visna virus, bacteria, and even parasites.[<xref ref-type="bibr" rid="ref16"> <a href='#ref16'> 16 </a> </xref>,<xref ref-type="bibr" rid="ref68"> <a href='#ref68'> 68 </a> </xref>,<xref ref-type="bibr" rid="ref78"> <a href='#ref78'> 78 </a> </xref>,<xref ref-type="bibr" rid="ref89"> <a href='#ref89'> 89 </a> </xref>]</p><h3 class = "title3">Innate immune response and neuroinflammation</h3><p>The innate immune system is considered the first line of defense against microorganisms and foreign bodies.[<xref ref-type="bibr" rid="ref103"> <a href='#ref103'> 103 </a> </xref>] This part of the immune system consists of several components, including epithelium cells acting as a physical barrier, TLRs, antimicrobial enzymes, and particular defending cells in each tissue.[<xref ref-type="bibr" rid="ref71"> <a href='#ref71'> 71 </a> </xref>] In the CNS, microglia and astrocytes are the essential parts of the innate immune system unique to this tissue. Microglia cells, which originate from the myeloid progenitors, monitor the brain tissue when they are in their resting state.[<xref ref-type="bibr" rid="ref73"> <a href='#ref73'> 73 </a> </xref>] These cells can be activated following neuroinflammation through TLR activation and then release different cytokines, especially TNF-a, which exacerbates microglia activity within the CNS.[<xref ref-type="bibr" rid="ref112"> <a href='#ref112'> 112 </a> </xref>] In addition, astrocytes are the other unique group of innate immune cells in the CNS, which various cytokines can stimulate from reactive microglia.[<xref ref-type="bibr" rid="ref99"> <a href='#ref99'> 99 </a> </xref>] Furthermore, it is shown that reactive astrogliosis can release different cytokines, such as TNF and IL-6, exaggerating neuroinflammation in the brain tissue.[<xref ref-type="bibr" rid="ref58"> <a href='#ref58'> 58 </a> </xref>,<xref ref-type="bibr" rid="ref109"> <a href='#ref109'> 109 </a> </xref>] The innate immune system plays a significant role in restricting invading pathogens and activating the adaptive immune system. However, its function is highly dependent on the secretion of cytokines and chemokines.</p><h3 class = "title3">Cytokine and chemokine release</h3><p>Cytokines and chemokines can improve and trigger the function of microglia and astrocytes.[<xref ref-type="bibr" rid="ref96"> <a href='#ref96'> 96 </a> </xref>] TNF-a is released by activated microglia, exacerbating the activity of innate immune cells within the brain tissue. Other cytokines released following neuroinflammation include IL-1 and IL-6, which promote the inflammatory process within the CNS.[<xref ref-type="bibr" rid="ref96"> <a href='#ref96'> 96 </a> </xref>] On the other hand, microglia also produce cytokines such as IL-10 and TGF-b to modulate the immune response, preventing excessive tissue damage.[<xref ref-type="bibr" rid="ref19"> <a href='#ref19'> 19 </a> </xref>,<xref ref-type="bibr" rid="ref61"> <a href='#ref61'> 61 </a> </xref>,<xref ref-type="bibr" rid="ref114"> <a href='#ref114'> 114 </a> </xref>] In addition, astrocytes also secrete particular chemokines and cytokines, including IL-1, IL-6, and IFN-gamma.[<xref ref-type="bibr" rid="ref59"> <a href='#ref59'> 59 </a> </xref>,<xref ref-type="bibr" rid="ref109"> <a href='#ref109'> 109 </a> </xref>] It is reported that following the surgery trauma, astrocytes release a particular chemokine named CCL2, which plays a key role in the stimulation of microglia cells.[<xref ref-type="bibr" rid="ref117"> <a href='#ref117'> 117 </a> </xref>] Therefore, particular cells in the innate immune system release different cytokines and chemokines in response to virus invasion through which they can enhance the function of the innate system itself and provoke the adaptive immune system.</p><h3 class = "title3">BBB disruption</h3><p>Beyond the activation of the innate immune system following surgery trauma, released cytokines and chemokines can cause disruption in BBB to help macrophages enter the CNS.[<xref ref-type="bibr" rid="ref64"> <a href='#ref64'> 64 </a> </xref>] It is reported that interruption of BBB can be directly due to anesthesia and surgery trauma weakening the junction of BBB endothelial cells.[<xref ref-type="bibr" rid="ref120"> <a href='#ref120'> 120 </a> </xref>] In addition, some studies demonstrated increased amounts of mast cells within the brain tissue after surgery, resulting in decreased BBB integrity through reducing tight junction proteins such as claudin-5.[<xref ref-type="bibr" rid="ref126"> <a href='#ref126'> 126 </a> </xref>] Disrupted BBB can also exacerbate neuroinflammation by losing integrity and admitting pathogens and inflammatory molecules to enter the brain tissue. BBB is the primary defense part of the brain, which is damaged by the neuroinflammation process triggered by the function of the innate immune system in response to neuroinvasion, posing the brain tissue to diverse pathogens and immune cells and proteins.[<xref ref-type="bibr" rid="ref104"> <a href='#ref104'> 104 </a> </xref>]</p><h3 class = "title3">Adaptive immune response</h3><h3 class = "title3" style="color:green;">T-cell activation and function</h3><p>The activation of the innate immune system may lead to the induction of the adaptive immune system through antigen presentation by dendritic cells, macrophages, and B cells to the naïve T cells, the main initiators of the adaptive system reaction.[<xref ref-type="bibr" rid="ref12"> <a href='#ref12'> 12 </a> </xref>,<xref ref-type="bibr" rid="ref27"> <a href='#ref27'> 27 </a> </xref>] In addition, all cells infected by viruses carry major histocompatibility complex (MHC) class I molecules, particularly molecules on their membranes, which trigger naïve T cells.[<xref ref-type="bibr" rid="ref12"> <a href='#ref12'> 12 </a> </xref>] Presenting of antigens to T cells within the CNS is carried out by microglia cells, which are the major antigen-presenting cells in the brain tissue.[<xref ref-type="bibr" rid="ref79"> <a href='#ref79'> 79 </a> </xref>,<xref ref-type="bibr" rid="ref100"> <a href='#ref100'> 100 </a> </xref>] Within the CNS, CD4+ T cells act as helper cells to enhance adaptive immune reactions, and CD8+ cytotoxic T cells function as the central part of the adaptive immune cells against pathogens within the brain tissue, targeting cells expressing MHC class I, including astrocytes, microglia, and infected neurons.[<xref ref-type="bibr" rid="ref41"> <a href='#ref41'> 41 </a> </xref>,<xref ref-type="bibr" rid="ref90"> <a href='#ref90'> 90 </a> </xref>,<xref ref-type="bibr" rid="ref100"> <a href='#ref100'> 100 </a> </xref>] CD8+ cytotoxic T cells utilize various methods to inhibit virus function within the CNS. These cells destroy infected neurons through apoptosis and eliminate infected microglia and astrocytes through the cytolysis process.[<xref ref-type="bibr" rid="ref51"> <a href='#ref51'> 51 </a> </xref>,<xref ref-type="bibr" rid="ref67"> <a href='#ref67'> 67 </a> </xref>,<xref ref-type="bibr" rid="ref76"> <a href='#ref76'> 76 </a> </xref>] T cells have the most important role in defending the CNS against viruses, whereas the collaboration of B cells and humoral immunity can increase the strength of the whole CNS immune system in destroying these pathogens.</p><h3 class = "title3" style="color:green;">Antibody production and humoral immunity</h3><p>B cells play a major role in humoral immunity by secreting antibodies and recognizing antigens.[<xref ref-type="bibr" rid="ref23"> <a href='#ref23'> 23 </a> </xref>] B cells have particular B-cell receptors, which bind to their specific antigens, resulting in the differentiation of B-cells into plasma cells and memory B-cells. Plasma cells are the main cells producing antibodies, including immunoglobulin (Ig)G, IgM, IgA, IgD, and IgE.[<xref ref-type="bibr" rid="ref94"> <a href='#ref94'> 94 </a> </xref>] In healthy conditions, B cells of CNS are found in the meninges, especially within the dura mater adjacent to its venous sinuses.[<xref ref-type="bibr" rid="ref92"> <a href='#ref92'> 92 </a> </xref>] In addition, it is reported that IgA+ plasma cells, which play a crucial role in CNS humoral immunity, can be found in dural venous sinuses.[<xref ref-type="bibr" rid="ref26"> <a href='#ref26'> 26 </a> </xref>] Once a pathogen enters the CNS, most of the antibody-secreting cells (ASCs) in early phases of humoral immunity activity are IgM+, and after a while, IgG<sup>+</sup> secreting cells become the major part of the ASCs.[<xref ref-type="bibr" rid="ref69"> <a href='#ref69'> 69 </a> </xref>] Secreted antibodies can neutralize various kinds of pathogens entering the brain tissue in two ways, including the stimulation of the proteins of the complement system and opsonization, the process through which antibodies coat the pathogen and induce the macrophages to eliminate it.[<xref ref-type="bibr" rid="ref85"> <a href='#ref85'> 85 </a> </xref>] Thus, interaction of the B cells and T cells as the humoral and adaptive immune system alongside the function of the innate immune system consisting of microglia, astrocytes, cytokines, and chemokines plays a core role in destroying viruses invade the CNS following a neurosurgical intervention.</p><h3 class="blogheading Main-Title"><a href="javascript:void(0);" name="POSTOPERATIVE VIRAL INFECTION AND COGNITION">POSTOPERATIVE VIRAL INFECTION AND COGNITION</a></h3><div class="clearfix"></div><div class="hline"></div><h3 class = "title3">Mechanisms of neuroinflammation, neuronal damage, and cognitive impairments brought on by viruses</h3><p>Surgery-induced systemic inflammation is found to raise plasma levels of TNF-a, IL-1, IL-6, and other inflammatory markers,[<xref ref-type="bibr" rid="ref80"> <a href='#ref80'> 80 </a> </xref>] which trigger the CNS inflammatory response through various pathways. Due to relative permeability and the absence of a continuous BBB, inflammatory substances enter the CNS through diffusion in the periventricular region. Some transporters may also actively carry inflammatory substances into the CNS within the intact BBB. In addition, in pathological circumstances, the BBB’s permeability is altered. The production of TNF-a during the perioperative systemic inflammatory response increases BBB permeability.[<xref ref-type="bibr" rid="ref127"> <a href='#ref127'> 127 </a> </xref>] Animal studies show that surgical trauma triggers peripheral TNF-a release, compromising the BBB. This leads to an influx of inflammatory cells, mainly macrophages, and factors into the CNS, provoking further inflammatory responses.[<xref ref-type="bibr" rid="ref15"> <a href='#ref15'> 15 </a> </xref>,<xref ref-type="bibr" rid="ref36"> <a href='#ref36'> 36 </a> </xref>] In addition, it was discovered that TNF-a antagonist therapy after inflammatory stimulation enhances cognitive function compared to the control group.[<xref ref-type="bibr" rid="ref6"> <a href='#ref6'> 6 </a> </xref>,<xref ref-type="bibr" rid="ref119"> <a href='#ref119'> 119 </a> </xref>] Peripheral inflammatory reactions involve the infiltration of inflammatory factors into the CNS, activating its immune response. Specifically, peripheral factors like IL-1 bind to receptors on BBB endothelial cells, prompting these cells to produce immunoreactive molecules that activate microglia and astrocytes in the CNS.[<xref ref-type="bibr" rid="ref6"> <a href='#ref6'> 6 </a> </xref>] When activated by external inflammatory stimuli, vagal afferent nerves quickly initiate central inflammatory pathways, leading to responses in the CNS. This activation triggers local gastrointestinal and cardiovascular reflexes, enhancing the body’s defense mechanisms. Immune cells respond to inflammation by releasing mediators that activate primary afferent neurons in the vagal sensory ganglion. These mediators bind to receptors on vagal afferent fibers, stimulating an immunological response in the CNS. Vagal sensory neurons can also express messenger RNAs for IL-1 and prostaglandin receptors.[<xref ref-type="bibr" rid="ref31"> <a href='#ref31'> 31 </a> </xref>] The term “gut-brain axis” refers to the relationship between gut microorganisms and the brain.[<xref ref-type="bibr" rid="ref98"> <a href='#ref98'> 98 </a> </xref>] The intestinal flora comprises many bacteria, viruses, other microbes, and the gut microbiota. Surprisingly, disruption of the gut microbiota led to the release of inflammatory markers, including TNF-a and IL-6, and the advancement of neurodegenerative disorders.[<xref ref-type="bibr" rid="ref121"> <a href='#ref121'> 121 </a> </xref>] An increasing amount of animal research has demonstrated in recent years that anesthesia and surgery can cause an imbalance in the gut microbiota, which can subsequently impact brain function through specific processes.[<xref ref-type="bibr" rid="ref125"> <a href='#ref125'> 125 </a> </xref>]</p><p>An increase in useful bacteria in the gut, such as Lactobacillus, Bifidobacterium, and Galactose oligosaccharide, may help to mitigate these pathogenic mechanisms.[<xref ref-type="bibr" rid="ref121"> <a href='#ref121'> 121 </a> </xref>] In addition, current studies have demonstrated that appropriate exercise reduced gut dysbiosis and elevated valeric acid, improving postoperative neuroplasticity and cognitive function.[<xref ref-type="bibr" rid="ref50"> <a href='#ref50'> 50 </a> </xref>] It is still unknown how the brain’s neurons and gut microbiota interacted with one another and how this influenced cognitive performance in the normal brain.[<xref ref-type="bibr" rid="ref14"> <a href='#ref14'> 14 </a> </xref>] It should be mentioned that the blood-cerebrospinal fluid and BBB, which separate the brain from the rest of the immune system and prevent it from being impacted by it, are essential to the CNS. However, Iliff <i>et al</i>.[<xref ref-type="bibr" rid="ref39"> <a href='#ref39'> 39 </a> </xref>] recently identified glymphatic pathways in the CNS. Louveau <i>et al</i>.[<xref ref-type="bibr" rid="ref62"> <a href='#ref62'> 62 </a> </xref>] demonstrated that meningeal lymphatic channels in the CNS interacted with the peripheral immune system. These results contributed to debunking the theory that the brain is an immune-privileged organ. <xref ref-type="fig" rid="F1"> <a href='#F1'> Figure 1 </a> </xref> depicts a summary of post-neurosurgical viral infection and following homeostasis disruption and cognitive decline.</p><div class="row"> <div class="col-xs-12 content-figure col-wrap"> <div class="col-xs-2 figure-body col"><a href='javascript:void(0);' name='F1'></a> <br /><img src='https://i0.wp.com/surgicalneurologyint.com/wp-content/uploads/2025/06/13627/SNI-16-241-g001.png?w=604&#038;ssl=1' data-recalc-dims="1" /></div><div class="col-xs-10 col"> <div class="figure-content"><h3>Figure 1:</h3><p>Viral immunological complications in neurological surgery. Procedures such as craniotomy and laminectomy increase the risk of CNS infections, allowing viral pathogens to infiltrate through CSF or BBB disruptions. This triggers microglial and astrocytic activation, releasing pro-inflammatory cytokines (TNF-α, IL-1), leading to potential POCD, memory loss, and neuronal damage. CNS: Central nervous system, CSF: cerebrospinal fluid, BBB: Blood-brain barrier, TNF-α: Tumor necrosis factor-alpha, IL-1: Interleukin 1, POCD: Postoperative cognitive dysfunction.</p></div> </div> </div> </div><div class="clearfix">&nbsp;</div><h3 class = "title3">Breakdown of the neuronal network and malfunctioning of synapses</h3><p>The density of synapses is reduced with age, although aging affects various brain parts differently.[<xref ref-type="bibr" rid="ref105"> <a href='#ref105'> 105 </a> </xref>] The prefrontal cortex and hippocampus showed more noticeable alterations than other brain regions.[<xref ref-type="bibr" rid="ref5"> <a href='#ref5'> 5 </a> </xref>,<xref ref-type="bibr" rid="ref9"> <a href='#ref9'> 9 </a> </xref>] Neurodegenerative illnesses result from malfunctioning synapses due to a lack of neuronal activity or cell death.[<xref ref-type="bibr" rid="ref9"> <a href='#ref9'> 9 </a> </xref>] Newly generated synapses underwent constant modification to meet their behavioral needs in a continuously changing environment.[<xref ref-type="bibr" rid="ref32"> <a href='#ref32'> 32 </a> </xref>] These alterations may occur in the development of new synapses or in synaptic plasticity, which is the improvement of synaptic efficacy.[<xref ref-type="bibr" rid="ref2"> <a href='#ref2'> 2 </a> </xref>] Inflammatory cytokines produced by surgical or anesthetic drugs cross the BBB, allowing them to enter the CNS and activate inflammatory cells. Damaged proteins, neurotoxins, and inflammatory cytokines are just a few pathological proteins that these inflammatory cells would eventually release. These proteins could interact with neurons and synapses, synaptic loss, causing neuronal death, and impaired cell signaling, ultimately resulting in POCD.[<xref ref-type="bibr" rid="ref37"> <a href='#ref37'> 37 </a> </xref>] There are two potential mechanisms: first, aggregations of phosphorylated Tau protein and synaptic terminal Ab plaques cause damage to the signaling function and the structure of neighboring neurons and synapses[<xref ref-type="bibr" rid="ref25"> <a href='#ref25'> 25 </a> </xref>] and second, intoxication may result from the production of TNF-a from glial cells and IL-16 from lymphocytes, which impede metabolism and cause an excessive build-up of glutamate.[<xref ref-type="bibr" rid="ref38"> <a href='#ref38'> 38 </a> </xref>,<xref ref-type="bibr" rid="ref97"> <a href='#ref97'> 97 </a> </xref>] Parvalbumin neurons, an important class of GABAergic inhibitory interneurons, were downregulated, leading to a reduction in inhibitory neurotransmission through GABA receptors. This change could disturb the balance between excitatory and inhibitory neurotransmission, resulting in lower neuronal excitability and decreased synaptic activity.[<xref ref-type="bibr" rid="ref82"> <a href='#ref82'> 82 </a> </xref>] After surgery or anesthesia, mitochondria-derived reactive oxygen species (ROS) are generated. These ROS not only functioned as upstream NOD-like receptor protein 3 (NLRP3) activators but also took part in the formation of downstream inflammasomes.[<xref ref-type="bibr" rid="ref113"> <a href='#ref113'> 113 </a> </xref>] Attacks by ROS were most likely to target mitochondria since they are the primary location of ROS production. Following oxidative damage, a vicious loop that ultimately resulted in nerve apoptosis was created when the mitochondrial respiratory chain was damaged.[<xref ref-type="bibr" rid="ref72"> <a href='#ref72'> 72 </a> </xref>] In general, the most important causes of POCD were neuronal injury and a decrease in synaptic plasticity. Future research will, therefore, concentrate on ways to intervene to lessen the loss of neuronal and synaptic function.[<xref ref-type="bibr" rid="ref105"> <a href='#ref105'> 105 </a> </xref>]</p><h3 class = "title3">Affected cognitive domains: Executive function, memory, and attention</h3><p>Brain damage brought on by neuronal apoptosis can lead patients to experience emotional disturbances, cognitive decline, memory loss, and reduced learning capacities. Age decreases the ability of adult brain neurons to mend themselves through a process called neuronal regeneration.[<xref ref-type="bibr" rid="ref110"> <a href='#ref110'> 110 </a> </xref>] Sun <i>et al</i>. found that Alzheimer’s disease (AD) mice’s hippocampus neurons had a much higher apoptotic index than the wild-type control group.[<xref ref-type="bibr" rid="ref102"> <a href='#ref102'> 102 </a> </xref>] Cognitive performance was improved, and hippocampus neuronal death was decreased in AD mice treated with dexmedetomidine.[<xref ref-type="bibr" rid="ref102"> <a href='#ref102'> 102 </a> </xref>] In their POCD model, Yuan <i>et al</i>. stated that autophagy and neuronal death are caused by activation of the NF-kB pathway in neurons, which leads to cognitive impairment.[<xref ref-type="bibr" rid="ref124"> <a href='#ref124'> 124 </a> </xref>] The experiment was conducted using elderly mice with the left extrahepatic lobe excised. Shao <i>et al</i>. used sevoflurane to create the POCD model. They found that through blocking the NLRP3/caspase-1 pathway, Chikusetsu saponin IVa (ChIV) has a neuroprotective effect against sevoflurane-induced neuroinflammation and cognitive impairment. According to Shao <i>et al</i>., this indicates that ChIV may be a viable clinical approach for treating older patients’ anesthesia-induced POCD.[<xref ref-type="bibr" rid="ref95"> <a href='#ref95'> 95 </a> </xref>] Disruption of synaptic plasticity refers to the independent modification of the strength of connections between nerve cell synapses. These changes can result in temporary modifications, such as inhibition, facilitation, and enhancement, as well as lasting alterations, including long-term potentiation and long-term inhibition. These processes form the neural basis for memory and learning mechanisms.[<xref ref-type="bibr" rid="ref65"> <a href='#ref65'> 65 </a> </xref>] The post-synaptic density protein 95, or PSD95, is essential for synaptic plasticity. Gao <i>et al</i>. found that insults from surgery and anesthesia decreased synaptophysin and PSD95 in the hippocampal regions of mice, which affected postoperative cognitive function.[<xref ref-type="bibr" rid="ref28"> <a href='#ref28'> 28 </a> </xref>] It was found that expression levels of histone deacetylase 3 were elevated in the dorsal hippocampus of an aged mouse model of POCD following exploratory laparotomy performed under anesthesia. This increase was associated with a significant decrease in dendritic spine density and in proteins related to synaptic plasticity. However, the cognitive impairment observed after surgery was restored in the dorsal hippocampus by specifically inhibiting histone deacetylase 3, which led to the recovery of dendritic spine density and the levels of proteins associated with synaptic plasticity.[<xref ref-type="bibr" rid="ref54"> <a href='#ref54'> 54 </a> </xref>,<xref ref-type="bibr" rid="ref118"> <a href='#ref118'> 118 </a> </xref>]</p><p>It was discovered that surgery caused an increase in the activity of histone deacetylase 2 and a decrease in dendritic arborization and spine density in the hippocampus of mice with POCD. The medically induced alterations were counteracted by administering suberanilohydroxamic acid by intraperitoneal injection. This compound is a highly specific inhibitor of histone deacetylase 2.[<xref ref-type="bibr" rid="ref63"> <a href='#ref63'> 63 </a> </xref>] Xiao <i>et al</i>. found that inhibiting prostaglandin E2-receptor 3 using a stereotaxic virus injection into the dorsal hippocampus restored plasticity-related proteins, including activity and cyclic adenosine monophosphate (cAMP) response element-binding protein-controlled cytoskeletal-associated protein.[<xref ref-type="bibr" rid="ref116"> <a href='#ref116'> 116 </a> </xref>] This intervention reduced the cognitive decline caused by surgery.[<xref ref-type="bibr" rid="ref116"> <a href='#ref116'> 116 </a> </xref>] <xref ref-type="table" rid="T2"> <a href='#T2'> Table 2 </a> </xref> explains a summary of how viral infections can lead to cognitive impairment and homeostatic disturbances after neurological surgery.</p><div class="row"> <div class="col-xs-12 content-figure col-wrap"> <div class="col-xs-2 figure-body col"><a href='javascript:void(0);' name='T2'></a> <br /><img src='https://i2.wp.com/surgicalneurologyint.com/wp-content/uploads/2025/06/13627/SNI-16-241-t002.png?w=604&#038;ssl=1' data-recalc-dims="1" /></div><div class="col-xs-10 col"> <div class="figure-content"><h3>Table 2:</h3><p>Mechanisms of viral-induced cognitive impairment and homeostatic disturbances.</p></div> </div> </div> </div><div class="clearfix">&nbsp;</div><h3 class="blogheading Main-Title"><a href="javascript:void(0);" name="PREVENTION AND MANAGEMENT STRATEGY">PREVENTION AND MANAGEMENT STRATEGY</a></h3><div class="clearfix"></div><div class="hline"></div><h3 class = "title3">Patient-related risk factors</h3><p>Certain patient-related factors increase the likelihood of viral infections following neurosurgery. These include being immunocompromised, whether due to underlying conditions such as untreated HIV, hematological malignancies, or the use of immunosuppressive therapies. In addition, a history of recent travel, contact with infected individuals, and behaviors such as injection drug use or unprotected sexual activity further elevate the risk. Use of steroids, exposure to radiotherapy, physical trauma, geographic location, the time of year, and vaccination history also influence susceptibility to viral reactivation or new infections. Recognizing these factors is crucial for identifying patients who may be at greater risk of developing complications, including cognitive decline after surgery.[<xref ref-type="bibr" rid="ref3"> <a href='#ref3'> 3 </a> </xref>,<xref ref-type="bibr" rid="ref46"> <a href='#ref46'> 46 </a> </xref>]</p><h3 class = "title3">Procedure-related risk factors</h3><p>Dural tears, laminectomy, and operation time >3 h have been identified as independent risk factors for bacterial meningitis following spinal surgery. Emergency surgeries, operations classified as clean-contaminated or dirty, procedures lasting more than 4 h, and recent neurosurgical interventions have been identified as independent predictive factors for developing SSIs. Despite the extensive data on bacterial complications, there is a significant gap in the literature regarding viral infections following neurosurgery.[<xref ref-type="bibr" rid="ref55"> <a href='#ref55'> 55 </a> </xref>,<xref ref-type="bibr" rid="ref77"> <a href='#ref77'> 77 </a> </xref>]</p><h3 class = "title3">Infection control protocols and antiviral prophylaxis</h3><p>HSV is the leading cause of sporadic encephalitis, a severe and potentially fatal condition with a high risk of long-term disability in survivors.[<xref ref-type="bibr" rid="ref83"> <a href='#ref83'> 83 </a> </xref>] The introduction of acyclovir treatment has significantly reduced mortality from around 70% to below 20%. Intravenous acyclovir is the preferred treatment for HSV encephalitis. The standard dose is 10 mg/kg every 8 h, typically for 14–21 days, with adjustments for kidney function as necessary. Studies indicate that delaying treatment beyond 48 h after admission increases the risk of long-term neurological complications. The choice between a 14-day and 21-day treatment duration depends on the severity of the condition. If the chance of HSV encephalitis is low and a CSF HSV PCR test performed over 72 h after symptom onset is negative, acyclovir can be discontinued. However, if there is a strong suspicion of HSV encephalitis, treatment should proceed despite a negative CSF HSV PCR, unless another diagnosis is established.[<xref ref-type="bibr" rid="ref108"> <a href='#ref108'> 108 </a> </xref>]</p><h3 class = "title3">Managing homeostatic disturbances and cognitive impairments</h3><p>The inflammation caused by HSV-1 can compromise the BBB, this is largely driven by high levels of cytokines such as IL-1b and TNF-a, which bind to ICAM-1 glycoproteins on brain endothelial cells. Mitochondrial dysfunction in astrocytes exacerbates these effects, causing cell death and altering aquaporin 4 (AQP4), a protein involved in fluid regulation, which leads to brain edema. HSV-1 triggers ongoing microglial activation, which results in significant neuroinflammation and the release of numerous cytokines and chemokines. Microglia and astrocytes recognize viral particles, known as pathogen-associated molecular patterns (PAMPs), through PRRs like TLRs, leading to an intense antiviral response. This response includes the production of inflammatory molecules such as TNF, IL-1, IFN-alpha, and IL-6. Microglia also produce chemokines and antimicrobial proteins, such as chemokine ligand 5 (CCL5), C-X-C motif chemokine ligand 10 (CXCL10), nitric oxide, and inducible nitric oxide synthase, which are essential in directing immune responses and controlling inflammation.[<xref ref-type="bibr" rid="ref10"> <a href='#ref10'> 10 </a> </xref>,<xref ref-type="bibr" rid="ref17"> <a href='#ref17'> 17 </a> </xref>,<xref ref-type="bibr" rid="ref30"> <a href='#ref30'> 30 </a> </xref>] HSV-1 infection disrupts normal neuronal activity, leading to increased excitability, calcium imbalance, and heightened production of amyloid precursor protein, amyloid plaques, and hyper phosphorylated tau–hallmarks of AD.[<xref ref-type="bibr" rid="ref81"> <a href='#ref81'> 81 </a> </xref>] Markers of HSV-1 infection, such as anti-HSV-1 IgM antibodies, have been linked to a greater risk of developing AD.[<xref ref-type="bibr" rid="ref53"> <a href='#ref53'> 53 </a> </xref>] Mitochondrial dysfunction and oxidative stress contribute to neuronal apoptosis, while pro-inflammatory cytokines exacerbate neurotoxicity. This cycle of neuroinflammation and cellular damage emphasizes the importance of managing homeostatic disturbances to prevent cognitive decline over the long-term.</p><h3 class = "title3">Novel antiviral therapies and immunomodulatory agents</h3><p>Among all strategies for controlling viral infections after neurosurgical procedures, there are novel therapies yet to be considered; monoclonal antibodies are engineered proteins designated to target specific antigens. They can affect the proteins that are essential for the replication of virus thereby reducing the overall viral load.[<xref ref-type="bibr" rid="ref45"> <a href='#ref45'> 45 </a> </xref>] Clustered Regularly Interspaced Short Palindromic Repeats (CRISPR) is a revolutionary gene-editing technology that allows scientists to make precise changes to DNA. It is based on a naturally occurring defense mechanism found in bacteria and archaea, which use CRISPR to protect themselves from viruses and other foreign genetic elements. CRISPR-based antiviral therapies are currently in the early stages of clinical development. Ongoing research is focused on optimizing delivery methods, improving specificity, and evaluating the safety and efficacy of these therapies in humans. CRISPRCas9 can be programmed to target specific DNA sequences, including those found within viral genomes. By targeting essential viral genes, researchers can disrupt viral replication, assembly, or entry processes, effectively eliminating infected cells and preventing the spread of the virus.[<xref ref-type="bibr" rid="ref122"> <a href='#ref122'> 122 </a> </xref>]</p><h3 class = "title3">Long-term Follow-up Studies and Patient Outcomes</h3><p>Even mild forms of viral meningoencephalitis can cause permanent long-term sequelae, and the severity of the disease during the acute phase has a significant impact on the long-term outcome. A significant proportion of survivors from acute encephalitis experience long-term effects, including memory problems, headaches, fatigue, and mood disorders. The severity of these challenges can vary widely, and some survivors may require ongoing support and resources to manage their symptoms and improve their quality of life.[<xref ref-type="bibr" rid="ref93"> <a href='#ref93'> 93 </a> </xref>]</p><h3 class="blogheading Main-Title"><a href="javascript:void(0);" name="DISCUSSION">DISCUSSION</a></h3><div class="clearfix"></div><div class="hline"></div><p>Viral infections that occur after neurosurgical procedures can significantly impact CNS homeostasis and cognitive function. After surgery, disruptions in the BBB and CSF pathways can facilitate viral infiltration. This infiltration triggers neuroinflammatory processes involving microglia and astrocytes. The release of cytokines, such as TNF-a and IL-1, promotes neuroinflammation, disrupts synaptic connections, and causes neuronal damage, all of which can lead to cognitive decline. While previous studies have highlighted the role of neuroinflammation in cognitive impairment, this review specifically examines these mechanisms within the context of neurosurgery, emphasizing the brain’s increased vulnerability to viral pathogens following invasive procedures.</p><h3 class = "title3">Limitations</h3><p>The existing literature often lacks comprehensive, surgery-specific data on viral infections and neuroinflammation. Variations in patient immune responses, surgical techniques, and post-operative care can lead to differing outcomes, making it challenging to generalize findings. Future research should aim to incorporate longitudinal data and standardized metrics for measuring neuroinflammation and cognitive function post-surgery. This approach would strengthen the interpretation of results and help establish causal links. A better understanding of neuroimmunology enhances our awareness of the risks associated with viral infections in neurosurgical settings. One important goal is to preserve brain homeostasis during and after neurosurgery, alongside the need for improved infection control measures.</p><h3 class = "title3">Future direction</h3><p>Future research should focus on developing neuroprotective interventions, such as anti-inflammatory agents or compounds that stabilize the BBB, to reduce the risk of CNS infections and neuroinflammation in the perioperative period. In addition, more studies are needed to investigate patient-specific factors, such as genetic predispositions or variations in immune response, which may influence susceptibility to POCD following neurosurgery. Finally, clinical trials that examine the effectiveness of preventive measures in diverse patient populations could provide crucial insights for tailored strategies to mitigate the cognitive impacts of viral infections in neurosurgical contexts.</p><h3 class="blogheading Main-Title"><a href="javascript:void(0);" name="CONCLUSION">CONCLUSION</a></h3><div class="clearfix"></div><div class="hline"></div><p>Neurosurgical procedures can increase susceptibility to CNS viral infections, which may disrupt brain homeostasis and contribute to cognitive decline through neuroinflammatory mechanisms. Understanding these pathways and the associated risks offers new perspectives on post-surgical care and points to potential interventions for safeguarding cognitive health. Enhanced infection control, combined with future neuroprotective therapies, holds promise for minimizing the cognitive impacts of neurosurgery and improving long-term patient outcomes.</p><p></p><h3 class="blogheading Main-Title"><a href="javascript:void(0);" name="Authors’ Contributions: ">Authors’ Contributions: </a></h3><div class="clearfix"></div><div class="hline"></div><p>MM, RZ, MJE, and HS prepared the first draft, revised the manuscript, and designed the table and figures. ASK conceptualized the title, edited and finalized the draft, critically revised the manuscript, and supervised the project. All the authors have read and approved the final draft of the manuscript.</p><h3 class="blogheading Main-Title"><a href="javascript:void(0);" name="Ethical approval: ">Ethical approval: </a></h3><div class="clearfix"></div><div class="hline"></div><p>The Institutional Review Board approval is not required.</p><h3 class="blogheading Main-Title"><a href="javascript:void(0);" name="Declaration of patient consent: ">Declaration of patient consent: </a></h3><div class="clearfix"></div><div class="hline"></div><p>Patient’s consent was not required as there are no patients in this study.</p><h3 class="blogheading Main-Title"><a href="javascript:void(0);" name="Financial support and sponsorship: ">Financial support and sponsorship: </a></h3><div class="clearfix"></div><div class="hline"></div><p>Nil.</p><h3 class="blogheading Main-Title"><a href="javascript:void(0);" name="Conflicts of interest: ">Conflicts of interest: </a></h3><div class="clearfix"></div><div class="hline"></div><p>There are no conflicts of interest.</p><h3 class="blogheading Main-Title"><a href="javascript:void(0);" name="Use of artificial intelligence (AI)-assisted technology for manuscript preparation: ">Use of artificial intelligence (AI)-assisted technology for manuscript preparation: </a></h3><div class="clearfix"></div><div class="hline"></div><p>The authors confirm that there was no use of artificial intelligence (AI)-assisted technology for assisting in the writing or editing of the manuscript and no images were manipulated using AI.</p><h3 class="blogheading Main-Title"><a href="javascript:void(0);" name="Disclaimer">Disclaimer</a></h3><div class="clearfix"></div><div class="hline"></div><p>The views and opinions expressed in this article are those of the authors and do not necessarily reflect the official policy or position of the Journal or its management. The information contained in this article should not be considered to be medical advice; patients should consult their own physicians for advice as to their specific medical needs.</p></div> </div></div><div><div class="row"> <div class="blogparagraph col-lg-9 col-sm-8 col-xs-12"><h3 class="blogheading pull-left Main-Title col-lg-9 col-sm-8 col-xs-12"><a href="javascript:void(0);" name="Acknowledgments: ">Acknowledgments: </a></h3><div class="clearfix"></div><div class="hline"></div><p>Figure 1 has been created with BioRender.com.</p></div> </div><div class="row"> <div class="blogparagraph col-lg-9 col-sm-8 col-xs-12"> <h3 class="blogheading pull-left Main-Title"><a name="References" href="javascript:void(0);">References</a></h3> <div class="clearfix"></div> <div class="hline"></div> <p><a href='javascript:void(0);' name='ref1' style='text-decoration: none;'>1.</a> Allan SM, Rothwell NJ. Cytokines and acute neurodegeneration. Nat Rev Neurosci. 2001. 2: 734-44</p><p><a href='javascript:void(0);' name='ref2' style='text-decoration: none;'>2.</a> Apple DM, Solano-Fonseca R, Kokovay E. Neurogenesis in the aging brain. Biochem Pharmacol. 2017. 141: 77-85</p><p><a href='javascript:void(0);' name='ref3' style='text-decoration: none;'>3.</a> Asensio VC, Campbell IL. Chemokines and viral diseases of the central nervous system. Adv Virus Res. 2001. 56: 127-73</p><p><a href='javascript:void(0);' name='ref4' style='text-decoration: none;'>4.</a> Ayala-Nunez NV, Gaudin R. A viral journey to the brain: Current considerations and future developments. PLoS Pathog. 2020. 16: e1008434</p><p><a href='javascript:void(0);' name='ref5' style='text-decoration: none;'>5.</a> Ball MJ. Neuronal loss, neurofibrillary tangles and granulovacuolar degeneration in the hippocampus with ageing and dementia. A quantitative study. Acta Neuropathol. 1977. 37: 111-8</p><p><a href='javascript:void(0);' name='ref6' style='text-decoration: none;'>6.</a> Banks WA, Plotkin SR, Kastin AJ. Permeability of the blood-brain barrier to soluble cytokine receptors. Neuroimmunomodulation. 1995. 2: 161-5</p><p><a href='javascript:void(0);' name='ref7' style='text-decoration: none;'>7.</a> Behzad F, Sefidgar E, Samadi A, Lin W, Pouladi I, Pi J. An overview of zinc oxide nanoparticles produced by plant extracts for anti-tuberculosis treatments. Curr Med Chem. 2022. 29: 86-98</p><p><a href='javascript:void(0);' name='ref8' style='text-decoration: none;'>8.</a> Brochard V, Combadière B, Prigent A, Laouar Y, Perrin A, Beray-Berthat V. Infiltration of CD4+ lymphocytes into the brain contributes to neurodegeneration in a mouse model of Parkinson disease. J Clin Invest. 2009. 119: 182-92</p><p><a href='javascript:void(0);' name='ref9' style='text-decoration: none;'>9.</a> Burke SN, Barnes CA. Neural plasticity in the ageing brain. Nat Rev Neurosci. 2006. 7: 30-40</p><p><a href='javascript:void(0);' name='ref10' style='text-decoration: none;'>10.</a> Campos EM, Rodrigues LD, Oliveira LF, Dos Santos JC. Dementia and cognitive impairment in adults as sequels of HSV-1-related encephalitis: A review. Dement Neuropsychol. 2021. 15: 164-72</p><p><a href='javascript:void(0);' name='ref11' style='text-decoration: none;'>11.</a> Carpenter J, Stapleton S, Holliman R. Retrospective analysis of 49 cases of brain abscess and review of the literature. Eur J Clin Microbiol Infect Dis. 2007. 26: 1-11</p><p><a href='javascript:void(0);' name='ref12' style='text-decoration: none;'>12.</a> Carson MJ, Doose JM, Melchior B, Schmid CD, Ploix CC. CNS immune privilege: Hiding in plain sight. Immunol Rev. 2006. 213: 48-65</p><p><a href='javascript:void(0);' name='ref13' style='text-decoration: none;'>13.</a> Chanthick C, Suttitheptumrong A, Rawarak N, Pattanakitsakul SN. Transcytosis Involvement in transport system and endothelial permeability of vascular leakage during dengue virus infection. Viruses. 2018. 10: 69</p><p><a href='javascript:void(0);' name='ref14' style='text-decoration: none;'>14.</a> Chen Y, Xu J, Chen Y. Regulation of neurotransmitters by the gut microbiota and effects on cognition in neurological disorders. Nutrients. 2021. 13: 2099</p><p><a href='javascript:void(0);' name='ref15' style='text-decoration: none;'>15.</a> Cheon SY, Kim JM, Kam EH, Ho CC, Kim EJ, Chung S. Cell-penetrating interactomic inhibition of nuclear factor-kappa B in a mouse model of postoperative cognitive dysfunction. Sci Rep. 2017. 7: 13482</p><p><a href='javascript:void(0);' name='ref16' style='text-decoration: none;'>16.</a> Choi MR, Stanton-Maxey KJ, Stanley JK, Levin CS, Bardhan R, Akin D. A cellular Trojan Horse for delivery of therapeutic nanoparticles into tumors. Nano Lett. 2007. 7: 3759-65</p><p><a href='javascript:void(0);' name='ref17' style='text-decoration: none;'>17.</a> Chu H, Xiang J, Wu P, Su J, Ding H, Tang Y. The role of aquaporin 4 in apoptosis after intracerebral hemorrhage. J Neuroinflammation. 2014. 11: 184</p><p><a href='javascript:void(0);' name='ref18' style='text-decoration: none;'>18.</a> Clemens JA, Stephenson DT, Smalstig EB, Dixon EP, Little SP. Global ischemia activates nuclear factor-kB in forebrain neurons of rats. Stroke. 1997. 28: 1073-81</p><p><a href='javascript:void(0);' name='ref19' style='text-decoration: none;'>19.</a> Constam DB, Philipp J, Malipiero UV, Ten Dijke P, Schachner M, Fontana A. Differential expression of transforming growth factor-beta 1,-beta 2, and-beta 3 by glioblastoma cells, astrocytes, and microglia. J Immunol. 1992. 148: 1404-10</p><p><a href='javascript:void(0);' name='ref20' style='text-decoration: none;'>20.</a> Davies D. Blood-brain barrier breakdown and oedema formation in systemic sepsis and human brain tumours. J Anat. 2002. 200: 528-29</p><p><a href='javascript:void(0);' name='ref21' style='text-decoration: none;'>21.</a> De Tiège X, Rozenberg F, Des Portes V, Lobut JB, Lebon P, Ponsot G. Herpes simplex encephalitis relapses in children: Differentiation of two neurologic entities. Neurology. 2003. 61: 241-3</p><p><a href='javascript:void(0);' name='ref22' style='text-decoration: none;'>22.</a> Degos V, Vacas S, Han Z, Van Rooijen N, Gressens P, Su H. Depletion of bone marrow-derived macrophages perturbs the innate immune response to surgery and reduces postoperative memory dysfunction. Anesthesiology. 2013. 118: 527-36</p><p><a href='javascript:void(0);' name='ref23' style='text-decoration: none;'>23.</a> Dorrier CE, McGavern DB. Humoral immune defense of the central nervous system. Curr Opin Immunol. 2022. 76: 102179</p><p><a href='javascript:void(0);' name='ref24' style='text-decoration: none;'>24.</a> Engblom D, Monica E, Saha S, Ericsson-Dahlstrand A, Jakobsson PJ, Blomqvist A. Prostaglandins as inflammatory messengers across the blood-brain barrier. J Mol Med (Berl). 2002. 80: 5-15</p><p><a href='javascript:void(0);' name='ref25' style='text-decoration: none;'>25.</a> Fang EF, Hou Y, Palikaras K, Adriaanse BA, Kerr JS, Yang B. Mitophagy inhibits amyloid-b and tau pathology and reverses cognitive deficits in models of Alzheimer’s disease. Nat Neurosci. 2019. 22: 401-12</p><p><a href='javascript:void(0);' name='ref26' style='text-decoration: none;'>26.</a> Fitzpatrick Z, Frazer G, Ferro A, Clare S, Bouladoux N, Ferdinand J. Gut-educated IgA plasma cells defend the meningeal venous sinuses. Nature. 2020. 587: 472-6</p><p><a href='javascript:void(0);' name='ref27' style='text-decoration: none;'>27.</a> Galea I, Bechmann I, Perry VH. What is immune privilege (not)?. Trends Immunol. 2007. 28: 12-8</p><p><a href='javascript:void(0);' name='ref28' style='text-decoration: none;'>28.</a> Gao S, Zhang S, Zhou H, Tao X, Ni Y, Pei D. Role of mTOR-regulated autophagy in synaptic plasticity related proteins downregulation and the reference memory deficits induced by anesthesia/surgery in aged mice. Front Aging Neurosci. 2021. 13: 628541</p><p><a href='javascript:void(0);' name='ref29' style='text-decoration: none;'>29.</a> Giovane RA, Lavender PD. Central nervous system infections. Prim Care. 2018. 45: 505-18</p><p><a href='javascript:void(0);' name='ref30' style='text-decoration: none;'>30.</a> Glass CK, Saijo K, Winner B, Marchetto MC, Gage FH. Mechanisms underlying inflammation in neurodegeneration. Cell. 2010. 140: 918-34</p><p><a href='javascript:void(0);' name='ref31' style='text-decoration: none;'>31.</a> Goehler LE, Gaykema RP, Opitz N, Reddaway R, Badr N, Lyte M. Activation in vagal afferents and central autonomic pathways: Early responses to intestinal infection with Campylobacter jejuni. Brain Behav Immun. 2005. 19: 334-44</p><p><a href='javascript:void(0);' name='ref32' style='text-decoration: none;'>32.</a> Gomez AM, Traunmüller L, Scheiffele P. Neurexins: Molecular codes for shaping neuronal synapses. Nat Rev Neurosci. 2021. 22: 137-51</p><p><a href='javascript:void(0);' name='ref33' style='text-decoration: none;'>33.</a> Gordon L, McQuaid S, Cosby SL. Detection of herpes simplex virus (types 1 and 2) and human herpesvirus 6 DNA in human brain tissue by polymerase chain reaction. Clin Diagn Virol. 1996. 6: 33-40</p><p><a href='javascript:void(0);' name='ref34' style='text-decoration: none;'>34.</a> Gruenbaum SE, Toscani L, Fomberstein KM, Ruskin KJ, Dai F, Qeva E. Severe intraoperative hyperglycemia is independently associated with postoperative composite infection after craniotomy: An observational study. Anesth Analg. 2017. 125: 556-61</p><p><a href='javascript:void(0);' name='ref35' style='text-decoration: none;'>35.</a> Hanamsagar R, Hanke ML, Kielian T. Toll-like receptor (TLR) and inflammasome actions in the central nervous system. Trends Immunol. 2012. 33: 333-42</p><p><a href='javascript:void(0);' name='ref36' style='text-decoration: none;'>36.</a> He HJ, Wang Y, Le Y, Duan KM, Yan XB, Liao Q. Surgery upregulates high mobility group box-1 and disrupts the blood-brain barrier causing cognitive dysfunction in aged rats. CNS Neurosci Ther. 2012. 18: 994-1002</p><p><a href='javascript:void(0);' name='ref37' style='text-decoration: none;'>37.</a> Hovens IB, Schoemaker RG, Van der Zee EA, Heineman E, Izaks GJ, Van Leeuwen BL. Thinking through postoperative cognitive dysfunction: How to bridge the gap between clinical and pre-clinical perspectives. Brain Behav Immun. 2012. 26: 1169-79</p><p><a href='javascript:void(0);' name='ref38' style='text-decoration: none;'>38.</a> Hridi SU, Franssen AJ, Jiang HR, Bushell TJ. Interleukin-16 inhibits sodium channel function and GluA1 phosphorylation via CD4-and CD9-independent mechanisms to reduce hippocampal neuronal excitability and synaptic activity. Mol Cell Neurosci. 2019. 95: 71-8</p><p><a href='javascript:void(0);' name='ref39' style='text-decoration: none;'>39.</a> Iliff JJ, Goldman SA, Nedergaard M. Implications of the discovery of brain lymphatic pathways. Lancet Neurol. 2015. 14: 977-9</p><p><a href='javascript:void(0);' name='ref40' style='text-decoration: none;'>40.</a> Jorgačevski J, Potokar M. Immune functions of astrocytes in viral neuroinfections. Int J Mol Sci. 2023. 24: 3514</p><p><a href='javascript:void(0);' name='ref41' style='text-decoration: none;'>41.</a> Kang SS, McGavern DB. Inflammation on the mind: Visualizing immunity in the central nervous system. Curr Top Microbiol Immunol. 2009. 334: 227-63</p><p><a href='javascript:void(0);' name='ref42' style='text-decoration: none;'>42.</a> Kapoor R, Hussain I, Hussain I, Stieg P, editors. Basic neurosurgical procedures. Cham: Springer; 2015. p. </p><p><a href='javascript:void(0);' name='ref43' style='text-decoration: none;'>43.</a> Kigerl KA, De Rivero Vaccari JP, Dietrich WD, Popovich PG, Keane RW. Pattern recognition receptors and central nervous system repair. Exp Neurol. 2014. 258: 5-16</p><p><a href='javascript:void(0);' name='ref44' style='text-decoration: none;'>44.</a> Kivisäkk P, Mahad DJ, Callahan MK, Trebst C, Tucky B, Wei T. Human cerebrospinal fluid central memory CD4+ T cells: Evidence for trafficking through choroid plexus and meninges via P-selectin. Proc Natl Acad Sci U S A. 2003. 100: 8389-94</p><p><a href='javascript:void(0);' name='ref45' style='text-decoration: none;'>45.</a> Krawczyk A, Arndt MA, Grosse-Hovest L, Weichert W, Giebel B, Dittmer U. Overcoming drug-resistant herpes simplex virus (HSV) infection by a humanized antibody. Proc Natl Acad Sci U S A. 2013. 110: 6760-5</p><p><a href='javascript:void(0);' name='ref46' style='text-decoration: none;'>46.</a> Krett JD, Beckham JD, Tyler KL, Piquet AL, Chauhan L, Wallace CJ, et al. Neurology of acute viral infections. Neurohospitalist. 2022. 12: 632-46</p><p><a href='javascript:void(0);' name='ref47' style='text-decoration: none;'>47.</a> Kulikov A, Krovko Y, Nikitin A, Shmigelsky A, Zagidullin T, Ershova O. Severe intraoperative hyperglycemia and infectious complications after elective brain neurosurgical procedures: Prospective observational study. Anesth Analg. 2022. 135: 1082-8</p><p><a href='javascript:void(0);' name='ref48' style='text-decoration: none;'>48.</a> Kurtel H, Fujimoto K, Zimmerman BJ, Granger DN, Tso P. Ischemia-reperfusion-induced mucosal dysfunction: Role of neutrophils. Am J Physiol. 1991. 261: G490-6</p><p><a href='javascript:void(0);' name='ref49' style='text-decoration: none;'>49.</a> Lai Z, Shan W, Li J, Min J, Zeng X, Zuo Z. Appropriate exercise level attenuates gut dysbiosis and valeric acid increase to improve neuroplasticity and cognitive function after surgery in mice. Mol Psychiatry. 2021. 26: 7167-87</p><p><a href='javascript:void(0);' name='ref50' style='text-decoration: none;'>50.</a> Lane TE, Hardison JL, Walsh KB. Functional diversity of chemokines and chemokine receptors in response to viral infection of the central nervous system. Curr Top Microbiol Immunol. 2006. 303: 1-27</p><p><a href='javascript:void(0);' name='ref51' style='text-decoration: none;'>51.</a> Lange N, Berndt M, Jörger AK, Wagner A, Lummel N, Ryang YM. Clinical characteristics and course of postoperative brain abscess. World Neurosurg. 2018. 120: 675-83</p><p><a href='javascript:void(0);' name='ref52' style='text-decoration: none;'>52.</a> Lee KS, Borbas B, Chaudhry D, Kuri A, Best L, Gillespie CS. Surgical site infection after craniotomy in neurooncology (SINO): A protocol for an international prospective multicentre service evaluation across the United Kingdom and Ireland. PLoS One. 2025. 20: e0316237</p><p><a href='javascript:void(0);' name='ref53' style='text-decoration: none;'>53.</a> Letenneur L, Pérès K, Fleury H, Garrigue I, Barberger-Gateau P, Helmer C. Seropositivity to herpes simplex virus antibodies and risk of Alzheimer’s disease: A population-based cohort study. PLoS One. 2008. 3: e3637</p><p><a href='javascript:void(0);' name='ref54' style='text-decoration: none;'>54.</a> Li WX, Luo RY, Chen C, Li X, Ao JS, Liu Y. Effects of propofol, dexmedetomidine, and midazolam on postoperative cognitive dysfunction in elderly patients: A randomized controlled preliminary trial. Chin Med J (Engl). 2019. 132: 437-45</p><p><a href='javascript:void(0);' name='ref55' style='text-decoration: none;'>55.</a> Liang S, Wang Z, Wu P, Chen Z, Yang X, Li Y. Risk factors and outcomes of central nervous system infection after spinal surgery: A retrospective cohort study. World Neurosurg. 2023. 170: e170-9</p><p><a href='javascript:void(0);' name='ref56' style='text-decoration: none;'>56.</a> Libbey JE, Fujinami RS. Adaptive immune response to viral infections in the central nervous system. Handb Clin Neurol. 2014. 123: 225-47</p><p><a href='javascript:void(0);' name='ref57' style='text-decoration: none;'>57.</a> Liblau RS, Gonzalez-Dunia D, Wiendl H, Zipp F. Neurons as targets for T cells in the nervous system. Trends Neurosci. 2013. 36: 315-24</p><p><a href='javascript:void(0);' name='ref58' style='text-decoration: none;'>58.</a> Lieberman AP, Pitha PM, Shin HS, Shin ML. Production of tumor necrosis factor and other cytokines by astrocytes stimulated with lipopolysaccharide or a neurotropic virus. Proc Natl Acad Sci U S A. 1989. 86: 6348-52</p><p><a href='javascript:void(0);' name='ref59' style='text-decoration: none;'>59.</a> Lin L, Wang X, Yu Z. Ischemia-reperfusion injury in the brain: Mechanisms and potential therapeutic strategies. Biochem Pharmacol (Los Angel). 2016. 5: 213</p><p><a href='javascript:void(0);' name='ref60' style='text-decoration: none;'>60.</a> Liu Y, Yin Y. Emerging roles of immune cells in postoperative cognitive dysfunction. Mediators Inflamm. 2018. 2018: 6215350</p><p><a href='javascript:void(0);' name='ref61' style='text-decoration: none;'>61.</a> Lodge PA, Sriram S. Regulation of microglial activation by TGF-beta, IL-10, and CSF-1. J Leukoc Biol. 1996. 60: 502-8</p><p><a href='javascript:void(0);' name='ref62' style='text-decoration: none;'>62.</a> Louveau A, Smirnov I, Keyes TJ, Eccles JD, Rouhani SJ, Peske JD. Structural and functional features of central nervous system lymphatic vessels. Nature. 2015. 523: 337-41</p><p><a href='javascript:void(0);' name='ref63' style='text-decoration: none;'>63.</a> Luo F, Min J, Wu J, Zuo Z. Histone deacetylases may mediate surgery-induced impairment of learning, memory, and dendritic development. Mol Neurobiol. 2020. 57: 3702-11</p><p><a href='javascript:void(0);' name='ref64' style='text-decoration: none;'>64.</a> Lv S, Song HL, Zhou Y, Li LX, Cui W, Wang W. Tumour necrosis factor-alpha affects blood-brain barrier permeability and tight junction-associated occludin in acute liver failure. Liver Int. 2010. 30: 1198-210</p><p><a href='javascript:void(0);' name='ref65' style='text-decoration: none;'>65.</a> Magee JC, Grienberger C. Synaptic plasticity forms and functions. Annu Rev Neurosci. 2020. 43: 95-117</p><p><a href='javascript:void(0);' name='ref66' style='text-decoration: none;'>66.</a> Manninen PH, Raman SK, Boyle K, El-Beheiry H. Early postoperative complications following neurosurgical procedures. Can J Anaesth. 1999. 46: 7-14</p><p><a href='javascript:void(0);' name='ref67' style='text-decoration: none;'>67.</a> Medana IM, Gallimore A, Oxenius A, Martinic MM, Wekerle H, Neumann H. MHC class I-restricted killing of neurons by virus-specific CD8+ T lymphocytes is effected through the Fas/FasL, but not the perforin pathway. Eur J Immunol. 2000. 30: 3623-33</p><p><a href='javascript:void(0);' name='ref68' style='text-decoration: none;'>68.</a> Mendez OA, Koshy AA. Toxoplasma gondii Entry, association, and physiological influence on the central nervous system. PLoS Pathog. 2017. 13: e1006351</p><p><a href='javascript:void(0);' name='ref69' style='text-decoration: none;'>69.</a> Metcalf TU, Baxter VK, Nilaratanakul V, Griffin DE. Recruitment and retention of B cells in the central nervous system in response to alphavirus encephalomyelitis. J Virol. 2013. 87: 2420-9</p><p><a href='javascript:void(0);' name='ref70' style='text-decoration: none;'>70.</a> Moon SM, Kim T, Lee EM, Kang JK, Lee SA, Choi SH. Comparison of clinical manifestations, outcomes and cerebrospinal fluid findings between herpes simplex type 1 and type 2 central nervous system infections in adults. J Med Virol. 2014. 86: 1766-71</p><p><a href='javascript:void(0);' name='ref71' style='text-decoration: none;'>71.</a> Murphy K, Weaver C, editors. Innate immunity: The first lines of defense. Janeway’s immunobiology. United States: W.W. Norton and Company; 2017. p. 37-76</p><p><a href='javascript:void(0);' name='ref72' style='text-decoration: none;'>72.</a> Netto MB, De Oliveira Junior AN, Goldim M, Mathias K, Fileti ME, Da Rosa N. Oxidative stress and mitochondrial dysfunction contributes to postoperative cognitive dysfunction in elderly rats. Brain Behav Immun. 2018. 73: 661-9</p><p><a href='javascript:void(0);' name='ref73' style='text-decoration: none;'>73.</a> Nimmerjahn A, Kirchhoff F, Helmchen F. Resting microglial cells are highly dynamic surveillants of brain parenchyma in vivo. Science. 2005. 308: 1314-8</p><p><a href='javascript:void(0);' name='ref74' style='text-decoration: none;'>74.</a> Pappa M, Theodosiadis N, Tsounis A, Sarafis P. Pathogenesis and treatment of post-operative cognitive dysfunction. Electron Physician. 2017. 9: 3768-75</p><p><a href='javascript:void(0);' name='ref75' style='text-decoration: none;'>75.</a> Pardridge WM. CSF, blood-brain barrier, and brain drug delivery. Expert Opin Drug Deliv. 2016. 13: 963-75</p><p><a href='javascript:void(0);' name='ref76' style='text-decoration: none;'>76.</a> Parra B, Hinton DR, Marten NW, Bergmann CC, Lin MT, Yang CS. IFN-gamma is required for viral clearance from central nervous system oligodendroglia. J Immunol. 1999. 162: 1641-7</p><p><a href='javascript:void(0);' name='ref77' style='text-decoration: none;'>77.</a> Patel S, Thompson D, Innocent S, Narbad V, Selway R, Barkas K. Risk factors for surgical site infections in neurosurgery. Ann R Coll Surg Engl. 2019. 101: 220-5</p><p><a href='javascript:void(0);' name='ref78' style='text-decoration: none;'>78.</a> Pedemonte E, Mancardi G, Giunti D, Corcione A, Benvenuto F, Pistoia V. Mechanisms of the adaptive immune response inside the central nervous system during inflammatory and autoimmune diseases. Pharmacol Ther. 2006. 111: 555-66</p><p><a href='javascript:void(0);' name='ref79' style='text-decoration: none;'>79.</a> Peluso R, Haase A, Stowring L, Edwards M, Ventura P. A Trojan Horse mechanism for the spread of visna virus in monocytes. Virology. 1985. 147: 231-6</p><p><a href='javascript:void(0);' name='ref80' style='text-decoration: none;'>80.</a> Peng M, Wang YL, Wang CY, Chen C. Dexmedetomidine attenuates lipopolysaccharide-induced proinflammatory response in primary microglia. J Surg Res. 2013. 179: e219-25</p><p><a href='javascript:void(0);' name='ref81' style='text-decoration: none;'>81.</a> Piacentini R, Civitelli L, Ripoli C, Marcocci ME, De Chiara G, Garaci E. HSV-1 promotes Ca2+-mediated APP phosphorylation and Ab accumulation in rat cortical neurons. Neurobiol Aging. 2011. 32: 2323.e13-26</p><p><a href='javascript:void(0);' name='ref82' style='text-decoration: none;'>82.</a> Pribiag H, Stellwagen D. TNF-a downregulates inhibitory neurotransmission through protein phosphatase 1-dependent trafficking of GABAA receptors. J Neurosci. 2013. 33: 15879-93</p><p><a href='javascript:void(0);' name='ref83' style='text-decoration: none;'>83.</a> Raschilas F, Wolff M, Delatour F, Chaffaut C, De Broucker T, Chevret S. Outcome of and prognostic factors for herpes simplex encephalitis in adult patients: Results of a multicenter study. Clin Infect Dis. 2002. 35: 254-60</p><p><a href='javascript:void(0);' name='ref84' style='text-decoration: none;'>84.</a> Rempe RG, Hartz AM, Bauer B. Matrix metalloproteinases in the brain and blood-brain barrier: Versatile breakers and makers. J Cereb Blood Flow Metab. 2016. 36: 1481-507</p><p><a href='javascript:void(0);' name='ref85' style='text-decoration: none;'>85.</a> Rogers KJ, Vijay R, Butler NS. Anti-malarial humoral immunity: The long and short of it. Microbes Infect. 2021. 23: 104807</p><p><a href='javascript:void(0);' name='ref86' style='text-decoration: none;'>86.</a> Rouse BT, Sehrawat S. Immunity and immunopathology to viruses: What decides the outcome?. Nat Rev Immunol. 2010. 10: 514-26</p><p><a href='javascript:void(0);' name='ref87' style='text-decoration: none;'>87.</a> Salimi H, Klein R, editors. Disruption of the blood-brain barrier during neuroinflammatory and neuroinfectious diseases. Cham: Springer; 2019. p. </p><p><a href='javascript:void(0);' name='ref88' style='text-decoration: none;'>88.</a> Salimi H, Klein RS. Disruption of the blood-brain barrier during neuroinflammatory and neuroinfectious diseases. Neuroimmune Diseases. 2019. 1: 195-234</p><p><a href='javascript:void(0);' name='ref89' style='text-decoration: none;'>89.</a> Santiago-Tirado FH, Onken MD, Cooper JA, Klein RS, Doering TL. Trojan Horse transit contributes to blood-brain barrier crossing of a eukaryotic pathogen. mBio. 2017. 8: </p><p><a href='javascript:void(0);' name='ref90' style='text-decoration: none;'>90.</a> Savarin C, Bergmann CC. Neuroimmunology of central nervous system viral infections: The cells, molecules and mechanisms involved. Curr Opin Pharmacol. 2008. 8: 472-9</p><p><a href='javascript:void(0);' name='ref91' style='text-decoration: none;'>91.</a> Saxena S, Maze M. Impact on the brain of the inflammatory response to surgery. Presse Méd. 2018. 47: e73-81</p><p><a href='javascript:void(0);' name='ref92' style='text-decoration: none;'>92.</a> Schafflick D, Wolbert J, Heming M, Thomas C, Hartlehnert M, Börsch AL. Single-cell profiling of CNS border compartment leukocytes reveals that B cells and their progenitors reside in non-diseased meninges. Nat Neurosci. 2021. 24: 1225-34</p><p><a href='javascript:void(0);' name='ref93' style='text-decoration: none;'>93.</a> Schwitter J, Branca M, Bicvic A, Abbuehl LS, Suter-Riniker F, Leib SL. Long-term sequelae after viral meningitis and meningoencephalitis are frequent, even in mildly affected patients, a prospective observational study. Front Neurol. 2024. 15: 1411860</p><p><a href='javascript:void(0);' name='ref94' style='text-decoration: none;'>94.</a> Sebina I, Pepper M. Humoral immune responses to infection: Common mechanisms and unique strategies to combat pathogen immune evasion tactics. Curr Opin Immunol. 2018. 51: 46-54</p><p><a href='javascript:void(0);' name='ref95' style='text-decoration: none;'>95.</a> Shao A, Fei J, Feng S, Weng J. Chikusetsu saponin IVa alleviated sevoflurane-induced neuroinflammation and cognitive impairment by blocking NLRP3/caspase-1 pathway. Pharmacol Rep. 2020. 72: 833-45</p><p><a href='javascript:void(0);' name='ref96' style='text-decoration: none;'>96.</a> Shastri A, Bonifati DM, Kishore U. Innate immunity and neuroinflammation. Mediators Inflamm. 2013. 2013: 342931</p><p><a href='javascript:void(0);' name='ref97' style='text-decoration: none;'>97.</a> Skvarc DR, Berk M, Byrne LK, Dean OM, Dodd S, Lewis M. Post-operative cognitive dysfunction: An exploration of the inflammatory hypothesis and novel therapies. Neurosci Biobehav Rev. 2018. 84: 116-33</p><p><a href='javascript:void(0);' name='ref98' style='text-decoration: none;'>98.</a> Smith PA. The tantalizing links between gut microbes and the brain. Nature. 2015. 526: 312-4</p><p><a href='javascript:void(0);' name='ref99' style='text-decoration: none;'>99.</a> Sofroniew MV. Molecular dissection of reactive astrogliosis and glial scar formation. Trends Neurosci. 2009. 32: 638-47</p><p><a href='javascript:void(0);' name='ref100' style='text-decoration: none;'>100.</a> Steel CD, Hahto SM, Ciavarra RP. Peripheral dendritic cells are essential for both the innate and adaptive antiviral immune responses in the central nervous system. Virology. 2009. 387: 117-26</p><p><a href='javascript:void(0);' name='ref101' style='text-decoration: none;'>101.</a> Sulhan S, Lyon KA, Shapiro LA, Huang JH. Neuroinflammation and blood-brain barrier disruption following traumatic brain injury: Pathophysiology and potential therapeutic targets. J Neurosci Res. 2020. 98: 19-28</p><p><a href='javascript:void(0);' name='ref102' style='text-decoration: none;'>102.</a> Sun W, Zhao J, Li C. Dexmedetomidine provides protection against hippocampal neuron apoptosis and cognitive impairment in mice with Alzheimer’s disease by mediating the miR-129/YAP1/JAG1 axis. Mol Neurobiol. 2020. 57: 5044-55</p><p><a href='javascript:void(0);' name='ref103' style='text-decoration: none;'>103.</a> Taguchi T, Mukai K. Innate immunity signalling and membrane trafficking. Curr Opin Cell Biol. 2019. 59: 1-7</p><p><a href='javascript:void(0);' name='ref104' style='text-decoration: none;'>104.</a> Takata F, Nakagawa S, Matsumoto J, Dohgu S. Blood-brain barrier dysfunction amplifies the development of neuroinflammation: Understanding of cellular events in brain microvascular endothelial cells for prevention and treatment of BBB dysfunction. Front Cell Neurosci. 2021. 15: 661838</p><p><a href='javascript:void(0);' name='ref105' style='text-decoration: none;'>105.</a> Tan XX, Qiu LL, Sun J. Research progress on the role of inflammatory mechanisms in the development of postoperative cognitive dysfunction. Biomed Res Int. 2021. 2021: 3883204</p><p><a href='javascript:void(0);' name='ref106' style='text-decoration: none;'>106.</a> Telikani Z, Monson EA, Hofer MJ, Helbig KJ. Antiviral response within different cell types of the CNS. Front Immunol. 2022. 13: 1044721</p><p><a href='javascript:void(0);' name='ref107' style='text-decoration: none;'>107.</a> Tian A, Ma H, Cao X, Zhang R, Wang X, Wu B. Vitamin D improves cognitive function and modulates Th17/T reg cell balance after hepatectomy in mice. Inflammation. 2015. 38: 500-9</p><p><a href='javascript:void(0);' name='ref108' style='text-decoration: none;'>108.</a> Tunkel AR, Glaser CA, Bloch KC, Sejvar JJ, Marra CM, Roos KL. The management of encephalitis: Clinical practice guidelines by the infectious diseases society of America. Clin Infect Dis. 2008. 47: 303-27</p><p><a href='javascript:void(0);' name='ref109' style='text-decoration: none;'>109.</a> Van Neerven S, Nemes A, Imholz P, Regen T, Denecke B, Johann S. Inflammatory cytokine release of astrocytes in vitro is reduced by all-trans retinoic acid. J Neuroimmunol. 2010. 229: 169-79</p><p><a href='javascript:void(0);' name='ref110' style='text-decoration: none;'>110.</a> Vasic V, Barth K, Schmidt MH. Neurodegeneration and neuro-regeneration-Alzheimer’s disease and stem cell therapy. Int J Mol Sci. 2019. 20: 4272</p><p><a href='javascript:void(0);' name='ref111' style='text-decoration: none;'>111.</a> Velnar T, Kocivnik N, Bosnjak R. Clinical infections in neurosurgical oncology: An overview. World J Clin Cases. 2023. 11: 3418-33</p><p><a href='javascript:void(0);' name='ref112' style='text-decoration: none;'>112.</a> Von Zahn J, Möller T, Kettenmann H, Nolte C. Microglial phagocytosis is modulated by pro-and anti-inflammatory cytokines. Neuroreport. 1997. 8: 3851-6</p><p><a href='javascript:void(0);' name='ref113' style='text-decoration: none;'>113.</a> Wei P, Yang F, Zheng Q, Tang W, Li J. The potential role of the NLRP3 inflammasome activation as a link between mitochondria ROS generation and neuroinflammation in postoperative cognitive dysfunction. Front Cell Neurosci. 2019. 13: 73</p><p><a href='javascript:void(0);' name='ref114' style='text-decoration: none;'>114.</a> Williams K, Dooley N, Ulvestad E, Becher B, Antel JP. IL-10 production by adult human derived microglial cells. Neurochem Int. 1996. 29: 55-64</p><p><a href='javascript:void(0);' name='ref115' style='text-decoration: none;'>115.</a> Woodroofe MN. Cytokine production in the central nervous system. Neurology. 1995. 45: S6-10</p><p><a href='javascript:void(0);' name='ref116' style='text-decoration: none;'>116.</a> Xiao JY, Xiong BR, Zhang W, Zhou WC, Yang H, Gao F. PGE 2-EP 3 signaling exacerbates hippocampus-dependent cognitive impairment after laparotomy by reducing expression levels of hippocampal synaptic plasticity-related proteins in aged mice. CNS Neurosci Therapeut. 2018. 24: 917-29</p><p><a href='javascript:void(0);' name='ref117' style='text-decoration: none;'>117.</a> Xu J, Dong H, Qian Q, Zhang X, Wang Y, Jin W. Astrocyte-derived CCL2 participates in surgery-induced cognitive dysfunction and neuroinflammation via evoking microglia activation. Behav Brain Res. 2017. 332: 145-53</p><p><a href='javascript:void(0);' name='ref118' style='text-decoration: none;'>118.</a> Yang L, Hao JR, Gao Y, Yang X, Shen XR, Wang HY. HDAC3 of dorsal hippocampus induces postoperative cognitive dysfunction in aged mice. Behav Brain Res. 2022. 433: 114002</p><p><a href='javascript:void(0);' name='ref119' style='text-decoration: none;'>119.</a> Yang N, Liang Y, Yang P, Wang W, Zhang X, Wang J. TNF-a receptor antagonist attenuates isoflurane-induced cognitive impairment in aged rats. Exp Ther Med. 2016. 12: 463-8</p><p><a href='javascript:void(0);' name='ref120' style='text-decoration: none;'>120.</a> Yang S, Gu C, Mandeville ET, Dong Y, Esposito E, Zhang Y. Anesthesia and surgery impair blood-brain barrier and cognitive function in mice. Front Immunol. 2017. 8: 902</p><p><a href='javascript:void(0);' name='ref121' style='text-decoration: none;'>121.</a> Yang XD, Wang LK, Wu HY, Jiao L. Effects of prebiotic galactooligosaccharide on postoperative cognitive dysfunction and neuroinflammation through targeting of the gut-brain axis. BMC Anesthesiol. 2018. 18: 177</p><p><a href='javascript:void(0);' name='ref122' style='text-decoration: none;'>122.</a> Ying M, Wang H, Liu T, Han Z, Lin K, Shi Q. CLEAR strategy inhibited HSV proliferation using viral vectors delivered CRISPR-Cas9. Pathogens. 2023. 12: 814</p><p><a href='javascript:void(0);' name='ref123' style='text-decoration: none;'>123.</a> Young GB, Bolton CF, Archibald YM, Austin TW, Wells GA. The electroencephalogram in sepsis-associated encephalopathy. J Clin Neurophysiol. 1992. 9: 145-52</p><p><a href='javascript:void(0);' name='ref124' style='text-decoration: none;'>124.</a> Yuan N, Wang X, Zhang Y, Kong L, Yuan L, Ge Y. Intervention of NF-Kb signaling pathway and preventing post-operative cognitive dysfunction as well as neuronal apoptosis. Iran J Public Health. 2022. 51: 124-132</p><p><a href='javascript:void(0);' name='ref125' style='text-decoration: none;'>125.</a> Zhan G, Hua D, Huang N, Wang Y, Li S, Zhou Z. Anesthesia and surgery induce cognitive dysfunction in elderly male mice: The role of gut microbiota. Aging (Albany NY). 2019. 11: 1778-90</p><p><a href='javascript:void(0);' name='ref126' style='text-decoration: none;'>126.</a> Zhang S, Dong H, Zhang X, Li N, Sun J, Qian Y. Cerebral mast cells contribute to postoperative cognitive dysfunction by promoting blood brain barrier disruption. Behav Brain Res. 2016. 298: 158-66</p><p><a href='javascript:void(0);' name='ref127' style='text-decoration: none;'>127.</a> Zhvania MG, Chilachava LR, Japaridze NJ, Gelazonia LK, Lordkipanidze TG. Immediate and persisting effect of toluene chronic exposure on hippocampal cell loss in adolescent and adult rats. Brain Res Bull. 2012. 87: 187-92</p></div> </div></div>
  4. Momentum and Mission: A Year of Growth for the Spine Section

    Thu, 29 May 2025 18:08:30 -0000

    It is an exciting time for the AANS/CNS Section on Disorders of the Spine and Peripheral Nerves (DSPN). As the largest section in organized neurosurgery, we continue to build momentum through impactful education, advocacy, research support and cross-disciplinary collaboration. This past February, our annual Spine Summit in Tampa, Florida marked a record-breaking success. Under the leadership of Dr. Juan […]
    <p>It is an exciting time for the AANS/CNS Section on Disorders of the Spine and Peripheral Nerves (DSPN). As the largest section&nbsp;in organized neurosurgery, we continue to build momentum through impactful education, advocacy, research support and cross-disciplinary collaboration.</p> <p>This past February, our annual&nbsp;Spine Summit in Tampa, Florida&nbsp;marked a record-breaking success. Under the leadership of&nbsp;Dr. <strong>Juan Uribe</strong> (Section Chair),&nbsp;Drs. <strong>Dean Chou</strong> and <strong>Larry Lenke</strong> (Scientific Program Chairs) and&nbsp;Dr. <strong>Zack Ray</strong> (Annual Meeting Chair), the meeting welcomed an unprecedented number of attendees and industry partners. Corporate sponsorships, led by&nbsp;Dr. <strong>John Shin</strong>, exceeded expectations—reflecting both the strength of our educational programming and the growing interest in spine innovation. The meeting also welcomed more than 100 orthopedic spine colleagues, highlighting the evolving multidisciplinary nature of our field and initiatives like the Albert Scholars program.</p> <p>The scientific program was particularly strong, with over 1,500 abstracts submitted and thoughtful sessions covering degenerative spine disease, peripheral nerve, spine oncology and emerging technologies.&nbsp;Looking ahead, planning is already underway for our&nbsp;2026 meeting in Phoenix, Arizona. With&nbsp;Dr. <strong>Laura Snyder</strong> leading scientific content development as Scientific Program Chair, we’re excited to build a dynamic and forward-thinking program that reflects the evolving landscape of spine care. The meeting, themed&nbsp;“Beyond Limits: Elevating Quality in Spine Surgery”, will continue to advance our mission of innovation, collaboration and excellence. As Section Chair, I look forward to supporting the planning team and ensuring this remains a must-attend event for the spine and peripheral nerve community.</p> <p>The Section remains financially healthy and well-positioned for long-term sustainability. A newly formed Finance Committee, composed of&nbsp;Drs. <strong>Charles Sansur</strong>, <strong>Juan Uribe</strong>, <strong>Michael Wang</strong>, <strong>Dan Sciubba</strong> and <strong>Luis</strong> <strong>Tumialán</strong>, is exploring strategies to ensure continued growth and responsible stewardship of Section resources.</p> <p>Our partnership with the&nbsp;Neurosurgery Research &amp; Education Foundation (NREF)&nbsp;continues to expand. The Section now has over 20 spine-related&nbsp;Honor Your Mentor (HYM)&nbsp;funds, several of which are actively disbursing<ins> </ins><ins>funds</ins>. Notably, the&nbsp;<strong>Kevin Foley</strong>&nbsp;and&nbsp;<strong>Mark Shaffrey</strong>&nbsp;funds have seen strong engagement. These funds are instrumental in supporting mission-aligned research efforts, including collaborations with Spine CORe, our Quality Outcomes Database (QOD)&nbsp;and&nbsp;American Spine Registry (ASR) initiative, reinforcing the Section’s commitment to data-driven advancement in spine care.</p> <p>We are also proud to support and collaborate closely with the&nbsp;AANS Annual Scientific Meeting, where the Spine Section continues to play a central role in delivering high-quality, relevant content for both spine specialists and general neurosurgeons. Under the leadership of&nbsp;Dr. <strong>Anand Veeravagu</strong>&nbsp;and the Education Committee, the Section developed a dynamic lineup of sessions for this year’s meeting, including&nbsp;cervical spine debates,&nbsp;neuromonitoring dilemmas,&nbsp;complication-focused case discussions and&nbsp;rapid-fire abstract presentations&nbsp;in spine and peripheral nerve surgery. Additional highlights included the&nbsp;Kline Lecture&nbsp;in peripheral nerve and sessions devoted to&nbsp;leadership and career development.</p> <p>We also remain engaged in shaping spine content for the&nbsp;CNS Annual Meeting, where this year’s program will include a special symposium honoring the career and innovations of&nbsp;Dr. <strong>Richard Fessler</strong>. The CNS meeting will also feature spine-focused symposia and interactive sessions in areas such as&nbsp;spinal trauma,&nbsp;chronic low back pain and&nbsp;personalized spine surgery—further demonstrating the Section’s commitment to advancing spine care across all major academic forums.</p> <p>Our&nbsp;Media and Communications Committee, led by&nbsp;Dr. <strong>Cheerag Upadhyaya</strong>, has been working closely with the&nbsp;AANS marketing and communications team&nbsp;to enhance visibility across digital platforms. Regular strategy sessions have helped strengthen content coordination, social media engagement and meeting promotion. We’ve seen steady growth across LinkedIn, X and Instagram, and ongoing website updates continue to support increased traffic and user engagement. Efforts are also underway to explore additional multimedia channels and speaker toolkits to further enhance outreach and impact.</p> <p>Advocacy remains one of our top priorities. The&nbsp;Payor Response Committee, under the leadership of&nbsp;Dr. <strong>Yi Lu</strong>, continues to collaborate with the CPT team and&nbsp;Washington Committee&nbsp;to defend fair reimbursement and protect access to necessary spine procedures. Recent efforts include coordinated responses to denials of interbody fusion codes and addressing challenges related to procedural valuation. These initiatives reflect the Section’s commitment to representing the interests of spine surgeons nationally and ensuring our patients receive appropriate care.</p> <p>Internally, we’ve made strategic moves to strengthen continuity and long-term vision. The&nbsp;Strategic Planning Committee&nbsp;has transitioned to standing status, and a new&nbsp;Vice Chair role within the Scientific Program Committee&nbsp;will help ensure consistency and institutional memory across meetings. Our Section remains committed to inclusivity, with continued efforts to engage medical students, residents, APPs and early-career neurosurgeons.</p> <p>Finally, I’d like to thank the many volunteers, staff and partner organizations, whose collaboration and support have made this work possible. From advancing science and education to protecting our practice environment and growing the next generation of leaders, the Spine Section is proud to serve as a forward-looking, engaged community within organized neurosurgery.</p> <p>We look forward to continuing to elevate spine care together—<em>beyond limits.</em></p> <ul class="wp-block-social-links has-small-icon-size is-style-pill-shape is-layout-flex wp-block-social-links-is-layout-flex"><li class="wp-social-link wp-social-link-instagram wp-block-social-link"><a rel="noopener nofollow" target="_blank" href="https://www.instagram.com/spinesection/" class="wp-block-social-link-anchor"><svg width="24" height="24" viewBox="0 0 24 24" version="1.1" xmlns="http://www.w3.org/2000/svg" aria-hidden="true" focusable="false"><path d="M12,4.622c2.403,0,2.688,0.009,3.637,0.052c0.877,0.04,1.354,0.187,1.671,0.31c0.42,0.163,0.72,0.358,1.035,0.673 c0.315,0.315,0.51,0.615,0.673,1.035c0.123,0.317,0.27,0.794,0.31,1.671c0.043,0.949,0.052,1.234,0.052,3.637 s-0.009,2.688-0.052,3.637c-0.04,0.877-0.187,1.354-0.31,1.671c-0.163,0.42-0.358,0.72-0.673,1.035 c-0.315,0.315-0.615,0.51-1.035,0.673c-0.317,0.123-0.794,0.27-1.671,0.31c-0.949,0.043-1.233,0.052-3.637,0.052 s-2.688-0.009-3.637-0.052c-0.877-0.04-1.354-0.187-1.671-0.31c-0.42-0.163-0.72-0.358-1.035-0.673 c-0.315-0.315-0.51-0.615-0.673-1.035c-0.123-0.317-0.27-0.794-0.31-1.671C4.631,14.688,4.622,14.403,4.622,12 s0.009-2.688,0.052-3.637c0.04-0.877,0.187-1.354,0.31-1.671c0.163-0.42,0.358-0.72,0.673-1.035 c0.315-0.315,0.615-0.51,1.035-0.673c0.317-0.123,0.794-0.27,1.671-0.31C9.312,4.631,9.597,4.622,12,4.622 M12,3 C9.556,3,9.249,3.01,8.289,3.054C7.331,3.098,6.677,3.25,6.105,3.472C5.513,3.702,5.011,4.01,4.511,4.511 c-0.5,0.5-0.808,1.002-1.038,1.594C3.25,6.677,3.098,7.331,3.054,8.289C3.01,9.249,3,9.556,3,12c0,2.444,0.01,2.751,0.054,3.711 c0.044,0.958,0.196,1.612,0.418,2.185c0.23,0.592,0.538,1.094,1.038,1.594c0.5,0.5,1.002,0.808,1.594,1.038 c0.572,0.222,1.227,0.375,2.185,0.418C9.249,20.99,9.556,21,12,21s2.751-0.01,3.711-0.054c0.958-0.044,1.612-0.196,2.185-0.418 c0.592-0.23,1.094-0.538,1.594-1.038c0.5-0.5,0.808-1.002,1.038-1.594c0.222-0.572,0.375-1.227,0.418-2.185 C20.99,14.751,21,14.444,21,12s-0.01-2.751-0.054-3.711c-0.044-0.958-0.196-1.612-0.418-2.185c-0.23-0.592-0.538-1.094-1.038-1.594 c-0.5-0.5-1.002-0.808-1.594-1.038c-0.572-0.222-1.227-0.375-2.185-0.418C14.751,3.01,14.444,3,12,3L12,3z M12,7.378 c-2.552,0-4.622,2.069-4.622,4.622S9.448,16.622,12,16.622s4.622-2.069,4.622-4.622S14.552,7.378,12,7.378z M12,15 c-1.657,0-3-1.343-3-3s1.343-3,3-3s3,1.343,3,3S13.657,15,12,15z M16.804,6.116c-0.596,0-1.08,0.484-1.08,1.08 s0.484,1.08,1.08,1.08c0.596,0,1.08-0.484,1.08-1.08S17.401,6.116,16.804,6.116z"></path></svg><span class="wp-block-social-link-label screen-reader-text">Instagram</span></a></li> <li class="wp-social-link wp-social-link-x wp-block-social-link"><a rel="noopener nofollow" target="_blank" href="https://x.com/spinesection" class="wp-block-social-link-anchor"><svg width="24" height="24" viewBox="0 0 24 24" version="1.1" xmlns="http://www.w3.org/2000/svg" aria-hidden="true" focusable="false"><path d="M13.982 10.622 20.54 3h-1.554l-5.693 6.618L8.745 3H3.5l6.876 10.007L3.5 21h1.554l6.012-6.989L15.868 21h5.245l-7.131-10.378Zm-2.128 2.474-.697-.997-5.543-7.93H8l4.474 6.4.697.996 5.815 8.318h-2.387l-4.745-6.787Z" /></svg><span class="wp-block-social-link-label screen-reader-text">X</span></a></li> <li class="wp-social-link wp-social-link-linkedin wp-block-social-link"><a rel="noopener nofollow" target="_blank" href="https://www.linkedin.com/company/spinesection/" class="wp-block-social-link-anchor"><svg width="24" height="24" viewBox="0 0 24 24" version="1.1" xmlns="http://www.w3.org/2000/svg" aria-hidden="true" focusable="false"><path d="M19.7,3H4.3C3.582,3,3,3.582,3,4.3v15.4C3,20.418,3.582,21,4.3,21h15.4c0.718,0,1.3-0.582,1.3-1.3V4.3 C21,3.582,20.418,3,19.7,3z M8.339,18.338H5.667v-8.59h2.672V18.338z M7.004,8.574c-0.857,0-1.549-0.694-1.549-1.548 c0-0.855,0.691-1.548,1.549-1.548c0.854,0,1.547,0.694,1.547,1.548C8.551,7.881,7.858,8.574,7.004,8.574z M18.339,18.338h-2.669 v-4.177c0-0.996-0.017-2.278-1.387-2.278c-1.389,0-1.601,1.086-1.601,2.206v4.249h-2.667v-8.59h2.559v1.174h0.037 c0.356-0.675,1.227-1.387,2.526-1.387c2.703,0,3.203,1.779,3.203,4.092V18.338z"></path></svg><span class="wp-block-social-link-label screen-reader-text">LinkedIn</span></a></li></ul> <p></p>
  5. Central Nervous System Tuberculomas

    Thu, 08 May 2025 00:00:00 -0000

    A 57-year-old man with previously treated pulmonary tuberculosis presented with a 2-week history of neck pain, headache, and tingling in his hand. MRI of the head showed numerous small, peripherally enhancing nodules.
  6. Peripheral nerve surgery at risk: the consequences of NHS funding and governance reforms

    Fri, 25 Apr 2025 03:45:11 -0000

    Volume 39, Issue 3, June 2025, Page 287-288
    .
    <a href="/toc/ibjn20/39/3">Volume 39, Issue 3</a>, June 2025, Page 287-288<br/>. <br/>