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

Article Types

Countries / Regions

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Search Results (288)

Search Parameters:
Keywords = integrated biomarker response approach

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
37 pages, 1469 KiB  
Review
Oncolytic Therapies for Glioblastoma: Advances, Challenges, and Future Perspectives
by Omar Alomari, Habiba Eyvazova, Beyzanur Güney, Rana Al Juhmani, Hatice Odabasi, Lubna Al-Rawabdeh, Muhammed Edib Mokresh, Ufuk Erginoglu, Abdullah Keles and Mustafa K. Baskaya
Cancers 2025, 17(15), 2550; https://doi.org/10.3390/cancers17152550 (registering DOI) - 1 Aug 2025
Viewed by 33
Abstract
Glioblastoma (GBM) remains one of the most aggressive and treatment-resistant brain tumors, necessitating novel therapeutic approaches. Oncolytic treatments, particularly oncolytic viruses (OVs), have emerged as promising candidates by selectively infecting and lysing tumor cells while stimulating anti-tumor immunity. Various virus-based therapies are under [...] Read more.
Glioblastoma (GBM) remains one of the most aggressive and treatment-resistant brain tumors, necessitating novel therapeutic approaches. Oncolytic treatments, particularly oncolytic viruses (OVs), have emerged as promising candidates by selectively infecting and lysing tumor cells while stimulating anti-tumor immunity. Various virus-based therapies are under investigation, including genetically engineered herpes simplex virus (HSV), adenovirus, poliovirus, reovirus, vaccinia virus, measles virus, and Newcastle disease virus, each exploiting unique tumor-selective mechanisms. While some, such as HSV-based therapies including G207 and DelytactTM, have demonstrated clinical progress, significant challenges persist, including immune evasion, heterogeneity in patient response, and delivery barriers due to the blood–brain barrier. Moreover, combination strategies integrating OVs with immune checkpoint inhibitors, chemotherapy, and radiation are promising but require further clinical validation. Non-viral oncolytic approaches, such as tumor-targeting bacteria and synthetic peptides, remain underexplored. This review highlights current advancements while addressing critical gaps in the literature, including the need for optimized delivery methods, better biomarker-based patient stratification, and a deeper understanding of GBM’s immunosuppressive microenvironment. Future research should focus on enhancing OV specificity, engineering viruses to deliver therapeutic genes, and integrating OVs with precision medicine strategies. By identifying these gaps, this review provides a framework for advancing oncolytic therapies in GBM treatment. Full article
(This article belongs to the Section Cancer Immunology and Immunotherapy)
Show Figures

Figure 1

20 pages, 1318 KiB  
Review
A Genetically-Informed Network Model of Myelodysplastic Syndrome: From Splicing Aberrations to Therapeutic Vulnerabilities
by Sanghyeon Yu, Junghyun Kim and Man S. Kim
Genes 2025, 16(8), 928; https://doi.org/10.3390/genes16080928 (registering DOI) - 1 Aug 2025
Viewed by 31
Abstract
Background/Objectives: Myelodysplastic syndrome (MDS) is a heterogeneous clonal hematopoietic disorder characterized by ineffective hematopoiesis and leukemic transformation risk. Current therapies show limited efficacy, with ~50% of patients failing hypomethylating agents. This review aims to synthesize recent discoveries through an integrated network model and [...] Read more.
Background/Objectives: Myelodysplastic syndrome (MDS) is a heterogeneous clonal hematopoietic disorder characterized by ineffective hematopoiesis and leukemic transformation risk. Current therapies show limited efficacy, with ~50% of patients failing hypomethylating agents. This review aims to synthesize recent discoveries through an integrated network model and examine translation into precision therapeutic approaches. Methods: We reviewed breakthrough discoveries from the past three years, analyzing single-cell multi-omics technologies, epitranscriptomics, stem cell architecture analysis, and precision medicine approaches. We examined cell-type-specific splicing aberrations, distinct stem cell architectures, epitranscriptomic modifications, and microenvironmental alterations in MDS pathogenesis. Results: Four interconnected mechanisms drive MDS: genetic alterations (splicing factor mutations), aberrant stem cell architecture (CMP-pattern vs. GMP-pattern), epitranscriptomic dysregulation involving pseudouridine-modified tRNA-derived fragments, and microenvironmental changes. Splicing aberrations show cell-type specificity, with SF3B1 mutations preferentially affecting erythroid lineages. Stem cell architectures predict therapeutic responses, with CMP-pattern MDS achieving superior venetoclax response rates (>70%) versus GMP-pattern MDS (<30%). Epitranscriptomic alterations provide independent prognostic information, while microenvironmental changes mediate treatment resistance. Conclusions: These advances represent a paradigm shift toward personalized MDS medicine, moving from single-biomarker to comprehensive molecular profiling guiding multi-target strategies. While challenges remain in standardizing molecular profiling and developing clinical decision algorithms, this systems-level understanding provides a foundation for precision oncology implementation and overcoming current therapeutic limitations. Full article
(This article belongs to the Section Molecular Genetics and Genomics)
31 pages, 419 KiB  
Review
Neoadjuvant Treatment for Locally Advanced Rectal Cancer: Current Status and Future Directions
by Masayoshi Iwamoto, Kazuki Ueda and Junichiro Kawamura
Cancers 2025, 17(15), 2540; https://doi.org/10.3390/cancers17152540 - 31 Jul 2025
Viewed by 190
Abstract
Locally advanced rectal cancer (LARC) remains a major clinical challenge due to its high risk of local recurrence and distant metastasis. Although total mesorectal excision (TME) has been established as the gold standard surgical approach, high recurrence rates associated with surgery alone have [...] Read more.
Locally advanced rectal cancer (LARC) remains a major clinical challenge due to its high risk of local recurrence and distant metastasis. Although total mesorectal excision (TME) has been established as the gold standard surgical approach, high recurrence rates associated with surgery alone have driven the development of multimodal preoperative strategies, such as radiotherapy and chemoradiotherapy. More recently, total neoadjuvant therapy (TNT)—which integrates systemic chemotherapy and radiotherapy prior to surgery—and non-operative management (NOM) for patients who achieve a clinical complete response (cCR) have further expanded treatment options. These advances aim not only to improve oncologic outcomes but also to enhance quality of life (QOL) by reducing long-term morbidity and preserving organ function. However, several unresolved issues persist, including the optimal sequencing of therapies, precise risk stratification, accurate evaluation of treatment response, and effective surveillance protocols for NOM. The advent of molecular biomarkers, next-generation sequencing, and artificial intelligence (AI) presents new opportunities for individualized treatment and more accurate prognostication. This narrative review provides a comprehensive overview of the current status of preoperative treatment for LARC, critically examines emerging strategies and their supporting evidence, and discusses future directions to optimize both oncological and patient-centered outcomes. By integrating clinical, molecular, and technological advances, the management of rectal cancer is moving toward truly personalized medicine. Full article
(This article belongs to the Special Issue Multidisciplinary Management of Rectal Cancer)
35 pages, 887 KiB  
Review
Prognostic Factors in Colorectal Liver Metastases: An Exhaustive Review of the Literature and Future Prospectives
by Maria Conticchio, Emilie Uldry, Martin Hübner, Antonia Digklia, Montserrat Fraga, Christine Sempoux, Jean Louis Raisaro and David Fuks
Cancers 2025, 17(15), 2539; https://doi.org/10.3390/cancers17152539 - 31 Jul 2025
Viewed by 81
Abstract
Background: Colorectal liver metastasis (CRLM) represents a major clinical challenge in oncology, affecting 25–50% of colorectal cancer patients and significantly impacting survival. While multimodal therapies—including surgical resection, systemic chemotherapy, and local ablative techniques—have improved outcomes, prognosis remains heterogeneous due to variations in [...] Read more.
Background: Colorectal liver metastasis (CRLM) represents a major clinical challenge in oncology, affecting 25–50% of colorectal cancer patients and significantly impacting survival. While multimodal therapies—including surgical resection, systemic chemotherapy, and local ablative techniques—have improved outcomes, prognosis remains heterogeneous due to variations in tumor biology, patient factors, and institutional practices. Methods: This review synthesizes current evidence on prognostic factors influencing CRLM management, encompassing clinical (e.g., tumor burden, anatomic distribution, timing of metastases), biological (e.g., CEA levels, inflammatory markers), and molecular (e.g., RAS/BRAF mutations, MSI status, HER2 alterations) determinants. Results: Key findings highlight the critical role of molecular profiling in guiding therapeutic decisions, with RAS/BRAF mutations predicting resistance to anti-EGFR therapies and MSI-H status indicating potential responsiveness to immunotherapy. Emerging tools like circulating tumor DNA (ctDNA) and radiomics offer promise for dynamic risk stratification and early recurrence detection, while the gut microbiome is increasingly recognized as a modulator of treatment response. Conclusions: Despite advancements, challenges persist in standardizing resectability criteria and integrating multidisciplinary approaches. Current guidelines (NCCN, ESMO, ASCO) emphasize personalized strategies but lack granularity in terms of incorporating novel biomarkers. This exhaustive review underscores the imperative for the development of a unified, biomarker-integrated framework to refine CRLM management and improve long-term outcomes. Full article
Show Figures

Figure 1

19 pages, 707 KiB  
Review
Salivary α-Amylase as a Metabolic Biomarker: Analytical Tools, Challenges, and Clinical Perspectives
by Gita Erta, Gita Gersone, Antra Jurka and Peteris Tretjakovs
Int. J. Mol. Sci. 2025, 26(15), 7365; https://doi.org/10.3390/ijms26157365 - 30 Jul 2025
Viewed by 257
Abstract
Salivary α-amylase, primarily encoded by the AMY1 gene, initiates the enzymatic digestion of dietary starch in the oral cavity and has recently emerged as a potential biomarker in metabolic research. Variability in salivary amylase activity (SAA), driven largely by copy number variation of [...] Read more.
Salivary α-amylase, primarily encoded by the AMY1 gene, initiates the enzymatic digestion of dietary starch in the oral cavity and has recently emerged as a potential biomarker in metabolic research. Variability in salivary amylase activity (SAA), driven largely by copy number variation of AMY1, has been associated with postprandial glycemic responses, insulin secretion dynamics, and susceptibility to obesity. This review critically examines current analytical approaches for quantifying SAA, including enzymatic assays, colorimetric techniques, immunoassays, and emerging biosensor technologies. The methodological limitations related to sample handling, intra-individual variability, assay standardization, and specificity are highlighted in the context of metabolic and clinical studies. Furthermore, the review explores the physiological relevance of SAA in energy homeostasis and its associations with visceral adiposity and insulin resistance. We discuss the potential integration of SAA measurements into obesity risk stratification and personalized dietary interventions, particularly in individuals with altered starch metabolism. Finally, the review identifies key research gaps and future directions necessary to validate SAA as a reliable metabolic biomarker in clinical practice. Understanding the diagnostic and prognostic value of salivary amylase may offer new insights into the prevention and management of obesity and related metabolic disorders. Full article
Show Figures

Figure 1

36 pages, 5612 KiB  
Review
The Multifaceted Role of p53 in Cancer Molecular Biology: Insights for Precision Diagnosis and Therapeutic Breakthroughs
by Bolong Xu, Ayitila Maimaitijiang, Dawuti Nuerbiyamu, Zhengding Su and Wenfang Li
Biomolecules 2025, 15(8), 1088; https://doi.org/10.3390/biom15081088 - 27 Jul 2025
Viewed by 410
Abstract
The protein p53, often referred to as the “guardian of the genome,” is essential for preserving cellular balance and preventing cancerous transformations. As one of the most commonly altered genes in human cancers, its impaired function is associated with tumor initiation, development, and [...] Read more.
The protein p53, often referred to as the “guardian of the genome,” is essential for preserving cellular balance and preventing cancerous transformations. As one of the most commonly altered genes in human cancers, its impaired function is associated with tumor initiation, development, and resistance to treatment. Exploring the diverse roles of p53, which include regulating the cell cycle, repairing DNA, inducing apoptosis, reprogramming metabolism, and modulating immunity, provides valuable insights into cancer mechanisms and potential treatments. This review integrates recent findings on p53′s dual nature, functioning as both a tumor suppressor and an oncogenic promoter, depending on the context. Wild-type p53 suppresses tumors by inducing cell cycle arrest or apoptosis in response to genotoxic stress, while mutated variants often lose these functions or gain novel pro-oncogenic activities. Emerging evidence highlights p53′s involvement in non-canonical pathways, such as regulating tumor microenvironment interactions, metabolic flexibility, and immune evasion mechanisms. For instance, p53 modulates immune checkpoint expression and influences the efficacy of immunotherapies, including PD-1/PD-L1 blockade. Furthermore, advancements in precision diagnostics, such as liquid biopsy-based detection of p53 mutations and AI-driven bioinformatics tools, enable early cancer identification and stratification of patients likely to benefit from targeted therapies. Therapeutic strategies targeting p53 pathways are rapidly evolving. Small molecules restoring wild-type p53 activity or disrupting mutant p53 interactions, such as APR-246 and MDM2 inhibitors, show promise in clinical trials. Combination approaches integrating gene editing with synthetic lethal strategies aim to exploit p53-dependent vulnerabilities. Additionally, leveraging p53′s immunomodulatory effects through vaccine development or adjuvants may enhance immunotherapy responses. In conclusion, deciphering p53′s complex biology underscores its unparalleled potential as a biomarker and therapeutic target. Integrating multi-omics analyses, functional genomic screens, and real-world clinical data will accelerate the translation of p53-focused research into precision oncology breakthroughs, ultimately improving patient outcomes. Full article
(This article belongs to the Special Issue DNA Damage and Repair in Cancer Treatment)
Show Figures

Figure 1

18 pages, 605 KiB  
Review
Gut Microbiota, Microbial Metabolites, and Inflammation in Cardiac Surgery: Implications for Clinical Outcomes—A Narrative Review
by Panagiota Misokalou, Arezina N. Kasti, Konstantinos Katsas and Dimitrios C. Angouras
Microorganisms 2025, 13(8), 1748; https://doi.org/10.3390/microorganisms13081748 - 26 Jul 2025
Viewed by 436
Abstract
Cardiac surgery, particularly procedures involving cardiopulmonary bypass (CPB), is associated with a high risk of postoperative complications, including systemic inflammatory response syndrome (SIRS), postoperative atrial fibrillation (POAF), and infection. Growing evidence suggests that the gut–heart axis, through mechanisms involving intestinal barrier integrity and [...] Read more.
Cardiac surgery, particularly procedures involving cardiopulmonary bypass (CPB), is associated with a high risk of postoperative complications, including systemic inflammatory response syndrome (SIRS), postoperative atrial fibrillation (POAF), and infection. Growing evidence suggests that the gut–heart axis, through mechanisms involving intestinal barrier integrity and gut microbiota homeostasis, may influence these outcomes. This review summarizes the relationship between gut microbiota composition and the inflammatory response in patients undergoing cardiac surgery and the extent to which these alterations impact clinical outcomes. The reviewed studies consistently show that cardiac surgery induces notable alterations in microbial diversity and composition during the perioperative period. These changes, indicative of dysbiosis, are characterized by a reduction in health-associated bacteria such as Blautia, Faecalibacterium, and Bifidobacterium and an increase in opportunistic pathogens. Inflammatory biomarkers were frequently elevated postoperatively, even in patients without evident complications. Key microbial metabolites and biomarkers, including short-chain fatty acids (SCFAs), trimethylamine N-oxide (TMAO), and bile acids (BAs), were implicated in modulating inflammation and clinical outcomes. Additionally, vitamin D deficiency emerged as a contributing factor, correlating with increased systemic inflammation and a higher incidence of POAF. The findings suggest that gut microbiota composition prior to surgery may influence the severity of the postoperative inflammatory response and that perioperative modulation of the gut microbiota could represent a novel approach to improving surgical outcomes. However, the relationship between dysbiosis and acute illness in surgical patients is confounded by factors such as antibiotic use and other perioperative interventions. Large-scale, standardized clinical studies are needed to better define these interactions and guide future therapeutic strategies in cardiac surgery. Full article
Show Figures

Graphical abstract

14 pages, 1209 KiB  
Article
Investigation of Growth Differentiation Factor 15 as a Prognostic Biomarker for Major Adverse Limb Events in Peripheral Artery Disease
by Ben Li, Farah Shaikh, Houssam Younes, Batool Abuhalimeh, Abdelrahman Zamzam, Rawand Abdin and Mohammad Qadura
J. Clin. Med. 2025, 14(15), 5239; https://doi.org/10.3390/jcm14155239 - 24 Jul 2025
Viewed by 295
Abstract
Background/Objectives: Peripheral artery disease (PAD) impacts more than 200 million individuals globally and leads to mortality and morbidity secondary to progressive limb dysfunction and amputation. However, clinical management of PAD remains suboptimal, in part because of the lack of standardized biomarkers to predict [...] Read more.
Background/Objectives: Peripheral artery disease (PAD) impacts more than 200 million individuals globally and leads to mortality and morbidity secondary to progressive limb dysfunction and amputation. However, clinical management of PAD remains suboptimal, in part because of the lack of standardized biomarkers to predict patient outcomes. Growth differentiation factor 15 (GDF15) is a stress-responsive cytokine that has been studied extensively in cardiovascular disease, but its investigation in PAD remains limited. This study aimed to use explainable statistical and machine learning methods to assess the prognostic value of GDF15 for limb outcomes in patients with PAD. Methods: This prognostic investigation was carried out using a prospectively enrolled cohort comprising 454 patients diagnosed with PAD. At baseline, plasma GDF15 levels were measured using a validated multiplex immunoassay. Participants were monitored over a two-year period to assess the occurrence of major adverse limb events (MALE), a composite outcome encompassing major lower extremity amputation, need for open/endovascular revascularization, or acute limb ischemia. An Extreme Gradient Boosting (XGBoost) model was trained to predict 2-year MALE using 10-fold cross-validation, incorporating GDF15 levels along with baseline variables. Model performance was primarily evaluated using the area under the receiver operating characteristic curve (AUROC). Secondary model evaluation metrics were accuracy, sensitivity, specificity, negative predictive value (NPV), and positive predictive value (PPV). Prediction histogram plots were generated to assess the ability of the model to discriminate between patients who develop vs. do not develop 2-year MALE. For model interpretability, SHapley Additive exPlanations (SHAP) analysis was performed to evaluate the relative contribution of each predictor to model outputs. Results: The mean age of the cohort was 71 (SD 10) years, with 31% (n = 139) being female. Over the two-year follow-up period, 157 patients (34.6%) experienced MALE. The XGBoost model incorporating plasma GDF15 levels and demographic/clinical features achieved excellent performance for predicting 2-year MALE in PAD patients: AUROC 0.84, accuracy 83.5%, sensitivity 83.6%, specificity 83.7%, PPV 87.3%, and NPV 86.2%. The prediction probability histogram for the XGBoost model demonstrated clear separation for patients who developed vs. did not develop 2-year MALE, indicating strong discrimination ability. SHAP analysis showed that GDF15 was the strongest predictive feature for 2-year MALE, followed by age, smoking status, and other cardiovascular comorbidities, highlighting its clinical relevance. Conclusions: Using explainable statistical and machine learning methods, we demonstrated that plasma GDF15 levels have important prognostic value for 2-year MALE in patients with PAD. By integrating clinical variables with GDF15 levels, our machine learning model can support early identification of PAD patients at elevated risk for adverse limb events, facilitating timely referral to vascular specialists and aiding in decisions regarding the aggressiveness of medical/surgical treatment. This precision medicine approach based on a biomarker-guided prognostication algorithm offers a promising strategy for improving limb outcomes in individuals with PAD. Full article
(This article belongs to the Special Issue The Role of Biomarkers in Cardiovascular Diseases)
Show Figures

Figure 1

81 pages, 4295 KiB  
Systematic Review
Leveraging AI-Driven Neuroimaging Biomarkers for Early Detection and Social Function Prediction in Autism Spectrum Disorders: A Systematic Review
by Evgenia Gkintoni, Maria Panagioti, Stephanos P. Vassilopoulos, Georgios Nikolaou, Basilis Boutsinas and Apostolos Vantarakis
Healthcare 2025, 13(15), 1776; https://doi.org/10.3390/healthcare13151776 - 22 Jul 2025
Viewed by 640
Abstract
Background: This systematic review examines artificial intelligence (AI) applications in neuroimaging for autism spectrum disorder (ASD), addressing six research questions regarding biomarker optimization, modality integration, social function prediction, developmental trajectories, clinical translation challenges, and multimodal data enhancement for earlier detection and improved [...] Read more.
Background: This systematic review examines artificial intelligence (AI) applications in neuroimaging for autism spectrum disorder (ASD), addressing six research questions regarding biomarker optimization, modality integration, social function prediction, developmental trajectories, clinical translation challenges, and multimodal data enhancement for earlier detection and improved outcomes. Methods: Following PRISMA guidelines, we conducted a comprehensive literature search across 8 databases, yielding 146 studies from an initial 1872 records. These studies were systematically analyzed to address key questions regarding AI neuroimaging approaches in ASD detection and prognosis. Results: Neuroimaging combined with AI algorithms demonstrated significant potential for early ASD detection, with electroencephalography (EEG) showing promise. Machine learning classifiers achieved high diagnostic accuracy (85–99%) using features derived from neural oscillatory patterns, connectivity measures, and signal complexity metrics. Studies of infant populations have identified the 9–12-month developmental window as critical for biomarker detection and the onset of behavioral symptoms. Multimodal approaches that integrate various imaging techniques have substantially enhanced predictive capabilities, while longitudinal analyses have shown potential for tracking developmental trajectories and treatment responses. Conclusions: AI-driven neuroimaging biomarkers represent a promising frontier in ASD research, potentially enabling the detection of symptoms before they manifest behaviorally and providing objective measures of intervention efficacy. While technical and methodological challenges remain, advancements in standardization, diverse sampling, and clinical validation could facilitate the translation of findings into practice, ultimately supporting earlier intervention during critical developmental periods and improving outcomes for individuals with ASD. Future research should prioritize large-scale validation studies and standardized protocols to realize the full potential of precision medicine in ASD. Full article
Show Figures

Graphical abstract

28 pages, 1358 KiB  
Review
Understanding the Borderline Brain: A Review of Neurobiological Findings in Borderline Personality Disorder (BPD)
by Eleni Giannoulis, Christos Nousis, Ioanna-Jonida Sula, Maria-Evangelia Georgitsi and Ioannis Malogiannis
Biomedicines 2025, 13(7), 1783; https://doi.org/10.3390/biomedicines13071783 - 21 Jul 2025
Viewed by 703
Abstract
Borderline personality disorder (BPD) is a complex and heterogeneous condition characterized by emotional instability, impulsivity, and impaired regulation of interpersonal relationships. This narrative review integrates findings from recent neuroimaging, neurochemical, and treatment studies to identify core neurobiological mechanisms and highlight translational potential. Evidence [...] Read more.
Borderline personality disorder (BPD) is a complex and heterogeneous condition characterized by emotional instability, impulsivity, and impaired regulation of interpersonal relationships. This narrative review integrates findings from recent neuroimaging, neurochemical, and treatment studies to identify core neurobiological mechanisms and highlight translational potential. Evidence from 112 studies published up to 2025 is synthesized, encompassing structural MRI, resting-state and task-based functional MRI, EEG, PET, and emerging machine learning applications. Consistent disruptions are observed across the prefrontal–amygdala circuitry, the default mode network (DMN), and mentalization-related regions. BPD shows a dominant and stable pattern of hyperconnectivity in the precuneus. Transdiagnostic comparisons with PTSD and cocaine use disorder (CUD) suggest partial overlap in DMN dysregulation, though BPD-specific traits emerge in network topology. Machine learning models achieve a classification accuracy of 70–88% and may support the tracking of early treatment responses. Longitudinal fMRI studies indicate that psychodynamic therapy facilitates the progressive normalization of dorsal anterior cingulate cortex (dACC) activity and reductions in alexithymia. We discuss the role of phenotypic heterogeneity (internalizing versus externalizing profiles), the potential of neuromodulation guided by biomarkers, and the need for standardized imaging protocols. Limitations include small sample sizes, a lack of effective connectivity analyses, and minimal multicenter cohort representation. Future research should focus on constructing multimodal biomarker panels that integrate functional connectivity, epigenetics, and computational phenotyping. This review supports the use of a precision psychiatry approach for BPD by aligning neuroscience with scalable clinical tools. Full article
(This article belongs to the Section Neurobiology and Clinical Neuroscience)
Show Figures

Figure 1

32 pages, 1948 KiB  
Review
Writing the Future: Artificial Intelligence, Handwriting, and Early Biomarkers for Parkinson’s Disease Diagnosis and Monitoring
by Giuseppe Marano, Sara Rossi, Ester Maria Marzo, Alice Ronsisvalle, Laura Artuso, Gianandrea Traversi, Antonio Pallotti, Francesco Bove, Carla Piano, Anna Rita Bentivoglio, Gabriele Sani and Marianna Mazza
Biomedicines 2025, 13(7), 1764; https://doi.org/10.3390/biomedicines13071764 - 18 Jul 2025
Viewed by 437
Abstract
Parkinson’s disease (PD) is a progressive neurodegenerative disorder that impairs motor function, including the fine motor control required for handwriting. Traditional diagnostic methods often lack sensitivity and objectivity in the early stages, limiting opportunities for timely intervention. There is a growing need for [...] Read more.
Parkinson’s disease (PD) is a progressive neurodegenerative disorder that impairs motor function, including the fine motor control required for handwriting. Traditional diagnostic methods often lack sensitivity and objectivity in the early stages, limiting opportunities for timely intervention. There is a growing need for non-invasive, accessible tools capable of capturing subtle motor changes that precede overt clinical symptoms. Among early PD manifestations, handwriting impairments such as micrographia have shown potential as digital biomarkers. However, conventional handwriting analysis remains subjective and limited in scope. Recent advances in artificial intelligence (AI) and machine learning (ML) enable automated analysis of handwriting dynamics, such as pressure, velocity, and fluency, collected via digital tablets and smartpens. These tools support the detection of early-stage PD, monitoring of disease progression, and assessment of therapeutic response. This paper highlights how AI-enhanced handwriting analysis provides a scalable, non-invasive method to support diagnosis, enable remote symptom tracking, and personalize treatment strategies in PD. This approach integrates clinical neurology with computer science and rehabilitation, offering practical applications in telemedicine, digital health, and personalized medicine. By capturing dynamic features often missed by traditional assessments, AI-based handwriting analysis contributes to a paradigm shift in the early detection and long-term management of PD, with broad relevance across neurology, digital diagnostics, and public health innovation. Full article
(This article belongs to the Section Neurobiology and Clinical Neuroscience)
Show Figures

Figure 1

26 pages, 1644 KiB  
Review
Therapeutic Targeting of Apoptosis, Autophagic Cell Death, Necroptosis, Pyroptosis, and Ferroptosis Pathways in Oral Squamous Cell Carcinoma: Molecular Mechanisms and Potential Strategies
by Po-Chih Hsu, Chung-Che Tsai, Ya-Hsuan Lin and Chan-Yen Kuo
Biomedicines 2025, 13(7), 1745; https://doi.org/10.3390/biomedicines13071745 - 16 Jul 2025
Viewed by 448
Abstract
Oral squamous cell carcinoma (OSCC) is a prevalent and aggressive malignancy with poor prognosis, largely due to its high metastatic potential and resistance to conventional therapies. Recent advances in cancer biology have underscored the significance of regulated cell death pathways, including apoptosis, autophagic [...] Read more.
Oral squamous cell carcinoma (OSCC) is a prevalent and aggressive malignancy with poor prognosis, largely due to its high metastatic potential and resistance to conventional therapies. Recent advances in cancer biology have underscored the significance of regulated cell death pathways, including apoptosis, autophagic cell death (ACD), necroptosis, pyroptosis, and ferroptosis, in modulating tumor progression and therapeutic responses. This review provides the current insights into the molecular mechanisms underlying these cell death pathways and explores their therapeutic relevance in OSCC. Restoration of apoptosis using BH3 mimetics, tumor necrosis factor (TNF)-related apoptosis-inducing ligand (TRAIL) receptor agonists, and p53 reactivators shows promise for sensitizing OSCC cells to treatment. Autophagy plays context-dependent roles in cancer, acting as a tumor suppressor during early carcinogenesis by maintaining cellular homeostasis, and as a tumor promoter in established tumors by supporting cancer cell survival under stress. Targeting necroptosis and pyroptosis has emerged as a novel strategy for inducing cancer cell death, with compounds such as acetylshikonin and okanin demonstrating antitumor effects. Additionally, the induction of ferroptosis via lipid peroxidation and glutathione peroxidase 4 (GPX4) inhibition offers a promising avenue for overcoming drug resistance, with agents such as quercetin and trifluoperazine exhibiting preclinical success. Integration of these therapeutic approaches may enhance the OSCC treatment efficacy, reduce chemoresistance, and provide novel prognostic biomarkers for clinical management. Future studies should focus on optimizing combinatorial strategies that effectively leverage these pathways to improve OSCC patient outcomes. Full article
(This article belongs to the Special Issue Oral Cancer: From Pathophysiology to Novel Therapeutic Approach)
Show Figures

Graphical abstract

36 pages, 1414 KiB  
Review
A Systems Biology Approach to Memory Health: Integrating Network Pharmacology, Gut Microbiota, and Multi-Omics for Health Functional Foods
by Heng Yuan, Junyu Zhou, Hongbao Li, Suna Kang and Sunmin Park
Int. J. Mol. Sci. 2025, 26(14), 6698; https://doi.org/10.3390/ijms26146698 - 12 Jul 2025
Viewed by 409
Abstract
Memory impairment, ranging from mild memory impairment to neurodegenerative diseases such as Alzheimer’s disease, poses an escalating global health challenge that necessitates multi-targeted interventions to prevent progression. Health functional foods (HFFs), which include bioactive dietary compounds that not only provide basic nutrition but [...] Read more.
Memory impairment, ranging from mild memory impairment to neurodegenerative diseases such as Alzheimer’s disease, poses an escalating global health challenge that necessitates multi-targeted interventions to prevent progression. Health functional foods (HFFs), which include bioactive dietary compounds that not only provide basic nutrition but also function beyond that to modulate physiological pathways, offer a promising non-pharmacological strategy to preserve memory function. This review presents an integrative framework for the discovery, evaluation, and clinical translation of biomarkers responsive to HFFs in the context of preventing memory impairment. We examine both established clinical biomarkers, such as amyloid-β and tau in the cerebrospinal fluid, neuroimaging indicators, and memory assessments, as well as emerging nutritionally sensitive markers including cytokines, microRNAs, gut microbiota signatures, epigenetic modifications, and neuroactive metabolites. By leveraging systems biology approaches, we explore how network pharmacology, gut–brain axis modulation, and multi-omics integration can help to elucidate the complex interactions between HFF components and memory-related pathways such as neuroinflammation, oxidative stress, synaptic plasticity, and metabolic regulation. The review also addresses the translational pipeline for HFFs, from formulation and standardization to regulatory frameworks and clinical development, with an emphasis on precision nutrition strategies and cross-disciplinary integration. Ultimately, we propose a paradigm shift in memory health interventions, positioning HFFs as scientifically validated compounds for personalized nutrition within a preventative memory function framework. Full article
(This article belongs to the Special Issue Molecular Mechanisms of Alzheimer’s Disease)
Show Figures

Figure 1

22 pages, 853 KiB  
Article
Intelligent Multi-Modeling Reveals Biological Mechanisms and Adaptive Phenotypes in Hair Sheep Lambs from a Semi-Arid Region
by Robson Mateus Freitas Silveira, Fábio Augusto Ribeiro, João Pedro dos Santos, Luiz Paulo Fávero, Luis Orlindo Tedeschi, Anderson Antonio Carvalho Alves, Danilo Augusto Sarti, Anaclaudia Alves Primo, Hélio Henrique Araújo Costa, Neila Lidiany Ribeiro, Amanda Felipe Reitenbach, Fabianno Cavalcante de Carvalho and Aline Vieira Landim
Genes 2025, 16(7), 812; https://doi.org/10.3390/genes16070812 - 11 Jul 2025
Viewed by 418
Abstract
Background: Heat stress challenges small ruminants in semi-arid regions, requiring integrative multi-modeling approaches to identify adaptive thermotolerance traits. This study aimed to identify phenotypic biomarkers and explore the relationships between thermoregulatory responses and hematological, behavioral, morphometric, carcass, and meat traits in lambs. Methods: [...] Read more.
Background: Heat stress challenges small ruminants in semi-arid regions, requiring integrative multi-modeling approaches to identify adaptive thermotolerance traits. This study aimed to identify phenotypic biomarkers and explore the relationships between thermoregulatory responses and hematological, behavioral, morphometric, carcass, and meat traits in lambs. Methods: Twenty 4-month-old non-castrated male lambs, with an average body weight of 19.0 ± 5.11 kg, were evaluated under natural heat stress. Results: Thermoregulatory variables were significantly associated with non-carcass components (p = 0.002), carcass performance (p = 0.027), commercial meat cuts (p = 0.032), and morphometric measures (p = 0.029), with a trend for behavioral responses (p = 0.078). The main phenotypic traits related to thermoregulation included idleness duration, cold carcass weight, blood, liver, spleen, shank, chest circumference, and body length. Exploratory factor analysis reduced the significant indicators to seven latent domains: carcass traits, commercial meat cuts, non-carcass components, idleness and feeding behavior, and morphometric and thermoregulatory responses. Bayesian network modeling revealed interdependencies, showing carcass traits influenced by morphometric and thermoregulatory responses and non-carcass traits linked to ingestive behavior. Thermoregulatory variables were not associated with meat quality or hematological traits. Conclusions: These findings highlight the complex biological relationships underlying heat adaptation and emphasize the potential of combining phenomic data with computational tools to support genomic selection for climate-resilient and welfare-oriented breeding programs. Full article
(This article belongs to the Section Animal Genetics and Genomics)
Show Figures

Graphical abstract

16 pages, 755 KiB  
Review
Hip Fracture as a Systemic Disease in Older Adults: A Narrative Review on Multisystem Implications and Management
by Silvia Andaloro, Stefano Cacciatore, Antonella Risoli, Rocco Maria Comodo, Vincenzo Brancaccio, Riccardo Calvani, Simone Giusti, Mathias Schlögl, Emanuela D’Angelo, Matteo Tosato, Francesco Landi and Emanuele Marzetti
Med. Sci. 2025, 13(3), 89; https://doi.org/10.3390/medsci13030089 - 11 Jul 2025
Viewed by 646
Abstract
Hip fractures are among the most serious health events in older adults, frequently leading to disability, loss of independence, and elevated mortality. In 2019, an estimated 9.6 million new cases occurred globally among adults aged ≥ 55 years, with an incidence rate of [...] Read more.
Hip fractures are among the most serious health events in older adults, frequently leading to disability, loss of independence, and elevated mortality. In 2019, an estimated 9.6 million new cases occurred globally among adults aged ≥ 55 years, with an incidence rate of 681 per 100,000. Despite improved surgical care, one-year mortality remains high (15–30%), and fewer than half of survivors regain their pre-fracture functional status. Traditionally regarded as mechanical injuries, hip fractures are now increasingly recognized as systemic events reflecting and accelerating biological vulnerability and frailty progression. We synthesize evidence across biological, clinical, and social domains to explore the systemic implications of hip fracture, from the acute catabolic response and immune dysfunction to long-term functional decline. The concept of intrinsic capacity, introduced by the World Health Organization, offers a resilience-based framework to assess the multidimensional impact of hip fracture on physical, cognitive, and psychological function. We highlight the importance of orthogeriatric co-management, early surgical intervention, and integrated rehabilitation strategies tailored to the individual’s functional reserves and personal goals. Innovations such as digital health tools, biological aging biomarkers, and personalized surgical approaches represent promising avenues to enhance recovery and autonomy. Ultimately, we advocate for a shift toward interdisciplinary, capacity-oriented models of care that align with the goals of healthy aging and enable recovery that transcends survival, focusing instead on restoring function and quality of life. Full article
Show Figures

Figure 1

Back to TopTop