[go: up one dir, main page]
More Web Proxy on the site http://driver.im/
You seem to have javascript disabled. Please note that many of the page functionalities won't work as expected without javascript enabled.
 
 
Sign in to use this feature.

Years

Between: -

Subjects

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Journals

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Article Types

Countries / Regions

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Search Results (6,859)

Search Parameters:
Keywords = tumor biomarkers

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
17 pages, 6091 KiB  
Article
Immunohistochemical Analysis of Inter-Alpha-Trypsin Inhibitor Heavy Chain 2 and Enolase 1 in Canine Mammary Tumors: Associations with Tumor Aggressiveness and Prognostic Significance
by Luadna dos Santos e Silva, Pedro Henrique Fogaça Jordão, Beatriz Castilho Balieiro, Laura de Souza Baracioli, Daniela Farias de Nóbrega, Adriana Alonso Novais, Luiz Gustavo de Almeida Chuffa and Debora Aparecida Pires de Campos Zuccari
Vet. Sci. 2025, 12(2), 110; https://doi.org/10.3390/vetsci12020110 (registering DOI) - 2 Feb 2025
Abstract
Mammary neoplasms in dogs are a common clinical concern, especially in middle-aged and older intact females. These tumors share similarities with human breast cancer in terms of histology, disease progression, and risk factors, making dogs a relevant model for breast cancer research. The [...] Read more.
Mammary neoplasms in dogs are a common clinical concern, especially in middle-aged and older intact females. These tumors share similarities with human breast cancer in terms of histology, disease progression, and risk factors, making dogs a relevant model for breast cancer research. The search for biomarkers in canine mammary tumors is essential to understand tumor progression and identify potential therapeutic targets. This study investigated the expression of two potential biomarkers—Inter-Alpha-Trypsin Inhibitor Heavy Chain 2 (ITIH2) and Enolase 1 (ENO1)—in the mammary glands of healthy and tumor-bearing dogs using immunohistochemistry. Both proteins were identified in previous proteomic analyses of extracellular vesicles derived from the plasma of healthy and tumor-bearing dogs. A total of fifty-one canine mammary tissue samples were analyzed and categorized into three groups: (i) the control group, composed of five samples of normal mammary tissue without neoplasia; (ii) benign tumors, composed of nineteen samples of benign mixed tumors; and (iii) malignant tumors, which included six carcinomas in grade 1 mixed tumors, five carcinomas in grade 2 mixed tumors, thirteen solid carcinomas of grade 3, one papillary carcinoma, and two tubular carcinomas. Regarding the intensity of staining, quantified by histoscore, there were no significant differences in the comparison between the groups; for ITIH2, the p-value was 0.33, and for ENO1, the p-value was 0.57. Regarding the predictive potential of their respective ROC curves, the proteins demonstrated low predictive power in canine mammary tumors. These findings indicate that neither ITIH2 nor ENO1 demonstrated strong prognostic value in this setting, as demonstrated by their moderate AUC values, wide confidence intervals, and lack of statistical significance. However, this study found distinct tissue localization patterns for ITIH2 and subcellular localization for ENO1. As an additional way to examine possible associations of these proteins with epithelial–mesenchymal transition, the ZEB1 antibody was tested by both single and double immunohistochemistry, demonstrating a tendency to be more intensely expressed in the malignant group and tending to be associated with ENO1 in canine mammary tumors. Full article
(This article belongs to the Special Issue Advanced Therapy in Companion Animals)
Show Figures

Figure 1

Figure 1
<p>Photomicrographs of the positive and negative controls of ITIH2, ENO1, and ZEB1. (<b>A</b>)—Positive control of ITIH2 (fallopian tube) and (<b>D</b>)—negative control (tonsil); (<b>B</b>)—Positive control of ENO1 (liver) and (<b>E</b>)—negative control (antibody suppression in dog mammary gland tissue); (<b>C</b>)—Positive control of ZEB1 (lymph node) and (<b>F</b>)—negative control (placenta).</p>
Full article ">Figure 2
<p>(<b>A</b>) Box plot illustrating age distribution across the malignant, benign, and control groups. <span class="html-italic">p</span>-values were calculated using ANOVA with Bonferroni post hoc tests. The black cross represents the mean age within each group. (<b>B</b>) Proportion representation of breeds within the malignant, benign, and control groups.</p>
Full article ">Figure 3
<p>Immunohistochemical staining of ITIH2. Control group (<b>A</b>–<b>C</b>); benign tumors (<b>D</b>–<b>F</b>); and malignant tumors (<b>G</b>–<b>I</b>). Photomicrographs at 20 µm showing weak or absent immunostaining in the cytoplasm.</p>
Full article ">Figure 4
<p>Immunohistochemical staining of ENO1. Control group (<b>A</b>–<b>C</b>); benign tumors (<b>D</b>–<b>F</b>); and malignant tumors (<b>G</b>–<b>I</b>). Photomicrographs at 20 µm showing strong immunostaining in nucleus, cytoplasm, and membrane (<b>I</b>).</p>
Full article ">Figure 5
<p>Protein expression (HistoScore) of ITIH2 (Inter-Alpha-Trypsin Inhibitor Heavy Chain 2) and ENO1 (Enolase 1). <span class="html-italic">p</span>-values for the Kruskal–Wallis test and Dunn’s post hoc test. The black cross represents the mean.</p>
Full article ">Figure 6
<p>Immunohistochemical localization of ENO1 across cellular compartments.</p>
Full article ">Figure 7
<p>Immunohistochemical comparison of ITIH2 (<b>1A</b>) and ZEB1 (<b>1B</b>) staining. The tissues in (<b>1A</b>) and (<b>1B</b>) are identical, differing only by the marker used. Tumor types and grades include carcinoma in mixed tumor grade 2 (<b>A</b>); carcinoma in mixed tumor grade 1 (<b>H</b>–<b>J</b>); tubular carcinoma (<b>C</b>); solid carcinoma grade 3 (<b>E</b>,<b>F</b>); and benign mixed tumor (<b>B</b>,<b>D</b>,<b>G</b>). Metastatic tumors are shown in (<b>A</b>,<b>C</b>,<b>E</b>).</p>
Full article ">Figure 8
<p>Double immunohistochemical staining showing ENO1 (magenta) and ZEB1 (brown) expression. Tumor types and grades include carcinoma in mixed tumor grade 2 (<b>A</b>); carcinoma in mixed tumor grade 1 (<b>H</b>–<b>J</b>); tubular carcinoma (<b>C</b>); solid carcinoma grade 3 (<b>E</b>,<b>F</b>); and benign mixed tumor (<b>B</b>,<b>D</b>,<b>G</b>).</p>
Full article ">Figure 9
<p>Roc curves of ITIH2 (Inter-Alpha-Trypsin Inhibitor Heavy Chain 2) and ENO1 (Enolase 1).</p>
Full article ">
20 pages, 3701 KiB  
Article
miRNA Signatures as Predictors of Therapy Response in Castration-Resistant Prostate Cancer: Insights from Clinical Liquid Biopsies and 3D Culture Models
by Jonathan Puente-Rivera, Stephanie I. Nuñez-Olvera, Verónica Fernández-Sánchez, Monica Alethia Cureño-Díaz, Erika Gómez-Zamora, Estibeyesbo Said Plascencia-Nieto, Elisa Elvira Figueroa-Angulo and María Elizbeth Alvarez-Sánchez
Genes 2025, 16(2), 180; https://doi.org/10.3390/genes16020180 (registering DOI) - 1 Feb 2025
Abstract
Background/Objectives: Prostate cancer (PCa) patients who do not respond to androgen deprivation therapy (ADT), referred to as castration-resistant prostate cancer (CRPC), remain a clinical challenge due to confirm the aggressive nature of CRPC and its resistance to conventional therapies. This study aims to [...] Read more.
Background/Objectives: Prostate cancer (PCa) patients who do not respond to androgen deprivation therapy (ADT), referred to as castration-resistant prostate cancer (CRPC), remain a clinical challenge due to confirm the aggressive nature of CRPC and its resistance to conventional therapies. This study aims to investigate the potential of microRNAs (miRNAs) as biomarkers for predicting therapeutic response in CRPC patients. Methods: We performed miRNA and mRNA expression analyses using publicly available datasets and applied 3D cell culture models to replicate more physiologically relevant tumor conditions. Genetic analysis techniques were employed on publicly available data, and expression profiles from 3D cell culture models were examined. Results: Eighteen miRNAs with differential expression were identified between patients who responded favorably to abiraterone therapy (responders) and those with advanced CRPC (non-responders). Specifically, miRNAs such as hsa-miR-152-3p and hsa-miR-34a-3p were found to be associated with critical pathways, including TGF-β signaling and P53, which are linked to therapeutic resistance. Several miRNAs were identified as potential predictors of treatment efficacy, including therapies like abiraterone. Conclusions: These results indicate that miRNAs could serve as non-invasive biomarkers for predicting therapeutic outcomes, facilitating a more personalized approach to CRPC treatment. This study provides a novel perspective on treatment strategies for CRPC, emphasizing the role of miRNAs in improving therapeutic precision and efficacy in this complex disease. Full article
(This article belongs to the Special Issue MicroRNA in Cancers)
Show Figures

Figure 1

Figure 1
<p>Study workflow for identifying miRNA predictors of therapy response in PCa. Diagram workflow illustrates the multi-step approach used in this study. (1) Data integration and analysis were performed using public PCa databases, including tissue and blood samples from responders, CRPC patients, and BPH cases, to identify common and differential miRNA profiles. (2) PC3 and DU145 PCa cell lines were cultured in both 3D and 2D models to analyze differential miRNA expression profiles and validate their roles in therapeutic resistance. (3) Liquid biopsy samples from ADT-treated PCa patients, categorized as responders, CRPC patients, and CTRL, were analyzed to correlate Gleason scores and miRNA expression. Differential miRNA signatures were correlated with therapy response, generating potential biomarkers to predict therapeutic outcomes in PCa. Purple dotted arrows represent the convergence of in silico data mining, experimental validation, and patient-derived samples. Upward green arrows indicate miRNA overexpression, while downward red arrows represent downregulation. Created in <a href="https://BioRender.com" target="_blank">https://BioRender.com</a> (accessed on 23 January 2025).</p>
Full article ">Figure 2
<p>miRNA expression profiles in non-responders CRPC datasets. (<b>A</b>,<b>B</b>) Volcano plot datasets of miRNAs with differential expression in CRPC vs. primary prostate tumors and BPH (fold change &gt; 1.5 and <span class="html-italic">p</span>-value 0.05). (<b>C</b>) Volcano plot dataset of miRNAs plasma miRNAs of responder versus non-responder metastatic castration-resistant PCa patients to abiraterone acetate (fold change &gt; 1.5, <span class="html-italic">p</span>-value of 0.05). (<b>D</b>) Common miRNAs across the three datasets. (<b>E</b>) Heatmap of common miRNAs in non-responders vs. responders (average linkage based on Euclidean distance measurements). Aqua blue color represents downregulation, and pink represents upregulation.</p>
Full article ">Figure 3
<p>miRNA expression profiles and targets in responder patients and identified pathways associated. (<b>A</b>) Pathways and biological processes related to miRNAs downregulated in responder patients. (<b>B</b>) Pathways and biological processes related to upregulated miRNAs in responder PCa patients. (<b>C</b>) Volcano plot datasets of mRNAs with differential expression in 3D culture vs. 2D culture from PC3 cells (fold change &gt; 1.5, <span class="html-italic">p</span>-value 0.05). (<b>D</b>) miRNA network regulation of top 50 genes upregulated in metastatic vs. primary PCa tumors from GSEA signature. (<b>E</b>) miRNA network regulation of top 50 genes upregulated in metastatic vs. primary PCa tumors from GSEA signature. (<b>F</b>) Representative 3D cell cultures of DU145 and PC3 cells observed by optical microscopy (40×) after 6 days of incubation over Matrigel. Bar represents 150 nm. Relative hsa-miR-411-3p (<b>G</b>), miR-34a-p (<b>H</b>), miR-152-3p (<b>I</b>), and miR-654a-3p (<b>J</b>) expression in 2D and 3D cell lines cultures of DU145 and PC3, respectively. The relative levels of miRNAs were obtained by qRT-PCR and calculated using 2<sup>−ΔΔCt</sup>. The box-plot graph indicates the median with quartiles, and error bars represent ± SD (two independent experiments by triplicate for each cell culture condition). A <span class="html-italic">t</span>-test and Tukey test were used for comparison; *** indicates <span class="html-italic">p</span> &lt; 0.001, ** indicates <span class="html-italic">p</span> &lt; 0.01. snRNU6 expression levels were used for the normalization of data.</p>
Full article ">Figure 4
<p>miRNAs expression profiles by Gleason score from PRAD-TGCA. Expression profile of miRNAa in patient tissue. Profile expression obtained from the TCGA-PRAD database through the Xena Browser comparing normal tissue and PCa tissue. (<b>A</b>) hsa-miR-411, (<b>B</b>) hsa-miR-654, (<b>C</b>) hsa-miR-152, (<b>D</b>) hsa-miR-345, (<b>E</b>) hsa-miR-629, and (<b>F</b>) hsa-miR-34a. Mean comparison was performed by ANOVA Tukey test. A fold change of 1.5 and a <span class="html-italic">p</span>-value of &lt;0.05 were used as cut-off values.</p>
Full article ">Figure 5
<p>miRNA expression profiles and correlation with Gleason score progression and aggressiveness from PRAD-TCGA. Comparison of expression miRNA levels of responder and non-responder PCa patients of (<b>A</b>) has-miR-152-3p, (<b>B</b>) has-miR-411-5p, (<b>C</b>) has-miR-34a-3p, and (<b>D</b>) hsa-miR-629-3p. The length of the bars represents the miRNA expression levels. Error bars represent ± SD, and mean comparison using a <span class="html-italic">t</span>-test was used to generate the <span class="html-italic">p</span>-values with a significance threshold set at 0.05. Evaluation of ROC curves and AUC in responder and non-responder patients. ROC and AUC values from (<b>E</b>) hsa-miR-152-3p, (<b>F</b>) hsa-miR-411-5p, (<b>G</b>) hsa-miR-34a-5p, and (<b>H</b>) hsa-miR-629-3p were calculated to assess the feasibility of using miRNA levels from patients as a potential diagnostic tool, highlighting significant differences that may correlate with PCa.</p>
Full article ">Figure 6
<p>Relative expression of selected miRNAs (<b>A</b>) has-miR-34a-3p, (<b>B</b>) has-miR-152-3p, (<b>C</b>) hsa-miR-411-5p, and (<b>D</b>) hsa-miR-629-3p in serum from responder (NCRPC) and non-responder (CRPC) PCa patients. miRNAs relative expression levels were determined by qRT-PCR and calculated with 2<sup>−ΔΔCt</sup>. The box-plot graph shows the median with quartile. ±SD are represented by error bars (two independent experiments by technical triplicate). Mean comparison using ANOVA test. ***: <span class="html-italic">p</span> &lt; 0.001. ns: not statically significance. The normalization of miRNA levels was performed with snRNU6.</p>
Full article ">
16 pages, 1006 KiB  
Review
Why Do Glioblastoma Treatments Fail?
by Alen Rončević, Nenad Koruga, Anamarija Soldo Koruga and Robert Rončević
Future Pharmacol. 2025, 5(1), 7; https://doi.org/10.3390/futurepharmacol5010007 (registering DOI) - 1 Feb 2025
Viewed by 100
Abstract
Glioblastoma (GBM) is the most aggressive brain tumor, characterized by high recurrence rates and poor patient outcomes. Treatment failure is driven by multiple factors, including complex tumor heterogeneity, the presence of cancer stem cells, the immunosuppressive tumor microenvironment (TME), and many others. GBM’s [...] Read more.
Glioblastoma (GBM) is the most aggressive brain tumor, characterized by high recurrence rates and poor patient outcomes. Treatment failure is driven by multiple factors, including complex tumor heterogeneity, the presence of cancer stem cells, the immunosuppressive tumor microenvironment (TME), and many others. GBM’s heterogeneity underlines its ability to resist therapies and adapt to the TME. The TME, which is highly immunosuppressive and shaped by hypoxia, impairs anti-tumor immunity and limits the efficacy of immunotherapy. The blood–brain barrier (BBB) remains a major obstacle to delivering sufficient drug concentrations to the tumor by restricting the penetration of therapeutic agents. Another problem is the lack of reliable biomarkers to perform better patient stratification or even guide personalized treatments, resulting in generalized therapeutic approaches that do not adequately address GBM complexities. This review highlights the multifactorial nature of GBM treatment failure and highlights the need for a paradigm shift and innovative, personalized strategies. A deeper understanding of tumor biology and advances in translational research will be crucial to developing effective therapies and improving patient outcomes in this devastating disease. Full article
(This article belongs to the Special Issue Feature Papers in Future Pharmacology 2024)
Show Figures

Figure 1

Figure 1
<p>Regional heterogeneity of the blood–brain barrier in glioblastoma.</p>
Full article ">Figure 2
<p>Glioblastoma treatment failure as a result of many interacting factors.</p>
Full article ">
22 pages, 771 KiB  
Review
Targeting the PD-1/PD-L1 Signaling Pathway for Cancer Therapy: Focus on Biomarkers
by Areti Strati, Christos Adamopoulos, Ioannis Kotsantis, Amanda Psyrri, Evi Lianidou and Athanasios G. Papavassiliou
Int. J. Mol. Sci. 2025, 26(3), 1235; https://doi.org/10.3390/ijms26031235 - 31 Jan 2025
Viewed by 534
Abstract
The PD1/PD-L1 axis plays an important immunosuppressive role during the T-cell-mediated immune response, which is essential for the physiological homeostasis of the immune system. The biology of the immunological microenvironment is extremely complex and crucial for the development of treatment strategies for immunotherapy. [...] Read more.
The PD1/PD-L1 axis plays an important immunosuppressive role during the T-cell-mediated immune response, which is essential for the physiological homeostasis of the immune system. The biology of the immunological microenvironment is extremely complex and crucial for the development of treatment strategies for immunotherapy. Characterization of the immunological, genomic or transcriptomic landscape of cancer patients could allow discrimination between responders and non-responders to anti-PD-1/PD-L1 therapy. Immune checkpoint inhibitor (ICI) therapy has shown remarkable efficacy in a variety of malignancies in landmark trials and has fundamentally changed cancer therapy. Current research focuses on strategies to maximize patient selection for therapy, clarify mechanisms of resistance, improve existing biomarkers, including PD-L1 expression and tumor mutational burden (TMB), and discover new biomarkers. In this review, we focus on the function of the PD-1/PD-L1 signaling pathway and discuss the immunological, genomic, epigenetic and transcriptomic landscape in cancer patients receiving anti-PD-1/PD-L1 therapy. Finally, we provide an overview of the clinical trials testing the efficacy of antibodies against PD-1/PD-L1. Full article
(This article belongs to the Special Issue Immunotherapy: A New Perspective in Cancer Treatment)
Show Figures

Figure 1

Figure 1
<p>Immunological, genomic, epigenetic and transcriptomic landscape in cancer patients receiving anti-PD-1/PD-L1 therapy.</p>
Full article ">
16 pages, 2370 KiB  
Article
Connexin 43 Expression as Biomarker of Oral Squamous Cell Carcinoma and Its Association with Human Papillomavirus 16 and 18
by Jose Roberto Gutierrez-Camacho, Lorena Avila-Carrasco, Idalia Garza-Veloz, Joel Monárrez-Espino, Maria Calixta Martinez-Vazquez, Roxana Araujo-Espino, Perla M. Trejo-Ortiz, Rosa B. Martinez-Flores, Reinaldo Gurrola-Carlos, Lorena Troncoso-Vazquez and Margarita L. Martinez-Fierro
Int. J. Mol. Sci. 2025, 26(3), 1232; https://doi.org/10.3390/ijms26031232 - 30 Jan 2025
Viewed by 315
Abstract
Oral squamous cell carcinoma (OSCC) is the main form of head and neck cancer. Gap junctions (GJs) are communication channels involved in cell proliferation control; they consist of hemichannels formed by connexin (Cx) proteins. The abnormal expression/function of Cx43 has been associated with [...] Read more.
Oral squamous cell carcinoma (OSCC) is the main form of head and neck cancer. Gap junctions (GJs) are communication channels involved in cell proliferation control; they consist of hemichannels formed by connexin (Cx) proteins. The abnormal expression/function of Cx43 has been associated with tumor progression. Also, some human papillomaviruses (HPVs) have been linked to squamous cell cancer. Therefore, this study aimed at assessing Cx43 as a potential OSCC biomarker and exploring its association with histopathological differentiation and HPV infection. OSCC samples were inspected using hematoxylin and eosin staining, and Cx43 expression and HPV 16/18 were tested by immunofluorescence. Pearson correlation tests, ANOVA, and Kaplan–Meier curves were used in the analysis. Samples from 39 patients with OSCC were studied. Most had well-differentiated histology and 61.5% were HPV+. Cx43 expression was significantly associated with HPV infection (p = 0.047), differentiation (p < 0.001), and survival (p = 0.009), and HPV positivity was also associated with the degree of differentiation (p = 0.012). Cx43 shows potential as a prognostic biomarker for OSCC. Lower Cx43 expression, correlated with poorer differentiation, is associated with an unfavorable prognosis. Further studies are needed to confirm its clinical utility. Full article
37 pages, 2658 KiB  
Review
Long Non-Coding RNAs in Ovarian Cancer: Mechanistic Insights and Clinical Applications
by Sneha Basu, Revathy Nadhan and Danny N. Dhanasekaran
Cancers 2025, 17(3), 472; https://doi.org/10.3390/cancers17030472 - 30 Jan 2025
Viewed by 343
Abstract
Background/Objectives: Ovarian cancer is a leading cause of gynecological cancer mortality worldwide, often diagnosed at advanced stages due to vague symptoms and the lack of effective early detection methods. Long non-coding RNAs (lncRNAs) have emerged as key regulators in cancer biology, influencing cellular [...] Read more.
Background/Objectives: Ovarian cancer is a leading cause of gynecological cancer mortality worldwide, often diagnosed at advanced stages due to vague symptoms and the lack of effective early detection methods. Long non-coding RNAs (lncRNAs) have emerged as key regulators in cancer biology, influencing cellular processes such as proliferation, apoptosis, and chemoresistance. This review explores the multifaceted roles of lncRNAs in ovarian cancer pathogenesis and their potential as biomarkers and therapeutic targets. Methods: A comprehensive literature review was conducted to analyze the structural and functional characteristics of lncRNAs and their contributions to ovarian cancer biology. This includes their regulatory mechanisms, interactions with signaling pathways, and implications for therapeutic resistance. Advanced bioinformatics and omics approaches were also evaluated for their potential in lncRNA research. Results: The review highlights the dual role of lncRNAs as oncogenes and tumor suppressors, modulating processes such as cell proliferation, invasion, and angiogenesis. Specific lncRNAs, such as HOTAIR and GAS5, demonstrate significant potential as diagnostic biomarkers and therapeutic targets. Emerging technologies, such as single-cell sequencing, provide valuable insights into the tumor microenvironment and the heterogeneity of lncRNA expression. Conclusions: LncRNAs hold transformative potential in advancing ovarian cancer diagnosis, prognosis, and treatment. Targeting lncRNAs or their associated pathways offers promising strategies to overcome therapy resistance and enhance personalized medicine. Continued research integrating omics and bioinformatics will be essential to unlock the full clinical potential of lncRNAs in ovarian cancer management. Full article
(This article belongs to the Special Issue Insights from the Editorial Board Member)
Show Figures

Figure 1

Figure 1
<p>Classification tree for non-coding RNAs. Non-coding RNAs can be classified into three major groups, namely the small non-coding RNAs that are less than 200 nt in length, the long non-coding RNAs (lncRNAs) that are greater than 200 nt in length, and the specific subclass of regulatory non-coding RNAs. The smaller non-coding RNAs include the miRNA, piRNA, snoRNA, and other snRNAs. The regulatory non-coding RNAs encompasses the tsRNA, tRNA, and rRNA. The lncRNAs can be classified into sense, antisense, bidirectional, intergenic, and intronic based on their position in the genome. Based on the functional classification, lncRNAs can be either guide, cis-/trans-acting, imprinted, scaffold, circular, competing endogenous, spliceosome-associated, enhancer-associated, or promoter-associated lncRNAs. (ceRNA: competing endogenous lncRNA; e-lncRNA: enhancer-associated lncRNA; miRNA: microRNA; onco-lncRNA: oncogenic lncRNA; p-lncRNA: promoter-associated lncRNA; piRNA: piwi interacting RNA; rRNA: ribosomal RNA; SA-lncRNA: spliceosome-associated lncRNA; snRNA: small nuclear RNA; snoRNA: small nucleolar RNA; tRNA: transfer RNA; tsRNA: transfer RNA derived small RNAs; TS-lncRNA: tumor-suppressor lncRNA).</p>
Full article ">Figure 2
<p>Structural differences in lncRNAs across the three levels: primary, secondary, and tertiary. The primary structure of lncRNA includes a linear stretch of nucleotides and splice variants of the same lncRNA, showcasing sequence diversity. The secondary structure includes a triple helix, hairpin loop, G-quadruplex, stem-loop, and four-way junction demonstrating the complex folding patterns of the primary lncRNA. The tertiary structure is depicted with an example: the lncRNA TERRA, which forms a complex G-quadruplex to interact with the telomeric-repeat binding factor 2 (TRF2) to confer stability to the telomeres.</p>
Full article ">Figure 3
<p>Genomic classifications of lncRNAs. LncRNAs can be classified on the basis of their origin in the genome. (<b>A</b>) Intronic LncRNA: transcribed from the intronic region of a transcript; (<b>B</b>) Sense lncRNA and antisense lncRNA: transcribed from the sense and antisense strand, respectively; (<b>C</b>) Intergenic lncRNA: transcribed from the region between two genes; (<b>D</b>) Bidirectional lncRNA: transcribed from the promoters of coding genes but in an opposite direction. (LncRNAs—long non-coding RNAs).</p>
Full article ">Figure 4
<p>Functional diversity of lncRNAs. In the cytoplasm, lncRNAs function as (<b>A</b>) imprinted lncRNAs, (<b>B</b>) scaffold lncRNAs, and (<b>C</b>) ceRNA, showcasing their diverse regulatory mechanisms throughout the cell. Within the nucleus, the lncRNAs can (<b>D</b>) act as a guide lncRNA, function either as (<b>E</b>) trans- or (<b>F</b>) cis-regulatory elements, or can be associated with (<b>G</b>) the spliceosome complex, (<b>H</b>) promoters, or (<b>I</b>) enhancers.</p>
Full article ">Figure 5
<p>Functional roles of lncRNAs in ovarian cancer. LncRNAs are critically involved in cell proliferation, cell survival, angiogenesis, stemness, metabolic reprogramming, therapy resistance, cell migration, and, immune evasion in ovarian cancer. Examples of lncRNAs that induce these processes and act as oncogenes in OC are highlighted in red boxes, whereas those that tend to reduce these processes are highlighted in green boxes.</p>
Full article ">Figure 6
<p>Roadmap of lncRNA research in ovarian cancer. The roadmap can be divided in three main phases: The discovery phase involves identifying dysregulated lncRNAs through RNA sequencing (RNA-seq) and bioinformatics tools, as shown by volcano plots and clustering analyses. The functional characterization phase includes validating lncRNAs as biomarkers and elucidating their roles through in vitro functional assays using tumor cell lines and in vivo studies in animal models, linking lncRNA expression to patient survival. Finally, the clinical translation phase highlights therapeutic strategies, such as nanoparticle- or exosome-based delivery of antisense oligonucleotides (ASOs) and siRNAs, and the development of small-molecule inhibitors targeting specific lncRNAs for clinical applications.</p>
Full article ">
20 pages, 2063 KiB  
Article
Diagnostic and Prognostic Significance of a Four-miRNA Signature in Colorectal Cancer
by Giuseppe Gattuso, Federica Longo, Graziana Spoto, Daria Ricci, Alessandro Lavoro, Saverio Candido, Antonio Di Cataldo, Giuseppe Broggi, Lucia Salvatorelli, Gaetano Magro, Massimo Libra and Luca Falzone
Int. J. Mol. Sci. 2025, 26(3), 1219; https://doi.org/10.3390/ijms26031219 - 30 Jan 2025
Viewed by 232
Abstract
Colorectal cancer (CRC) is the fourth most commonly diagnosed cancer and one of the leading causes of cancer death worldwide. Despite diagnostic and therapeutic advances, CRC mortality remains high, especially in industrialized countries. Numerous studies have highlighted the pathogenetic role of altered microRNA [...] Read more.
Colorectal cancer (CRC) is the fourth most commonly diagnosed cancer and one of the leading causes of cancer death worldwide. Despite diagnostic and therapeutic advances, CRC mortality remains high, especially in industrialized countries. Numerous studies have highlighted the pathogenetic role of altered microRNA (miRNA) expression among the various factors contributing to the development and progression of colorectal cancer (CRC). However, the data regarding specific miRNAs involved in CRC pathogenesis remain inconsistent, and no miRNAs have been recognized so far as reliable or effective biomarkers for the diagnosis of this tumor type. To identify novel miRNA biomarkers in CRC, this study validated the expression levels of a four-miRNA signature predicted to be involved in CRC by analyzing both tissue and liquid biopsy samples. Our experimental and bioinformatics results highlighted the diagnostic potential of hsa-miR-21-5p, hsa-miR-503-5p, and hsa-miR-375, as well as the potential prognostic value of hsa-miR-497-5p overexpression and hsa-miR-375-3p downregulation. Overall, the results obtained suggest the diagnostic and prognostic significance of this four-miRNA signature in CRC. Full article
18 pages, 981 KiB  
Article
Radiomics-Based Prediction of Treatment Response to TRuC-T Cell Therapy in Patients with Mesothelioma: A Pilot Study
by Hubert Beaumont, Antoine Iannessi, Alexandre Thinnes, Sebastien Jacques and Alfonso Quintás-Cardama
Cancers 2025, 17(3), 463; https://doi.org/10.3390/cancers17030463 - 29 Jan 2025
Viewed by 362
Abstract
Background/Objectives: T cell receptor fusion constructs (TRuCs), a next generation engineered T cell therapy, hold great promise. To accelerate the clinical development of these therapies, improving patient selection is a crucial pathway forward. Methods: We retrospectively analyzed 23 mesothelioma patients (85 target tumors) [...] Read more.
Background/Objectives: T cell receptor fusion constructs (TRuCs), a next generation engineered T cell therapy, hold great promise. To accelerate the clinical development of these therapies, improving patient selection is a crucial pathway forward. Methods: We retrospectively analyzed 23 mesothelioma patients (85 target tumors) treated in a phase 1/2 single arm clinical trial (NCT03907852). Five imaging sites were involved, the settings for the evaluations were Blinded Independent Central Reviews (BICRs) with double reads. The reproducibility of 3416 radiomics and delta-radiomics (Δradiomics) was assessed. The univariate analysis evaluated correlations at the target tumor level with (1) tumor diameter response; (2) tumor volume response, according to the Quantitative Imaging Biomarker Alliance; and (3) the mean standard uptake value (SUV) response, as defined by the positron emission tomography response criteria in solid tumors (PERCISTs). A random forest model predicted the response of the target pleural tumors. Results: Tumor anatomical distribution was 55.3%, 17.6%, 14.1%, and 10.6% in the pleura, lymph nodes, peritoneum, and soft tissues, respectively. Radiomics/Δradiomics reproducibility differed across tumor localizations. Radiomics were more reproducible than Δradiomics. In the univariate analysis, none of the radiomics/Δradiomics correlated with any response criteria. With an accuracy ranging from 0.75 to 0.9, three radiomics/Δradiomics were able to predict the response of target pleural tumors. Pivotal studies will require a sample size of 250 to 400 tumors. Conclusions: The prediction of responding target pleural tumors can be achieved using a machine learning-based radiomics/Δradiomics analysis. Tumor-specific reproducibility and the average values indicated that using tumor models to create an effective patient model would require combining several target tumor models. Full article
(This article belongs to the Special Issue Biomarkers and Targeted Therapy in Malignant Pleural Mesothelioma)
15 pages, 1200 KiB  
Review
Long Intergenic Non-Coding RNAs and BRCA1 in Breast Cancer Pathogenesis: Neighboring Companions or Nemeses?
by Olalekan Olatunde Fadebi, Thabiso Victor Miya, Richard Khanyile, Zodwa Dlamini and Rahaba Marima
Non-Coding RNA 2025, 11(1), 9; https://doi.org/10.3390/ncrna11010009 - 29 Jan 2025
Viewed by 347
Abstract
Breast cancer is one of the leading causes of mortality among women, primarily due to its complex molecular landscape and heterogeneous nature. The tendency of breast cancer patients to develop metastases poses significant challenges in clinical management. Notably, mutations in the breast cancer [...] Read more.
Breast cancer is one of the leading causes of mortality among women, primarily due to its complex molecular landscape and heterogeneous nature. The tendency of breast cancer patients to develop metastases poses significant challenges in clinical management. Notably, mutations in the breast cancer gene 1 (BRCA1) significantly elevate breast cancer risk. The current research endeavors employ diverse molecular approaches, including RNA, DNA, and protein studies, to explore avenues for the early diagnosis and treatment of breast cancer. Recent attention has shifted towards long non-coding RNAs (lncRNAs) as promising diagnostic, prognostic, and therapeutic targets in the multifaceted progression of breast cancer. Among these, long intergenic non-coding RNAs (lincRNAs), a specific class of lncRNAs, play critical roles in regulating various aspects of tumorigenesis, including cell proliferation, apoptosis, epigenetic modulation, tumor invasion, and metastasis. Their distinctive expression patterns in cellular and tissue contexts underscore their importance in breast cancer development and progression. Harnessing lincRNAs’ sensitivity and precision as diagnostic, therapeutic, and prognostic markers holds significant promise for the clinical management of breast cancer. However, the potential of lincRNAs remains relatively underexplored, particularly in the context of BRCA1-mutated breast cancer and other clinicopathological parameters such as receptor status and patient survival. Consequently, there is an urgent need for comprehensive investigations into novel diagnostic and prognostic breast cancer biomarkers. This review examines the roles of lincRNAs associated with BRCA1 in the landscape of breast cancer, highlighting the potential avenues for future research and clinical applications. Full article
Show Figures

Figure 1

Figure 1
<p>Estimated cancer incidence and mortality rates worldwide in 2022. Data Source: GLOBOCAN 2022, Graph Production: Global Cancer Observatory <a href="http://gco.iarc.fr" target="_blank">http://gco.iarc.fr</a> (accessed on 8 October 2024). International Agency for Research on Cancer.</p>
Full article ">Figure 2
<p>(<b>a</b>) Main roles of <span class="html-italic">BRCA1</span>. An overview of <span class="html-italic">BRCA1</span>’<span class="html-italic">s</span> physiological functions in HR, repair during the DNA-replication recruitment of DNA damage site, G1/MS-phase checkpoint regulation, and DNA end resection. (<b>b</b>) <span class="html-italic">BRCA1</span>-mutated breast cancer associated with TNBC based on their expression of hormone receptors (ER, PR, and HER2). The image was self-created with BioRender.com.</p>
Full article ">Figure 3
<p>Regulation of the pRB and p53 pathways by the lincRNAs <span class="html-italic">ANRIL</span>, <span class="html-italic">lincRNA-p21</span>, and <span class="html-italic">lincRNA-ROR</span>. These lincRNAs regulate the pRB and p53 pathways by modulating the expression of CDK inhibitors p15 and p16, which impact pRB activity. <span class="html-italic">ANRIL</span> acts as a coregulator by binding to polycomb proteins. In response to DNA damage, p53 produces <span class="html-italic">lincRNA-p21</span>, <span class="html-italic">lincRNA-ROR</span>, and <span class="html-italic">PANDA</span> to regulate apoptosis. The image was self-created using BioRender.com.</p>
Full article ">
14 pages, 789 KiB  
Review
Understanding microRNA-Mediated Chemoresistance in Colorectal Cancer Treatment
by Guillermo Valenzuela, Héctor R. Contreras, Katherine Marcelain, Mauricio Burotto and Jaime González-Montero
Int. J. Mol. Sci. 2025, 26(3), 1168; https://doi.org/10.3390/ijms26031168 - 29 Jan 2025
Viewed by 329
Abstract
Colorectal cancer (CRC) remains the second most lethal cancer worldwide, with incidence rates expected to rise substantially by 2040. Although biomarker-driven therapies have improved treatment, responses to standard chemotherapeutics, such as 5-fluorouracil (5-FU), oxaliplatin, and irinotecan, vary considerably. This clinical heterogeneity emphasizes the [...] Read more.
Colorectal cancer (CRC) remains the second most lethal cancer worldwide, with incidence rates expected to rise substantially by 2040. Although biomarker-driven therapies have improved treatment, responses to standard chemotherapeutics, such as 5-fluorouracil (5-FU), oxaliplatin, and irinotecan, vary considerably. This clinical heterogeneity emphasizes the urgent need for novel biomarkers that can guide therapeutic decisions and overcome chemoresistance. microRNAs (miRNAs) have emerged as key post-transcriptional regulators that critically influence chemotherapy responses. miRNAs orchestrate post-transcriptional gene regulation and modulate diverse pathways linked to chemoresistance. They influence drug transport by regulating ABC transporters and affect metabolic enzymes like thymidylate synthase (TYMS). These activities shape responses to standard CRC chemotherapy agents. Furthermore, miRNAs can regulate the epithelial–mesenchymal transition (EMT). The miR-200 family (e.g., miR-200c and miR-141) can reverse EMT phenotypes, restoring chemosensitivity. Additionally, miRNAs like miR-19a and miR-625-3p show predictive value for chemotherapy outcomes. Despite these promising findings, the clinical translation of miRNA-based biomarkers faces challenges, including methodological inconsistencies and the dynamic nature of miRNA expression, influenced by the tumor microenvironment. This review highlights the critical role of miRNAs in elucidating chemoresistance mechanisms and their promise as biomarkers and therapeutic targets in CRC, paving the way for a new era of precision oncology. Full article
(This article belongs to the Special Issue Molecular Mechanisms and Therapies of Colorectal Cancer: 4th Edition)
Show Figures

Figure 1

Figure 1
<p>miRNA-associated mechanisms of resistance to 5-fluorouracil, oxaliplatin, and irinotecan. Each circle represents a key regulatory site modulated by a microRNA that promotes either chemosensitivity (green circles) or chemoresistance (red circles).</p>
Full article ">
15 pages, 2273 KiB  
Article
A Pilot Study on Qualitative Metabolomics to Characterize Lewis Lung Carcinoma in Mice
by Agnieszka Stawarska, Magdalena Bamburowicz-Klimkowska, Dariusz Maciej Pisklak, Maciej Gawlak and Ireneusz P. Grudzinski
Life 2025, 15(2), 202; https://doi.org/10.3390/life15020202 - 29 Jan 2025
Viewed by 550
Abstract
Metabolomics is a powerful tool that can be used to identify different stages in cancer development. In this study, the metabolomic profile of Lewis lung carcinoma (LLC) was characterized in C57BL/6 mice bearing LLC tumors. Magnetic resonance spectroscopy (nuclear magnetic resonance—NMR) was applied [...] Read more.
Metabolomics is a powerful tool that can be used to identify different stages in cancer development. In this study, the metabolomic profile of Lewis lung carcinoma (LLC) was characterized in C57BL/6 mice bearing LLC tumors. Magnetic resonance spectroscopy (nuclear magnetic resonance—NMR) was applied using a 400 MHz 1H NMR spectrometer. Two types of metabolites (polar and non-polar) were identified on LLC based on the analysis of methanol/water and chloroform extracts collected from lung cancer samples in mice. The investigated metabolomics show that the neoplastic processes of growing LLC on mice may affect carbohydrate; alanine and glutamate; leucine and isoleucine; lysine; creatine; and choline metabolism, whereas hypoxia states were identified due to elevated lactate in lung cancer tissues. The metabolomic profile of Lewis lung carcinoma could be considered to be a valuable biomarker in translational lung cancer research. Full article
(This article belongs to the Section Physiology and Pathology)
Show Figures

Figure 1

Figure 1
<p>A schematic diagram illustrating the experimental workflow.</p>
Full article ">Figure 2
<p>Representative <sup>1</sup>H NMR spectra of chloroform extracts of LLC tumors in mice. <sup>1</sup>H NMR was applied using 400 MHz NMR system.</p>
Full article ">Figure 3
<p>Representative <sup>1</sup>H NMR spectra of water/methanol extract of LLC tumors in mice. Aliphatic region of <sup>1</sup>H NMR spectra of water/methanol extract with assigned signals (Lac—lactate; Ala—alanine; Cho—choline; Cr—creatine; Glu—glutamine; glutamate; sIn—scyllo-inositol; Leu—leucine; Ile—isoleucine; Lys—lysine; Ac—acetate). <sup>1</sup>H NMR was applied using 400 MHz NMR system.</p>
Full article ">Figure 4
<p>High-field region of <sup>1</sup>H NMR spectrum of water/methanol extract with assigned signals. (Tyr—tyrosine; His—histidine; Phe—phenylalanine; Ade—adenine; For—formate; Ino—inosine). <sup>1</sup>H NMR was applied using 400 MHz NMR system.</p>
Full article ">
23 pages, 2689 KiB  
Review
Dual Biomarker Strategies for Liquid Biopsy: Integrating Circulating Tumor Cells and Circulating Tumor DNA for Enhanced Tumor Monitoring
by Ga Young Moon, Basak Dalkiran, Hyun Sung Park, Dongjun Shin, Chaeyeon Son, Jung Hyun Choi, Seha Bang, Hosu Lee, Il Doh, Dong Hyung Kim, Woo-jin Jeong and Jiyoon Bu
Biosensors 2025, 15(2), 74; https://doi.org/10.3390/bios15020074 - 28 Jan 2025
Viewed by 359
Abstract
The liquid biopsy has gained significant attention in cancer diagnostics, with circulating tumor cells (CTCs) and circulating tumor DNA (ctDNA) being recognized as key biomarkers for tumor detection and monitoring. However, each biomarker possesses inherent limitations that restrict its standalone clinical utility, such [...] Read more.
The liquid biopsy has gained significant attention in cancer diagnostics, with circulating tumor cells (CTCs) and circulating tumor DNA (ctDNA) being recognized as key biomarkers for tumor detection and monitoring. However, each biomarker possesses inherent limitations that restrict its standalone clinical utility, such as the rarity and heterogeneity of CTCs and the variable sensitivity and specificity of ctDNA assays. This highlights the necessity of integrating both biomarkers to maximize diagnostic and prognostic potential, offering a more comprehensive understanding of the tumor biology and therapeutic response. In this review, we summarize clinical studies that have explored the combined analysis of CTCs and ctDNA as biomarkers, providing insights into their synergistic value in diverse tumor types. Specifically, this paper examines the individual advantages and limitations of CTCs and ctDNA, details the findings of combined biomarker studies across various cancers, highlights the benefits of dual biomarker approaches over single-biomarker strategies, and discusses future prospects for advancing personalized oncology through liquid biopsies. By offering a comprehensive overview of clinical studies combining CTCs and ctDNA, this review serves as a guideline for researchers and clinicians aiming to enhance biomarker-based strategies in oncology and informs biosensor design for improved biomarker detection. Full article
(This article belongs to the Special Issue Immunoassays and Biosensing (2nd Edition))
13 pages, 996 KiB  
Article
Impact of Membranous Nectin-4 on Outcomes of Platinum-Based Chemotherapy in Metastatic Urothelial Carcinoma
by Fu-Jen Hsueh, Chung-Chieh Wang, Jhe-Cyuan Guo, Shih-Chieh Chueh and Yu-Chieh Tsai
Cancers 2025, 17(3), 433; https://doi.org/10.3390/cancers17030433 - 27 Jan 2025
Viewed by 348
Abstract
Background: Platinum-based chemotherapy (Plt-ChT) remains a cornerstone treatment for metastatic urothelial carcinoma (mUC). The nectin-4-targeting antibody–drug conjugate enfortumab vedotin (EV), in combination with immune checkpoint inhibitors, has expanded first-line treatment options. However, biomarkers to guide treatment selection are lacking. Membranous nectin-4 (mNectin-4) expression, [...] Read more.
Background: Platinum-based chemotherapy (Plt-ChT) remains a cornerstone treatment for metastatic urothelial carcinoma (mUC). The nectin-4-targeting antibody–drug conjugate enfortumab vedotin (EV), in combination with immune checkpoint inhibitors, has expanded first-line treatment options. However, biomarkers to guide treatment selection are lacking. Membranous nectin-4 (mNectin-4) expression, a potential marker of EV efficacy, has not been fully explored in the context of Plt-ChT. Methods: We retrospectively analyzed 96 patients with mUC treated with first-line gemcitabine plus cisplatin or carboplatin (GC/GCarbo) and classified mNectin-4 expression using immunohistochemical staining as either high (histochemical score (H-score): 100–300, mNectin-4High) or low (H-score: 0–99, mNectin-4Low). Results: Among these patients, 54.1% exhibited mNectin-4High tumors, which were associated with bladder-origin tumors, a pure urothelial carcinoma histology, de novo presentation, and lymph node-only metastasis. The mNectin-4High subgroup showed a non-significant trend toward better outcomes, including response rate (46.1% vs. 36.3%; p = 0.32), median progression-free survival (7.0 months vs. 4.0 months; adjusted p = 0.06), and median overall survival (20.0 months vs. 8.8 months; adjusted p = 0.06), compared with the mNectin-4Low subgroup. The subgroup analysis revealed that the favorable outcomes for mNectin-4High tumors were predominantly observed in the GC cohort, which was not evident in the GCarbo cohort. Conclusions: Our study suggests a non-significant trend toward improved outcomes with first-line Plt-ChT in patients with mNectin-4High tumors, particularly with the GC regimen. Larger prospective studies are warranted to validate mNectin-4 as a potential predictive biomarker for mUC treatment. Full article
(This article belongs to the Special Issue Clinical Outcomes in Urologic Cancers)
18 pages, 6491 KiB  
Article
An Integrated Approach Utilizing Single-Cell and Bulk RNA-Sequencing for the Identification of a Mitophagy-Associated Genes Signature: Implications for Prognostication and Therapeutic Stratification in Prostate Cancer
by Yuke Zhang, Li Ding, Zhijin Zhang, Liliang Shen, Yadong Guo, Wentao Zhang, Yang Yu, Zhuoran Gu, Ji Liu, Aimaitiaji Kadier, Jiang Geng, Shiyu Mao and Xudong Yao
Biomedicines 2025, 13(2), 311; https://doi.org/10.3390/biomedicines13020311 - 27 Jan 2025
Viewed by 460
Abstract
Introduction: Prostate cancer, notably prostate adenocarcinoma (PARD), has high incidence and mortality rates. Although typically resistant to immunotherapy, recent studies have found immune targets for prostate cancer. Stratifying patients by molecular subtypes may identify those who could benefit from immunotherapy. Methods: [...] Read more.
Introduction: Prostate cancer, notably prostate adenocarcinoma (PARD), has high incidence and mortality rates. Although typically resistant to immunotherapy, recent studies have found immune targets for prostate cancer. Stratifying patients by molecular subtypes may identify those who could benefit from immunotherapy. Methods: We used single-cell and bulk RNA sequencing data from GEO and TCGA databases. We characterized the tumor microenvironment at the single-cell level, analyzing cell interactions and identifying fibroblasts linked to mitophagy. Target genes were narrowed down at the bulk transcriptome level to construct a PARD prognosis prediction nomogram. Unsupervised consensus clustering classified PARD into subtypes, analyzing differences in clinical features, immune infiltration, and immunotherapy. Furthermore, the cellular functions of the genes of interest were verified in vitro. Results: We identified ten cell types and 160 mitophagy-related single-cell differentially expressed genes (MR-scDEGs). Strong interactions were observed between fibroblasts, endothelial cells, CD8+ T cells, and NK cells. Fibroblasts linked to mitophagy were divided into six subtypes. Intersection of DEGs from three bulk datasets with MR-scDEGs identified 26 key genes clustered into two subgroups. COX regression analysis identified seven prognostic key genes, enabling a prognostic nomogram model. High and low-risk groups showed significant differences in clinical features, immune infiltration, immunotherapy, and drug sensitivity. In prostate cancer cell lines, CAV1, PALLD, and ITGB8 are upregulated, while CLDN7 is downregulated. Knockdown of PALLD significantly inhibits the proliferation and colony-forming ability of PC3 and DU145 cells, suggesting the important roles of this gene in prostate cancer progression. Conclusions: This study analyzed mitophagy-related genes in PARD, predicting prognosis and aiding in subtype identification and immunotherapy response analysis. This approach offers new strategies for treating prostate cancer with specific molecular subtypes and helps develop potential biomarkers for personalized medicine strategies. Full article
(This article belongs to the Section Cancer Biology and Oncology)
21 pages, 2675 KiB  
Review
Advances in Understanding Lipopolysaccharide-Mediated Hepatitis: Mechanisms and Pathological Features
by Kazuhiko Nakadate, Hayate Saitoh, Miina Sakaguchi, Fumito Miruno, Naoto Muramatsu, Nozomi Ito, Kanako Tadokoro and Kiyoharu Kawakami
Curr. Issues Mol. Biol. 2025, 47(2), 79; https://doi.org/10.3390/cimb47020079 - 27 Jan 2025
Viewed by 259
Abstract
Lipopolysaccharide (LPS), a key component of Gram-negative bacterial membranes, plays a central role in the pathogenesis of inflammatory liver diseases. In this review, we aimed to explore the role of LPS in hepatic injury. Upon hepatic infiltration, LPS activates Kupffer cells via toll-like [...] Read more.
Lipopolysaccharide (LPS), a key component of Gram-negative bacterial membranes, plays a central role in the pathogenesis of inflammatory liver diseases. In this review, we aimed to explore the role of LPS in hepatic injury. Upon hepatic infiltration, LPS activates Kupffer cells via toll-like receptor 4 (TLR4) signaling, inducing proinflammatory cytokines such as tumor necrosis factor-α and interleukin-1β. These mediators amplify hepatocyte apoptosis, endothelial damage, and platelet aggregation, thereby contributing to sinusoidal thrombosis and tissue ischemia. Pathological features, such as hepatocyte shrinkage, sinusoidal expansion, and fibrin deposition, are hallmark indicators of LPS-induced hepatic inflammation. Therapeutically, aspirin shows promise for attenuating cytokine release, protecting endothelial integrity, and reducing thrombogenesis. Emerging strategies include targeting TLR4 pathways, modulating the gut–liver axis, and utilizing biomolecular approaches such as RNA interference for LPS suppression. The integration of public health interventions, such as dietary optimization and microbiome regulation, offers additional preventive measures. In this review, the dual roles of LPS in inflammation and thrombosis have been emphasized. Advancing our understanding of LPS-driven mechanisms and enhancing treatment strategies are pivotal for managing hepatic inflammation and its systemic implications. Future research should focus on refining biomarkers, optimizing therapeutic efficacy, and addressing safety concerns for clinical applications. Full article
(This article belongs to the Special Issue Advances in Molecular Biology Methods in Hepatology Research)
Show Figures

Figure 1

Figure 1
<p>(<b>A</b>) Schematic representation of the cell surface structure of Gram-negative bacteria; (<b>B</b>) structure of lipopolysaccharide (LPS).</p>
Full article ">Figure 2
<p>Schematic diagram of intrahepatic sinusoids, various factors induced by LPS, and the mechanism of thrombogenesis. LPS—lipopolysaccharide; TLR4—toll-like receptor 4; IL-1β—interleukin-1β; TNF-α—tumor necrosis factor-α; IL-6—interleukin-6; TF—tissue factor; VII—factor VII of blood coagulation; TM—thrombomodulin; EPCR—endothelial protein C receptor; PAI-1—plasminogen activator inhibitor-1; t-PA—tissue-type plasminogen activator.</p>
Full article ">Figure 3
<p>(<b>A</b>,<b>B</b>) Hematoxylin and eosin-stained images of the liver lobules; (<b>C</b>) hematoxylin and eosin-stained image of the intrahepatic vein. (<b>A</b>) Normal mouse liver; (<b>B</b>,<b>C</b>) mouse liver 24 h after LPS administration. As shown in (<b>A</b>), the hepatocytes of normal mice are healthy, and staining is even. The sinusoids are normal and few hemocytes are observed in the sinusoids. As shown in (<b>B</b>), the hepatocytes in the LPS-treated group are not uniformly stained and are damaged. The sinusoids are swollen. There is an increase in the number of erythrocytes and leukocytes in sinusoids. In (<b>C</b>), a large number of blood cells are stored in the vein. All scale bars are 100 μm.</p>
Full article ">Figure 4
<p>Scanning electron microscopy images of intrahepatic veins 24 h after LPS administration. (<b>A</b>,<b>B</b>) show different vascular images; (<b>B′</b>) magnified image of the red frame in (<b>B</b>). A thrombus is observed in the veins. All scale bars are 100 μm.</p>
Full article ">Figure 5
<p>Identical histopathological images obtained using correlative light and electron microscopy. (<b>A</b>,<b>A′</b>) Hematoxylin-stained images; (<b>B</b>–<b>B″</b>) scanning electron microscopy images of the same field of view; (<b>A′</b>) magnified image of the red frame in (<b>A</b>); (<b>B′</b>) magnified image of the red frame in (<b>B</b>); (<b>B″</b>) magnified image of the blue frame in (<b>B′</b>); (<b>C</b>) magnified image of the yellow frame in (<b>B</b>). The morphology of the blood cells is observed using an optical microscope and confirmed using a scanning electron microscope. It is possible to observe blood cells and thrombi in the blood vessels in detail. Scale bar is 10 μm.</p>
Full article ">
Back to TopTop