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

Article Types

Countries / Regions

remove_circle_outline
remove_circle_outline
remove_circle_outline

Search Results (376)

Search Parameters:
Keywords = heterogeneity of biological tissues

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
21 pages, 2994 KiB  
Article
A Multi-Omics Integration Framework with Automated Machine Learning Identifies Peripheral Immune-Coagulation Biomarkers for Schizophrenia Risk Stratification
by Feitong Hong, Qiuming Chen, Xinwei Luo, Sijia Xie, Yijie Wei, Xiaolong Li, Kexin Li, Benjamin Lebeau, Crystal Ling, Fuying Dao, Hao Lin, Lixia Tang, Mi Yang and Hao Lv
Int. J. Mol. Sci. 2025, 26(15), 7640; https://doi.org/10.3390/ijms26157640 - 7 Aug 2025
Abstract
Schizophrenia (SCZ) is a complex psychiatric disorder with heterogeneous molecular underpinnings that remain poorly resolved by conventional single-omics approaches, limiting biomarker discovery and mechanistic insights. To address this gap, we applied an artificial intelligence (AI)-driven multi-omics framework to an open access dataset comprising [...] Read more.
Schizophrenia (SCZ) is a complex psychiatric disorder with heterogeneous molecular underpinnings that remain poorly resolved by conventional single-omics approaches, limiting biomarker discovery and mechanistic insights. To address this gap, we applied an artificial intelligence (AI)-driven multi-omics framework to an open access dataset comprising plasma proteomics, post-translational modifications (PTMs), and metabolomics to systematically dissect SCZ pathophysiology. In a cohort of 104 individuals, comparative analysis of 17 machine learning models revealed that multi-omics integration significantly enhanced classification performance, reaching a maximum AUC of 0.9727 (95% CI: 0.8889–1.000) using LightGBMXT, compared to 0.9636 (95% CI: 0.8636–1.0000) with CNNBiLSTM for proteomics alone. Interpretable feature prioritization identified carbamylation at immunoglobulin-constant region sites IGKC_K20 and IGHG1_K8, alongside oxidation of coagulation factor F10 at residue M8, as key discriminative molecular events. Functional analyses identified significantly enriched pathways including complement activation, platelet signaling, and gut microbiota-associated metabolism. Protein interaction networks further implicated coagulation factors F2, F10, and PLG, as well as complement regulators CFI and C9, as central molecular hubs. The clustering of these molecules highlights a potential axis linking immune activation, blood coagulation, and tissue homeostasis, biological domains increasingly recognized in psychiatric disorders. These results implicate immune–thrombotic dysregulation as a critical component of SCZ pathology, with PTMs of immune proteins serving as quantifiable disease indicators. Our work delineates a robust computational strategy for multi-omics integration into psychiatric research, offering biomarker candidates that warrant further validation for diagnostic and therapeutic applications. Full article
Show Figures

Figure 1

21 pages, 880 KiB  
Review
Regenerative Cartilage Treatment for Focal Chondral Defects in the Knee: Focus on Marrow-Stimulating and Cell-Based Scaffold Approaches
by Filippo Migliorini, Francesco Simeone, Tommaso Bardazzi, Michael Kurt Memminger, Gennaro Pipino, Raju Vaishya and Nicola Maffulli
Cells 2025, 14(15), 1217; https://doi.org/10.3390/cells14151217 - 7 Aug 2025
Abstract
Focal chondral defects of the knee are a common cause of pain and functional limitation in active individuals and may predispose to early degenerative joint changes. Given the limited regenerative capacity of hyaline cartilage, biologically based surgical strategies have emerged to promote tissue [...] Read more.
Focal chondral defects of the knee are a common cause of pain and functional limitation in active individuals and may predispose to early degenerative joint changes. Given the limited regenerative capacity of hyaline cartilage, biologically based surgical strategies have emerged to promote tissue repair and restore joint function. This narrative review critically examines current treatment approaches that rely on autologous cell sources and scaffold-supported regeneration. Particular emphasis is placed on techniques that stimulate endogenous repair or support chondrocyte-based tissue restoration through the use of autologous biomaterial constructs. The influence of lesion morphology, joint biomechanics, and patient-specific variables on treatment selection is discussed in detail, focusing on the differences between tibiofemoral and patellofemoral involvement. Biologically driven approaches have shown promising mid- to long-term outcomes in selected patients, and are increasingly favoured over traditional methods in specific clinical scenarios. However, the literature remains limited by heterogeneity in study design, follow-up duration, and outcome measures. This review aims to provide an evidence-based, morphology-informed framework to support the clinical decision-making process in the management of knee cartilage defects. Full article
Show Figures

Figure 1

28 pages, 1877 KiB  
Review
Unconventional Immunotherapies in Cancer: Opportunities and Challenges
by Meshael Alturki, Abdullah A. Alshehri, Ahmad M. Aldossary, Mohannad M. Fallatah, Fahad A. Almughem, Nojoud Al Fayez, Majed A. Majrashi, Ibrahim A. Alradwan, Mohammad Alkhrayef, Mohammad N. Alomary and Essam A. Tawfik
Pharmaceuticals 2025, 18(8), 1154; https://doi.org/10.3390/ph18081154 - 4 Aug 2025
Viewed by 337
Abstract
Conventional immunotherapy, including immune checkpoint blockade and chimeric antigen receptor (CAR)-T cells, has revolutionized cancer therapy over the past decade. Yet, the efficacy of these therapies is limited by tumor resistance, antigen escape mechanisms, poor persistence, and T-cell exhaustion, particularly in the treatment [...] Read more.
Conventional immunotherapy, including immune checkpoint blockade and chimeric antigen receptor (CAR)-T cells, has revolutionized cancer therapy over the past decade. Yet, the efficacy of these therapies is limited by tumor resistance, antigen escape mechanisms, poor persistence, and T-cell exhaustion, particularly in the treatment of solid tumors. The emergence of unconventional immunotherapies offers novel opportunities by leveraging diverse immune cell subsets and synthetic biologics. This review explores various immunotherapy platforms, including gamma delta T cells, invariant natural killer T cells, mucosal-associated invariant T cells, engineered regulatory T cells, and universal CAR platforms. Additionally, it expands on biologics, including bispecific and multispecific antibodies, cytokine fusions, agonists, and oncolytic viruses, showcasing their potential for modular engineering and off-the-shelf applicability. Distinct features of unconventional platforms include independence from the major histocompatibility complex (MHC), tissue-homing capabilities, stress ligand sensing, and the ability to bridge adaptive and innate immunity. Their compatibility with engineering approaches highlights their potential as scalable, efficient, and cost-effective therapies. To overcome translational challenges such as functional heterogeneity, immune exhaustion, tumor microenvironment-mediated suppression, and limited persistence, novel strategies will be discussed, including metabolic and epigenetic reprogramming, immune cloaking, gene editing, and the utilization of artificial intelligence for patient stratification. Ultimately, unconventional immunotherapies extend the therapeutic horizon of cancer immunotherapy by breaking barriers in solid tumor treatment and increasing accessibility. Continued investments in research for mechanistic insights and scalable manufacturing are key to unlocking their full clinical potential. Full article
(This article belongs to the Section Biopharmaceuticals)
Show Figures

Figure 1

17 pages, 432 KiB  
Article
Anomalous Drug Transport in Biological Tissues: A Caputo Fractional Approach with Non-Classical Boundary Modeling
by Ahmed Ghezal, Ahmed A. Al Ghafli and Hassan J. Al Salman
Fractal Fract. 2025, 9(8), 508; https://doi.org/10.3390/fractalfract9080508 - 4 Aug 2025
Viewed by 133
Abstract
This paper focuses on the numerical modeling of drug diffusion in biological tissues using fractional time-dependent parabolic equations with non-local boundary conditions. The model includes a Caputo fractional derivative to capture the non-local effects and memory inherent in biological processes, such as drug [...] Read more.
This paper focuses on the numerical modeling of drug diffusion in biological tissues using fractional time-dependent parabolic equations with non-local boundary conditions. The model includes a Caputo fractional derivative to capture the non-local effects and memory inherent in biological processes, such as drug absorption and transport. The theoretical framework of the problem is based on the work of Alhazzani, et al.,which demonstrates the solution’s goodness, existence, and uniqueness. Building on this foundation, we present a robust numerical method designed to deal with the complexity of fractional derivatives and non-local interactions at the boundaries of biological tissues. Numerical simulations reveal how fractal order and non-local boundary conditions affect the drug concentration distribution over time, providing valuable insights into drug delivery dynamics in biological systems. The results underscore the potential of fractal models to accurately represent diffusion processes in heterogeneous and complex biological environments. Full article
Show Figures

Figure 1

18 pages, 3891 KiB  
Review
Navigating Brain Organoid Maturation: From Benchmarking Frameworks to Multimodal Bioengineering Strategies
by Jingxiu Huang, Yingli Zhu, Jiong Tang, Yang Liu, Ming Lu, Rongxin Zhang and Alfred Xuyang Sun
Biomolecules 2025, 15(8), 1118; https://doi.org/10.3390/biom15081118 - 4 Aug 2025
Viewed by 266
Abstract
Brain organoid technology has revolutionized in vitro modeling of human neurodevelopment and disease, providing unprecedented insights into cortical patterning, neural circuit assembly, and pathogenic mechanisms of neurological disorders. Critically, human brain organoids uniquely recapitulate human-specific developmental processes—such as the expansion of outer radial [...] Read more.
Brain organoid technology has revolutionized in vitro modeling of human neurodevelopment and disease, providing unprecedented insights into cortical patterning, neural circuit assembly, and pathogenic mechanisms of neurological disorders. Critically, human brain organoids uniquely recapitulate human-specific developmental processes—such as the expansion of outer radial glia and neuromelanin—that are absent in rodent models, making them indispensable for studying human brain evolution and dysfunction. However, a major bottleneck persists: Extended culture periods (≥6 months) are empirically required to achieve late-stage maturation markers like synaptic refinement, functional network plasticity, and gliogenesis. Yet prolonged conventional 3D culture exacerbates metabolic stress, hypoxia-induced necrosis, and microenvironmental instability, leading to asynchronous tissue maturation—electrophysiologically active superficial layers juxtaposed with degenerating cores. This immaturity/heterogeneity severely limits their utility in modeling adult-onset disorders (e.g., Alzheimer’s disease) and high-fidelity drug screening, as organoids fail to recapitulate postnatal transcriptional signatures or neurovascular interactions without bioengineering interventions. We summarize emerging strategies to decouple maturation milestones from rigid temporal frameworks, emphasizing the synergistic integration of chronological optimization (e.g., vascularized co-cultures) and active bioengineering accelerators (e.g., electrical stimulation and microfluidics). By bridging biological timelines with scalable engineering, this review charts a roadmap to generate translationally relevant, functionally mature brain organoids. Full article
Show Figures

Figure 1

17 pages, 4219 KiB  
Article
Identification of Differentially Expressed Genes and Pathways in Non-Diabetic CKD and Diabetic CKD by Integrated Human Transcriptomic Bioinformatics Analysis
by Clara Barrios, Marta Riera, Eva Rodríguez, Eva Márquez, Jimena del Risco, Melissa Pilco, Jorge Huesca, Ariadna González, Claudia Martyn, Jordi Pujol, Anna Buxeda and Marta Crespo
Int. J. Mol. Sci. 2025, 26(15), 7421; https://doi.org/10.3390/ijms26157421 - 1 Aug 2025
Viewed by 173
Abstract
Chronic kidney disease (CKD) is a heterogeneous condition with various etiologies, including type 2 diabetes mellitus (T2D), hypertension, and autoimmune disorders. Both diabetic CKD (CKD_T2D) and non-diabetic CKD (CKD_nonT2D) share overlapping clinical features, but understanding the molecular mechanisms underlying each subtype and distinguishing [...] Read more.
Chronic kidney disease (CKD) is a heterogeneous condition with various etiologies, including type 2 diabetes mellitus (T2D), hypertension, and autoimmune disorders. Both diabetic CKD (CKD_T2D) and non-diabetic CKD (CKD_nonT2D) share overlapping clinical features, but understanding the molecular mechanisms underlying each subtype and distinguishing diabetic from non-diabetic forms remain poorly defined. To identify differentially expressed genes (DEGs) and enriched biological pathways between CKD_T2D and CKD_nonT2D cohorts, including autoimmune (CKD_nonT2D_AI) and hypertensive (CKD_nonT2D_HT) subtypes, through integrative transcriptomic analysis. Publicly available gene expression datasets from human glomerular and tubulointerstitial kidney tissues were curated and analyzed from GEO and ArrayExpress. Differential expression analysis and Gene Set Enrichment Analysis (GSEA) were conducted to assess cohort-specific molecular signatures. A considerable overlap in DEGs was observed between CKD_T2D and CKD_nonT2D, with CKD_T2D exhibiting more extensive gene expression changes. Hypertensive-CKD shared greater transcriptomic similarity with CKD_T2D than autoimmune-CKD. Key DEGs involved in fibrosis, inflammation, and complement activation—including Tgfb1, Timp1, Cxcl6, and C1qa/B—were differentially regulated in diabetic samples, where GSEA revealed immune pathway enrichment in glomeruli and metabolic pathway enrichment in tubulointerstitium. The transcriptomic landscape of CKD_T2D reveals stronger immune and metabolic dysregulation compared to non-diabetic CKD. These findings suggest divergent pathological mechanisms and support the need for tailored therapeutic approaches. Full article
Show Figures

Figure 1

16 pages, 2762 KiB  
Article
PriorCCI: Interpretable Deep Learning Framework for Identifying Key Ligand–Receptor Interactions Between Specific Cell Types from Single-Cell Transcriptomes
by Hanbyeol Kim, Eunyoung Choi, Yujeong Shim and Joonha Kwon
Int. J. Mol. Sci. 2025, 26(15), 7110; https://doi.org/10.3390/ijms26157110 - 23 Jul 2025
Viewed by 235
Abstract
Understanding the interactions between specific cell types within tissue environments is essential for elucidating key biological processes, such as immune responses, cancer progression, inflammation, and development, in both physiological and pathological studies. The predominant methods for analyzing cell–cell interactions (CCI) rely primarily on [...] Read more.
Understanding the interactions between specific cell types within tissue environments is essential for elucidating key biological processes, such as immune responses, cancer progression, inflammation, and development, in both physiological and pathological studies. The predominant methods for analyzing cell–cell interactions (CCI) rely primarily on statistical inference using mapping or network-based techniques. However, these approaches often struggle to prioritize meaningful interactions owing to the high sparsity and heterogeneity inherent in single-cell RNA sequencing (scRNA-seq) data, where small but biologically important differences can be easily overlooked. To overcome these limitations, we developed PriorCCI, a deep-learning framework that leverages a convolutional neural network (CNN) alongside Grad-CAM++, an explainable artificial intelligence algorithm. This study aims to provide a scalable, interpretable, and biologically meaningful framework for systematically identifying and prioritizing key ligand–receptor interactions between defined cell-type pairs from single-cell RNA-seq data, particularly in complex environments such as tumors. PriorCCI effectively prioritizes interactions between cancer and other cell types within the tumor microenvironment and accurately identifies biologically significant interactions related to angiogenesis. By providing a visual interpretation of gene-pair contributions, our approach enables robust inference of gene–gene interactions across distinct cell types from scRNA-seq data. Full article
(This article belongs to the Special Issue New Insights in Translational Bioinformatics: Second Edition)
Show Figures

Figure 1

21 pages, 5525 KiB  
Article
A High-Throughput ImmunoHistoFluorescence (IHF) Method for Sub-Nuclear Protein Analysis in Tissue
by Kezia Catharina Oxe, Kristoffer Staal Rohrberg, Ulrik Lassen and Dorthe Helena Larsen
Cells 2025, 14(14), 1109; https://doi.org/10.3390/cells14141109 - 18 Jul 2025
Viewed by 471
Abstract
The current understanding of cellular protein distribution in clinical samples is limited. This is partially due to the complexity and heterogeneity of tissues combined with the qualitative nature of analysis by immunohistochemistry (IHC). The common use of manual assessment in the clinic is [...] Read more.
The current understanding of cellular protein distribution in clinical samples is limited. This is partially due to the complexity and heterogeneity of tissues combined with the qualitative nature of analysis by immunohistochemistry (IHC). The common use of manual assessment in the clinic is time-consuming and restricts both the complexity of scoring and the scale of patient tissue analysis. This has limited the transfer of biological observations into pathology and their integration into diagnostics. Immunofluorescence (IF) techniques allow detailed and high-throughput investigation of proteins in cell models, but their application to tissues has been hindered by poor antibody penetration, autofluorescence artefacts, and weak signals. With a growing focus on precision medicine, scalable techniques to investigate and analyse proteins are critically important. To address this, we generated a high-throughput ImmunoHistoFluorescence (IHF) approach, applying IF to tissue samples followed by automated acquisition and artificial intelligence (AI)-based analysis of sub-nuclear protein distribution to enable precise investigation of complex protein localization patterns. This advancement offers a method to transfer in vitro findings into human tissues to analyse protein localization patterns in physiologically relevant contexts for improved understanding of disease-driving mechanisms in patients, identification of new biomarkers, and acceleration of translational research. Full article
(This article belongs to the Special Issue Imaging Methods in Cell Biology)
Show Figures

Graphical abstract

18 pages, 735 KiB  
Review
Co-Occurrence of Endometriosis with Systemic Lupus Erythematosus: Genetic Aspects
by Maria I. Zervou, Theoni B. Tarlatzi, Grigoris F. Grimbizis, Timothy B. Niewold, Basil C. Tarlatzis, George Bertsias and George N. Goulielmos
Int. J. Mol. Sci. 2025, 26(14), 6841; https://doi.org/10.3390/ijms26146841 - 16 Jul 2025
Viewed by 618
Abstract
Previous studies have shown that patients with a history of endometriosis have an increased susceptibility for developing a big number of comorbidities, including various autoimmune diseases. Endometriosis is a complex, inflammatory, estrogen-dependent, heterogeneous gynecological disorder with an incidence of up to 10% in [...] Read more.
Previous studies have shown that patients with a history of endometriosis have an increased susceptibility for developing a big number of comorbidities, including various autoimmune diseases. Endometriosis is a complex, inflammatory, estrogen-dependent, heterogeneous gynecological disorder with an incidence of up to 10% in women of reproductive age. It is characterized by the implantation and growth of endometrial tissue outside the uterus and is associated with dysmenorrhea, deep dyspareunia, pelvic pain and infertility. Systemic lupus erythematosus (SLE) is a chronic, heterogeneous autoimmune disorder of the connective tissue, characterized by impaired innate and adaptive immune responses and the production of pathogenic autoantibodies that drive inflammation and damage in multiple organs. Its etiology is elusive yet associated with high heritability. Importantly, it has been found that endometriosis and SLE share some underlying molecular and cellular pathways. In the present study, we sought to delineate the co-occurrence of endometriosis with SLE from the biological and genetic viewpoint, aiming to identify the putative shared genetic components and clarify the underlying pathogenetic mechanisms. This information may contribute further to the design of new therapeutic protocols for both disorders under study. Full article
Show Figures

Figure 1

29 pages, 1953 KiB  
Review
Targeted Biologic Therapies in Severe Asthma: Mechanisms, Biomarkers, and Clinical Applications
by Renata Maria Văruț, Dop Dalia, Kristina Radivojevic, Diana Maria Trasca, George-Alin Stoica, Niculescu Stefan Adrian, Niculescu Elena Carmen and Cristina Elena Singer
Pharmaceuticals 2025, 18(7), 1021; https://doi.org/10.3390/ph18071021 - 10 Jul 2025
Viewed by 1199
Abstract
Asthma represents a heterogeneous disorder characterized by a dynamic balance between pro-inflammatory and anti-inflammatory forces, with allergic sensitization contributing substantially to airway hyperresponsiveness and remodeling. Central to its pathogenesis are cytokines such as IL-4, IL-5, IL-13, IL-17, and IL-33, which drive recruitment of [...] Read more.
Asthma represents a heterogeneous disorder characterized by a dynamic balance between pro-inflammatory and anti-inflammatory forces, with allergic sensitization contributing substantially to airway hyperresponsiveness and remodeling. Central to its pathogenesis are cytokines such as IL-4, IL-5, IL-13, IL-17, and IL-33, which drive recruitment of eosinophils, neutrophils, and other effector cells, thereby precipitating episodic exacerbations in response to viral and environmental triggers. Conventional biomarkers, including blood and sputum eosinophil counts, IgE levels, and fractional exhaled nitric oxide, facilitate phenotypic classification and guide the emerging biologic era. Monoclonal antibodies targeting IgE (omalizumab) and IL-5 (mepolizumab, benralizumab, reslizumab, depemokimab) have demonstrated the ability to reduce exacerbation frequency and improve lung function, with newer agents such as depemokimab offering extended dosing intervals. Itepekimab, an anti-IL-33 antibody, effectively engages its target and mitigates tissue eosinophilia, while CM310-stapokibart, tralokinumab, and lebrikizumab inhibit IL-4/IL-13 signaling with variable efficacy depending on patient biomarkers. Comparative analyses of these biologics, encompassing affinity, dosing regimens, and trial outcomes, underscore the imperative of personalized therapy to optimize disease control in severe asthma. Full article
Show Figures

Graphical abstract

54 pages, 3159 KiB  
Review
Biomimetic Tumour Model Systems for Pancreatic Ductal Adenocarcinoma in Relation to Photodynamic Therapy
by Olivia M. Smith, Nicole Lintern, Jiahao Tian, Bárbara M. Mesquita, Sabrina Oliveira, Veronika Vymetalkova, Jai Prakash, Andrew M. Smith, David G. Jayne, Michal Heger and Yazan S. Khaled
Int. J. Mol. Sci. 2025, 26(13), 6388; https://doi.org/10.3390/ijms26136388 - 2 Jul 2025
Viewed by 858
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is the most common type of pancreatic cancer and is associated with poor prognosis. Despite years of research and improvements in chemotherapy regimens, the 5-year survival rate of PDAC remains dismal. Therapies for PDAC often face resistance owing in [...] Read more.
Pancreatic ductal adenocarcinoma (PDAC) is the most common type of pancreatic cancer and is associated with poor prognosis. Despite years of research and improvements in chemotherapy regimens, the 5-year survival rate of PDAC remains dismal. Therapies for PDAC often face resistance owing in large part to an extensive desmoplastic stromal matrix. Modelling PDAC ex vivo to investigate novel therapeutics is challenging due to the complex tumour microenvironment and its heterogeneity in native tumours. Development of novel therapies is needed to improve PDAC survival rates, for which disease models that recapitulate the tumour biology are expected to bear utility. This review focuses on the existing preclinical models for human PDAC and discusses advancements in tissue remodelling to guide translational PDAC research. Further emphasis is placed on photodynamic therapy (PDT) due to the ability of this treatment modality to not only directly kill cancer cells by minimally invasive means, but also to perturb the tumour microenvironment and elicit a post-therapeutic anti-tumour immune response. Accordingly, more complex preclinical models that feature multiple biologically relevant PDAC components are needed to develop translatable PDT regimens in a preclinical setting. Full article
(This article belongs to the Special Issue Molecular Advances in Oncologic Photodynamic Therapy)
Show Figures

Figure 1

31 pages, 1423 KiB  
Review
Glioblastoma: Overview of Proteomic Investigations and Biobank Approaches for the Development of a Multidisciplinary Translational Network
by Giusy Ciuffreda, Sara Casati, Francesca Brambilla, Mauro Campello, Valentina De Falco, Dario Di Silvestre, Antonio Frigeri, Marco Locatelli, Lorenzo Magrassi, Andrea Salmaggi, Marco Salvetti, Francesco Signorelli, Yvan Torrente, Giuseppe Emanuele Umana, Raffaello Viganò and Pietro Luigi Mauri
Cancers 2025, 17(13), 2151; https://doi.org/10.3390/cancers17132151 - 26 Jun 2025
Viewed by 807
Abstract
Glioblastoma is a highly aggressive, infiltrative brain tumor of the central nervous system (CNS). Its extensive molecular and biochemical heterogenicity hinders the identification of reliable biomarkers and therapeutic targets, thereby making prognosis and existing therapy ineffective. In recent years, breakthroughs in the use of [...] Read more.
Glioblastoma is a highly aggressive, infiltrative brain tumor of the central nervous system (CNS). Its extensive molecular and biochemical heterogenicity hinders the identification of reliable biomarkers and therapeutic targets, thereby making prognosis and existing therapy ineffective. In recent years, breakthroughs in the use of proteomics on a range of biological samples, such as plasma, cerebrospinal fluid (CSF), tissues, brain cells, and exosomes, represent a potential improvement to GBM investigations. Mass spectrometry-based approaches represent an important technique in the characterization of the tumoral proteome, for the identification of differentially expressed proteins, and for studying altered molecular pathways involved in tumor stages. Proteomics studies advance our knowledge about GBM pathogenesis, the discovery of reliable diagnostic and prognostic markers, and therapeutic approaches, also. In this context, for the effective application of proteomics on GBM, it is mandatory to develop a translational network by integrating hospitals, biobanks, and research institutions into a single network, to enable a collaborative approach across disciplines, thereby enabling rapid translation to clinical application of new proteomic insights. Today, high-quality biobanks play a key role in enabling collaborative, ethically compliant research, supporting the effective application of proteomics in glioblastoma studies and the translation of discoveries into clinical practice. This review explores current trends in proteomics and GBM research, highlighting how leveraging biobank infrastructure and fostering institutional cooperation can drive the development of targeted pilot projects to enhance the impact and effectiveness of glioblastoma research. Full article
(This article belongs to the Section Cancer Therapy)
Show Figures

Figure 1

19 pages, 1191 KiB  
Review
Targeting Senescence: A Review of Senolytics and Senomorphics in Anti-Aging Interventions
by Timur Saliev and Prim B. Singh
Biomolecules 2025, 15(6), 860; https://doi.org/10.3390/biom15060860 - 13 Jun 2025
Cited by 1 | Viewed by 3107
Abstract
Cellular senescence is a fundamental mechanism in aging, marked by irreversible growth arrest and diverse functional changes, including, but not limited to, the development of a senescence-associated secretory phenotype (SASP). While transient senescence contributes to beneficial processes such as tissue repair and tumor [...] Read more.
Cellular senescence is a fundamental mechanism in aging, marked by irreversible growth arrest and diverse functional changes, including, but not limited to, the development of a senescence-associated secretory phenotype (SASP). While transient senescence contributes to beneficial processes such as tissue repair and tumor suppression, the persistent accumulation of senescent cells is implicated in tissue dysfunction, chronic inflammation, and age-related diseases. Notably, the SASP can exert both pro-inflammatory and immunosuppressive effects, depending on cell type, tissue context, and temporal dynamics, particularly in early stages where it may be profibrotic and immunomodulatory. Recent advances in senotherapeutics have led to two principal strategies for targeting senescent cells: senolytics, which selectively induce their apoptosis, and senomorphics, which modulate deleterious aspects of the senescence phenotype, including the SASP, without removing the cells. This review critically examines the molecular mechanisms, therapeutic agents, and clinical potential of both approaches in the context of anti-aging interventions. We discuss major classes of senolytics, such as tyrosine kinase inhibitors, BCL-2 family inhibitors, and natural polyphenols, alongside senomorphics including mTOR and JAK inhibitors, rapalogs, and epigenetic modulators. Additionally, we explore the biological heterogeneity of senescent cells, challenges in developing specific biomarkers, and the dualistic role of senescence in physiological versus pathological states. The review also highlights emerging tools, such as targeted delivery systems, multi-omics integration, and AI-assisted drug discovery, which are advancing precision geroscience and shaping future anti-aging strategies. Full article
(This article belongs to the Special Issue Molecular Advances in Mechanism and Regulation of Lifespan and Aging)
Show Figures

Figure 1

49 pages, 3130 KiB  
Review
Multimodal AI in Biomedicine: Pioneering the Future of Biomaterials, Diagnostics, and Personalized Healthcare
by Nargish Parvin, Sang Woo Joo, Jae Hak Jung and Tapas K. Mandal
Nanomaterials 2025, 15(12), 895; https://doi.org/10.3390/nano15120895 - 10 Jun 2025
Cited by 2 | Viewed by 2194
Abstract
Multimodal artificial intelligence (AI) is driving a paradigm shift in modern biomedicine by seamlessly integrating heterogeneous data sources such as medical imaging, genomic information, and electronic health records. This review explores the transformative impact of multimodal AI across three pivotal areas: biomaterials science, [...] Read more.
Multimodal artificial intelligence (AI) is driving a paradigm shift in modern biomedicine by seamlessly integrating heterogeneous data sources such as medical imaging, genomic information, and electronic health records. This review explores the transformative impact of multimodal AI across three pivotal areas: biomaterials science, medical diagnostics, and personalized medicine. In the realm of biomaterials, AI facilitates the design of patient-specific solutions tailored for tissue engineering, drug delivery, and regenerative therapies. Advanced tools like AlphaFold have significantly improved protein structure prediction, enabling the creation of biomaterials with enhanced biological compatibility. In diagnostics, AI systems synthesize multimodal inputs combining imaging, molecular markers, and clinical data—to improve diagnostic precision and support early disease detection. For precision medicine, AI integrates data from wearable technologies, continuous monitoring systems, and individualized health profiles to inform targeted therapeutic strategies. Despite its promise, the integration of AI into clinical practice presents challenges such as ensuring data security, meeting regulatory standards, and promoting algorithmic transparency. Addressing ethical issues including bias and equitable access remains critical. Nonetheless, the convergence of AI and biotechnology continues to shape a future where healthcare is more predictive, personalized, and responsive. Full article
(This article belongs to the Section Biology and Medicines)
Show Figures

Graphical abstract

27 pages, 2190 KiB  
Review
The Young’s Modulus as a Mechanical Biomarker in AFM Experiments: A Tool for Cancer Diagnosis and Treatment Monitoring
by Stylianos Vasileios Kontomaris, Anna Malamou and Andreas Stylianou
Sensors 2025, 25(11), 3510; https://doi.org/10.3390/s25113510 - 2 Jun 2025
Viewed by 998
Abstract
This review explores recent advances in data processing for atomic force microscopy (AFM) nanoindentation on soft samples, with a focus on “apparent” or “average” Young’s modulus distributions used for cancer diagnosis and treatment monitoring. Young’s modulus serves as a potential key biomarker, distinguishing [...] Read more.
This review explores recent advances in data processing for atomic force microscopy (AFM) nanoindentation on soft samples, with a focus on “apparent” or “average” Young’s modulus distributions used for cancer diagnosis and treatment monitoring. Young’s modulus serves as a potential key biomarker, distinguishing normal from cancerous cells or tissue by assessing stiffness variations at the nanoscale. However, user-independent, reproducible classification remains challenging due to assumptions in traditional mechanics models, particularly Hertzian theory. To enhance accuracy, depth-dependent mechanical properties and polynomial corrections have been introduced to address sample heterogeneity and finite thickness. Additionally, AFM measurements are affected by tip imperfections and the viscoelastic nature of biological samples, requiring careful data processing and consideration of loading conditions. Furthermore, a quantitative approach using distributions of mechanical properties is suitable for tissue classification and for evaluating treatment-induced changes in nanomechanical properties. As part of this review, the use of AFM-based mechanical properties as a tool for monitoring treatment outcomes—including treatments with antifibrotic drugs and photodynamic therapy—is also presented. By analyzing nanomechanical property distributions before and after treatment, AFM provides insights for optimizing therapeutic strategies, reinforcing its role in personalized cancer care and expanding its applications in research and clinical settings. Full article
Show Figures

Figure 1

Back to TopTop