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

Article Types

Countries / Regions

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Search Results (216)

Search Parameters:
Keywords = pattern recognition molecules

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
42 pages, 2435 KB  
Review
HMGB1: A Central Node in Cancer Therapy Resistance
by Bashar A. Alhasan, Boris A. Margulis and Irina V. Guzhova
Int. J. Mol. Sci. 2025, 26(24), 12010; https://doi.org/10.3390/ijms262412010 - 13 Dec 2025
Viewed by 42
Abstract
Cancer therapy resistance emerges from highly integrated molecular systems that enable tumor cells to evade cell death and survive cytotoxic therapeutic stress. High Mobility Group Box 1 (HMGB1) is increasingly gaining recognition as a central coordinator of these resistance programs. This review delineates [...] Read more.
Cancer therapy resistance emerges from highly integrated molecular systems that enable tumor cells to evade cell death and survive cytotoxic therapeutic stress. High Mobility Group Box 1 (HMGB1) is increasingly gaining recognition as a central coordinator of these resistance programs. This review delineates how HMGB1 functions as a molecular switch that dynamically redistributes between cellular compartments in response to stress, with each localization enabling a distinct layer of resistance. In the nucleus, HMGB1 enhances chromatin accessibility and facilitates the recruitment of DNA repair machinery, strengthening resistance to radio- and chemotherapeutic damage. Cytosolic HMGB1 drives pro-survival autophagy, maintains redox stability, and modulates multiple regulated cell death pathways, including apoptosis, ferroptosis, and necroptosis, thereby predominantly shifting cell-fate decisions toward survival under therapeutic pressure. Once released into the extracellular space, HMGB1 acts as a damage-associated molecular pattern (DAMP) that activates key pro-survival and inflammatory signaling pathways, establishing microenvironmental circuits that reinforce malignant progression and therapy escape. HMGB1 further intensifies resistance through upregulation of multidrug resistance transporters, amplifying drug efflux. Together, these compartmentalized functions position HMGB1 as a central node in the networks of cancer therapy resistance. Emerging HMGB1-targeted agents, ranging from peptides and small molecules to receptor antagonists and nanoformulations, show promise in reversing resistance, but clinical translation will require precise, context- and redox-informed HMGB1 targeting to overcome multifactorial resistance program in refractory cancers. Full article
Show Figures

Graphical abstract

28 pages, 7110 KB  
Article
New Dimethylpyridine-3-Carboxamide Derivatives as MMP-13 Inhibitors with Anticancer Activity
by Remigiusz Płaczek, Tomasz Janek, Małgorzata Strzelecka, Aleksandra Kotynia, Piotr Świątek and Żaneta Czyżnikowska
Molecules 2025, 30(24), 4662; https://doi.org/10.3390/molecules30244662 - 5 Dec 2025
Viewed by 258
Abstract
A series of dimethylpyridine-3-carboxamide derivatives was designed as potential, selective, non-zinc chelating inhibitors of matrix metalloproteinase 13 (MMP-13), and subsequently synthesized. The identity of the obtained compounds was confirmed by FT-IR, 1H/13C NMR, and HR-MS methods. Fluorescence spectroscopy was applied [...] Read more.
A series of dimethylpyridine-3-carboxamide derivatives was designed as potential, selective, non-zinc chelating inhibitors of matrix metalloproteinase 13 (MMP-13), and subsequently synthesized. The identity of the obtained compounds was confirmed by FT-IR, 1H/13C NMR, and HR-MS methods. Fluorescence spectroscopy was applied to study the interaction of synthesized compounds with human serum albumin, providing insight into their potential transport properties in plasma. In parallel, the electronic properties and reactivity parameters relevant to enzyme binding of the designed molecules were analyzed using density functional theory. Molecular docking and molecular dynamics simulations revealed the compounds to interact preferentially and stably within the S1 pocket of MMP-13 via hydrogen bonds and π-stacking interactions. The calculated binding free energy confirmed the stability and persistence of the complexes during simulation, indicating a strong and specific recognition pattern. On the other hand, their affinity towards MMP-8 was considerably weaker, which is consistent with the predicted selectivity profile. In addition, the biological evaluation confirmed MMP-13 inhibition. Finally, in vitro tests revealed their cytotoxic activity against cancer cell lines. Full article
Show Figures

Graphical abstract

22 pages, 5784 KB  
Review
An Overview of the Pathogenesis of Cutaneous Lupus Erythematosus
by Alice Verdelli, Emanuela Barletta, Elena Biancamaria Mariotti, Simone Landini, Alessandro Magnatta, Valentina Ruffo di Calabria, Alberto Corrà, Lavinia Quintarelli, Irene Bonanni, Luca Sanna, Virginia Corti and Marzia Caproni
J. Clin. Med. 2025, 14(23), 8285; https://doi.org/10.3390/jcm14238285 - 21 Nov 2025
Viewed by 753
Abstract
Background/Objectives: Cutaneous lupus erythematosus (CLE) is a complex autoimmune skin disease driven by genetic predisposition, environmental triggers, and immune dysregulation. Environmental factors such as ultraviolet radiation, smoking, and certain drugs can initiate disease onset by inducing keratinocyte apoptosis. The subsequent release of nucleic [...] Read more.
Background/Objectives: Cutaneous lupus erythematosus (CLE) is a complex autoimmune skin disease driven by genetic predisposition, environmental triggers, and immune dysregulation. Environmental factors such as ultraviolet radiation, smoking, and certain drugs can initiate disease onset by inducing keratinocyte apoptosis. The subsequent release of nucleic acids and danger-associated molecular patterns activates pattern recognition receptors (PRRs) on keratinocytes and immune cells, leading to the production of type I and type III interferons (IFNs) and pro-inflammatory cytokines. The objective of this review is to summarize recent advances in understanding the immunopathogenesis of CLE, with particular attention to emerging cellular players and their therapeutic implications. Methods: A narrative review of the recent literature was performed, including experimental, translational, and clinical studies investigating the cellular and molecular mechanisms underlying CLE and novel targeted treatments derived from these findings. Results: Although plasmacytoid dendritic cells (pDCs) have traditionally been considered the major producers of IFN-I, recent data indicate that pDCs in CLE are functionally impaired and are not the primary source. Other cells, such as keratinocytes have emerged as key producers of IFN-I, contributing to a prelesional, IFN-rich microenvironment. This promotes the recruitment and activation of dendritic cells and other inflammatory myeloid subsets, which are now recognized as central players in amplifying local inflammation. Concurrently, T cells infiltrate the skin, where cytotoxic CD8+ T cells attack keratinocytes and CD4+ T cells further propagate inflammation via cytokine production. B cells and plasma cells produce autoantibodies, forming immune complexes that perpetuate inflammation. Neutrophils release neutrophil extracellular traps (NETs), exposing autoantigens and further stimulating IFN pathways. Macrophages contribute by presenting autoantigens, producing pro-inflammatory mediators, and failing to effectively clear apoptotic cells and immune complexes. Conclusions: The dynamic interplay between the innate and adaptive immune systems sustains the chronic inflammatory state characteristic of CLE. Based on the pathogenetic novelties, new therapeutic agents targeting specific molecules have been developed, which may improve the treatment of this complex disease in the future. Full article
(This article belongs to the Special Issue Skin Diseases: From Diagnosis to Treatment)
Show Figures

Figure 1

36 pages, 2919 KB  
Review
Viewing Inflammation and Immunoregulation Under the Calpain System Lens
by Vijay Kumar and John H. Stewart
Cells 2025, 14(22), 1814; https://doi.org/10.3390/cells14221814 - 19 Nov 2025
Viewed by 736
Abstract
The controlled pro-inflammatory immune response is critical for fighting against external and endogenous threats, such as microbes/pathogens, allergens, xenobiotics, various antigens, and dying host cells and their mediators (DNA, RNA, and nuclear proteins) released into the circulation and cytosol (PAMPs, MAMPs, and DAMPs). [...] Read more.
The controlled pro-inflammatory immune response is critical for fighting against external and endogenous threats, such as microbes/pathogens, allergens, xenobiotics, various antigens, and dying host cells and their mediators (DNA, RNA, and nuclear proteins) released into the circulation and cytosol (PAMPs, MAMPs, and DAMPs). Several pattern recognition receptors (PRRs) and their downstream adaptor molecules, expressed by innate and adaptive immune cells, are critical in generating the inflammatory immune response by recognizing PAMPs, MAMPs, and DAMPs. However, their dysregulation may predispose the host to develop inflammation-associated organ damage, neurodegeneration, autoimmunity, cancer, and even death due to the absence of the inflammation resolution phase. The cytosolic calcium (Ca2+) level regulates the survival, proliferation, and immunological functions of immune cells. Cysteine-rich proteases, specifically calpains, are Ca2+-dependent proteases that become activated during inflammatory conditions, playing a critical role in the inflammatory process and associated organ damage. Therefore, this article discusses the expression and function of calpain-1 and calpain-2 (ubiquitous calpains) in various innate (epithelial, endothelial, dendritic, mast, and NK cells, as well as macrophages) and adaptive (T and B cells) immune cells, affecting inflammation and immune regulation. As inflammatory diseases are on the rise due to several factors, such as environment, lifestyle, and an aging population, we must not just investigate but strive for a deeper understanding of the inflammation and immunoregulation under the calpain system (calpain-1 and calpain-2 and their endogenous negative regulator calpastatin) lens, which is ubiquitous and senses cytosolic Ca2+ changes to impact immune response. Full article
Show Figures

Figure 1

27 pages, 8542 KB  
Article
The Receptor for Advanced Glycation End-Products (RAGE) Regulates Cell Adhesion Through Upregulation of ITGA8
by Swetha Thiyagarajan, Estelle Leclerc and Stefan W. Vetter
Cells 2025, 14(22), 1805; https://doi.org/10.3390/cells14221805 - 17 Nov 2025
Viewed by 493
Abstract
The Receptor for Advanced Glycation End-Products (RAGE) is a cell surface receptor of the immunoglobulin-like receptor superfamily. RAGE is a pattern-recognition, multi-ligand receptor that binds glycated proteins, specific non-glycated proteins, and nucleic acids. RAGE ligands are typically part of the group of damage-associated [...] Read more.
The Receptor for Advanced Glycation End-Products (RAGE) is a cell surface receptor of the immunoglobulin-like receptor superfamily. RAGE is a pattern-recognition, multi-ligand receptor that binds glycated proteins, specific non-glycated proteins, and nucleic acids. RAGE ligands are typically part of the group of damage-associated molecular patterns (DAMPs) or alarmins. As such, RAGE is a receptor for molecular products of cellular stress, abnormal metabolism, and inflammation. Activation of RAGE by its ligands leads to pro-inflammatory signaling, often resulting in persistent RAGE activation in various disease states. Consequently, RAGE has been investigated as a potential drug target in the treatment of diabetic complications, vascular disease, Alzheimer’s disease, and multiple types of cancer. An underexplored aspect of RAGE is its role in cell adhesion. Structural comparison of the extracellular domain of RAGE has revealed structural similarity to the activated leukocyte cell adhesion molecule (ALCAM). The present study reveals the role and mechanism of RAGE in regulating cell adhesion. We investigated the role of individual RAGE domains in cell adhesion to extracellular matrix proteins and the changes in protein expression resulting from RAGE upregulation. Key findings include that RAGE displays substrate-specific adhesion to extracellular matrix proteins, that the intracellular domain of RAGE is required for modulating cell spreading, and that regulation of ITGA8 depends on the cytoplasmic domain of RAGE. Full article
(This article belongs to the Section Cell Motility and Adhesion)
Show Figures

Figure 1

21 pages, 3884 KB  
Article
CpG ODN Activates TLR9 and Upregulates TLR3 via the p38 MAPK-ATF3 Signaling Axis to Synergistically Enhance Dendritic Cell Vaccine Efficacy
by Lv Zhou, Zhuowei Lei, Qian Jiang, Linpeng Xu, Quanji Wang, Yimin Huang and Ting Lei
Cells 2025, 14(22), 1785; https://doi.org/10.3390/cells14221785 - 13 Nov 2025
Viewed by 668
Abstract
Toll-like receptor 9 (TLR9) and Toll-like receptor 3 (TLR3), which are widely expressed in dendritic cells (DCs), function as key pattern recognition receptors (PRRs) in the immune system. Their primary roles involve specifically detecting pathogen-associated molecular patterns (PAMPs): TLR9 recognizes unmethylated CpG motifs [...] Read more.
Toll-like receptor 9 (TLR9) and Toll-like receptor 3 (TLR3), which are widely expressed in dendritic cells (DCs), function as key pattern recognition receptors (PRRs) in the immune system. Their primary roles involve specifically detecting pathogen-associated molecular patterns (PAMPs): TLR9 recognizes unmethylated CpG motifs predominantly found in bacterial and viral DNA, while TLR3 identifies viral double-stranded RNA (dsRNA), a molecular signature associated with viral replication. Their specific agonists [CpG ODN (a TLR9 agonist) and poly(I:C) (a TLR3 agonist)] can effectively activate DCs and enhance the expression of immune activation-related molecules. In this study, by establishing a mouse primary dendritic cell model and a glioma-bearing mouse model, and employing techniques such as transcriptome sequencing, we found that combined stimulation with CpG ODN and poly(I:C) significantly enhanced the anti-tumor function of DCs: in vitro, DCs subjected to combined stimulation showed upregulation of anti-tumor-related surface markers, enhanced migratory capacity, and a more effective activation of CD8+ T cells; in vivo, a DC vaccine loaded with tumor lysate antigen and stimulated with this combined regimen significantly delayed the progression of glioma in tumor-bearing mice. Further investigation revealed that the underlying mechanism for this enhanced effect may involve TLR9 activation promoting TLR3 upregulation through the p38 MAPK-ATF3 signaling axis. Consequently, we designed a sequential stimulation protocol (first CpG ODN then poly(I:C)), which demonstrated a stronger anti-glioma effect compared to simple combined stimulation. This study provides a new strategy for enhancing the immune efficacy of DC vaccines and has potential significance for promoting the clinical translation of DC vaccines. Full article
(This article belongs to the Topic Advances in Glioblastoma: From Biology to Therapeutics)
Show Figures

Graphical abstract

25 pages, 4105 KB  
Review
Structural and Functional Insights into Viral and Fungal Proteins Involved in Chronic Inflammation and Their Biologic Treatments
by Mohamed Halawa, Alicia L. Gallo and Valerie J. Carabetta
Pharmaceutics 2025, 17(11), 1466; https://doi.org/10.3390/pharmaceutics17111466 - 13 Nov 2025
Viewed by 647
Abstract
Chronic inflammation constitutes a significant characteristic of sustained infections caused by viral and fungal pathogens, with a strong correlation to the development of cancer, autoimmune disorders, and tissue fibrosis. Viral proteins such as HIV-1 Tat, HBV X (HBx), HPV E6/E7, and EBV LMP1 [...] Read more.
Chronic inflammation constitutes a significant characteristic of sustained infections caused by viral and fungal pathogens, with a strong correlation to the development of cancer, autoimmune disorders, and tissue fibrosis. Viral proteins such as HIV-1 Tat, HBV X (HBx), HPV E6/E7, and EBV LMP1 modulate the host’s immune signaling pathways, primarily through the activation of the NF-κB signaling cascade and the disruption of cytokine equilibrium. These molecular interactions result in a pro-inflammatory microenvironment that facilitates viral persistence, immune evasion, and the process of oncogenesis. Structural investigations have elucidated the mechanisms by which these viral proteins interact with host signaling complexes, thereby highlighting their potential as viable therapeutic targets. Similarly, fungal proteins, including secreted aspartyl proteases (Saps), ribotoxin Asp f1, and chitin-binding proteins, incite chronic inflammation by activating pattern recognition receptors and triggering inflammasome activation. Despite the limited structural information of these fungal proteins, emerging models and bioinformatic analyses identified conserved motifs that are crucial for host interactions. Biologic therapies, encompassing antiviral and antifungal peptides as well as monoclonal antibodies, are currently under development to disrupt these protein-host interactions and modulate inflammatory responses. This review provides structural and functional insight into viral and fungal inflammatory proteins and evaluates the potential of biologics as targeted therapeutic interventions for chronic inflammation associated with infections. We discuss the ongoing clinical trials involving neutralizing antibodies targeting HIV, peptide vaccines aimed at HPV and other promising molecules. Finally, we discuss the current limitations of biologics and possible solutions to translate these promising therapeutics into clinical practice. Full article
(This article belongs to the Special Issue Antibody–Drug Conjugates Therapeutics)
Show Figures

Graphical abstract

24 pages, 751 KB  
Review
Integrating Advanced Metabolomics and Machine Learning for Anti-Doping in Human Athletes
by Mohannad N. AbuHaweeleh, Ahmad Hamdan, Jawaher Al-Essa, Shaikha Aljaal, Nasser Al Saad, Costas Georgakopoulos, Francesco Botre and Mohamed A. Elrayess
Metabolites 2025, 15(11), 696; https://doi.org/10.3390/metabo15110696 - 27 Oct 2025
Viewed by 1412
Abstract
The ongoing challenge of doping in sports has triggered the adoption of advanced scientific strategies for the detection and prevention of doping abuse. This review examines the potential of integrating metabolomics aided by artificial intelligence (AI) and machine learning (ML) for profiling small-molecule [...] Read more.
The ongoing challenge of doping in sports has triggered the adoption of advanced scientific strategies for the detection and prevention of doping abuse. This review examines the potential of integrating metabolomics aided by artificial intelligence (AI) and machine learning (ML) for profiling small-molecule metabolites across biological systems to advance anti-doping efforts. While traditional targeted detection methods serve a primarily forensic role—providing legally defensible evidence by directly identifying prohibited substances—metabolomics offers complementary insights by revealing both exogenous compounds and endogenous physiological alterations that may persist beyond direct drug detection windows, rather than serving as an alternative to routine forensic testing. High-throughput platforms such as UHPLC-HRMS and NMR, coupled with targeted and untargeted metabolomic workflows, can provide comprehensive datasets that help discriminate between doped and clean athlete profiles. However, the complexity and dimensionality of these datasets necessitate sophisticated computational tools. ML algorithms, including supervised models like XGBoost and multi-layer perceptrons, and unsupervised methods such as clustering and dimensionality reduction, enable robust pattern recognition, classification, and anomaly detection. These approaches enhance both the sensitivity and specificity of diagnostic screening and optimize resource allocation. Case studies illustrate the value of integrating metabolomics and ML—for example, detecting recombinant human erythropoietin (r-HuEPO) use via indirect blood markers and uncovering testosterone and corticosteroid abuse with extended detection windows. Future progress will rely on interdisciplinary collaboration, open-access data infrastructure, and continuous methodological innovation to fully realize the complementary role of these technologies in supporting fair play and athlete well-being. Full article
(This article belongs to the Special Issue Artificial Intelligence and Machine Learning in Metabolomics)
Show Figures

Graphical abstract

18 pages, 1867 KB  
Article
Factors Driving Amyloid Beta Fibril Recognition by Cell Surface Receptors: A Computational Study
by Olivia Slater and Maria Kontoyianni
Molecules 2025, 30(20), 4116; https://doi.org/10.3390/molecules30204116 - 17 Oct 2025
Viewed by 584
Abstract
Alzheimer’s disease (AD) has been studied extensively and is characterized by plaques deposited throughout the brain. Plaques are made of beta-amyloid (Aβ) peptides which have undergone fibrillogenesis to form insoluble Aβ fibrils (fAβ) that are neurotoxic. Receptor for [...] Read more.
Alzheimer’s disease (AD) has been studied extensively and is characterized by plaques deposited throughout the brain. Plaques are made of beta-amyloid (Aβ) peptides which have undergone fibrillogenesis to form insoluble Aβ fibrils (fAβ) that are neurotoxic. Receptor for Advanced Glycation End end products (RAGE), toll-like receptors (TLRs) 2 and 4, and co-receptor CD14 recognize negatively charged binding regions on fAβ to activate microglia and release proinflammatory cytokines. In this study, we used two experimentally resolved fAβ structures (type I and II) isolated from AD brain tissue to elucidate binding patterns of fAβ with RAGE, TLR2, TLR4, and CD14 and investigated whether binding was affected by fibril structure or system pH. Receptors TLR2 and RAGE formed tight complexes with both type I and II fibrils, while TLR4 showed selectivity for type I. CD14 binding was less tight and selective for type II. Binding was pH dependent for CD14, TLR4, and RAGE but not TLR2. We explored the effects of familial mutations on fibril structure to determine whether mutants of type I or II structures are feasible. Finally, we investigated whether mutations affected binding interactions of fAβ with proteins. The Arctic (Glu22Gly), Dutch (Glu22Gln), and Iowa (Asp23Asn) mutations showed similar effects on binding affinity. Italian (Glu22Lys) mutations abrogated binding, whereas type I and II fibrils with Flemish (Ala21Gly) mutations were not shown to be feasible. Results highlight the adaptability of immune receptors in recognizing damaging molecules, with fibril structure and pH being the main recognition determinants predicated on disease progression. In silico mutations showed that aggregates similar to type I and II structures were plausible for some familial mutations. Full article
(This article belongs to the Section Computational and Theoretical Chemistry)
Show Figures

Figure 1

43 pages, 10755 KB  
Review
PRRs-Dependent and Independent Mechanisms of STING Signaling in Inflammatory and Autoimmune Diseases
by Le Xu, Jingrou Li, Xingchen Zhu, Liting Zhou, Zhirong Sun, Zhipeng Zhang, Wei Xu and Yahui Song
Biomedicines 2025, 13(10), 2533; https://doi.org/10.3390/biomedicines13102533 - 17 Oct 2025
Cited by 1 | Viewed by 1632
Abstract
The stimulator of interferon genes (STING) serves as a pivotal signaling hub in innate immunity, orchestrating type I interferon (IFN-I) and pro-inflammatory responses upon detection of cytosolic DNA. While the canonical cyclic GMP-AMP synthase (cGAS)-STING axis has been extensively studied in host defense [...] Read more.
The stimulator of interferon genes (STING) serves as a pivotal signaling hub in innate immunity, orchestrating type I interferon (IFN-I) and pro-inflammatory responses upon detection of cytosolic DNA. While the canonical cyclic GMP-AMP synthase (cGAS)-STING axis has been extensively studied in host defense and sterile inflammation, increasing evidence indicates that STING can also be activated through a variety of both pattern recognition receptors (PRRs)-dependent and PRRs-independent mechanisms. In this review, we comprehensively summarize the molecular pathways through which PRRs—including cGAS, interferon gamma inducible protein 16 (IFI16), DEAD-box helicase 41 (DDX41), and DNA-dependent protein kinase (DNA-PK)—engage and regulate STING activation. Beyond PRRs-triggered pathways, we explore emerging evidence of PRRs-independent STING activation, driven by genetic mutations, endoplasmic reticulum (ER) stress, dysregulated intracellular trafficking, and impaired protein degradation. These mechanisms contribute to the pathogenesis of a broad spectrum of inflammatory and autoimmune disorders affecting multiple organ systems, including the digestive, cardiovascular, renal, pulmonary, and nervous systems. We also highlight the current landscape of pharmacological inhibitors targeting cGAS and STING, categorized according to their mechanisms of action and therapeutic potential. The redundancy and complexity of components within the STING signaling network present challenges in effectively suppressing inflammatory overactivation by targeting a single molecule. Nevertheless, the central role of STING offers multiple opportunities for therapeutic intervention, whether by modulating upstream or downstream signaling elements. This review not only provides a systematic framework for understanding the intricacies of STING signaling, but offers insights into the development of next-generation therapeutics aimed at selectively modulating STING activity in disease contexts. Full article
Show Figures

Figure 1

20 pages, 2459 KB  
Review
The Immunoregulatory Mechanisms of Human Cytomegalovirus from Primary Infection to Reactivation
by Xiaodan Liu, Chang Liu and Ting Zhang
Pathogens 2025, 14(10), 998; https://doi.org/10.3390/pathogens14100998 - 2 Oct 2025
Cited by 1 | Viewed by 1832
Abstract
Human cytomegalovirus (HCMV) establishes lifelong latency following primary infection, residing within myeloid progenitor cells and monocytes. To achieve this, the virus employs multiple immune evasion strategies. It suppresses innate immune signaling by inhibiting Toll-like receptor and cGAS-STING pathways. In addition, the virus suppresses [...] Read more.
Human cytomegalovirus (HCMV) establishes lifelong latency following primary infection, residing within myeloid progenitor cells and monocytes. To achieve this, the virus employs multiple immune evasion strategies. It suppresses innate immune signaling by inhibiting Toll-like receptor and cGAS-STING pathways. In addition, the virus suppresses major histocompatibility complex (MHC)-dependent antigen presentation to evade T cell recognition. As the downregulation of MHC molecules may trigger NK cell activation, the virus compensates for this by expressing proteins such as UL40 and IL-10, which engage inhibitory NK cell receptors and block activating signals, thereby suppressing NK cell immune surveillance. Viral proteins like UL36 and UL37 block host cell apoptosis and necroptosis, allowing HCMV to persist undetected and avoid clearance. In settings of profound immunosuppression, such as after allogeneic hematopoietic stem cell transplantation (allo-HSCT) or solid organ transplantation, slow immune reconstitution creates a window for viral reactivation. Likewise, immunosenescence and chronic low-grade inflammation during aging increases the risk of reactivation. Once reactivated, HCMV triggers programmed cell death, releasing viral PAMPs (pathogen-associated molecular patterns) and host-derived DAMPs (damage-associated molecular patterns). This release fuels a potent inflammatory response, promoting further viral reactivation and exacerbating tissue damage, creating a vicious cycle. This cycle of inflammation and reactivation contributes to both transplant-related complications and the decline of antiviral immunity in the elderly. Therefore, understanding the immune regulatory mechanisms that govern the switch from latency to reactivation is critical, especially within the unique immune landscapes of transplantation and aging. Elucidating these pathways is essential for developing strategies to prevent and treat HCMV-related disease in these high-risk populations. Full article
(This article belongs to the Special Issue Pathogen–Host Interactions: Death, Defense, and Disease)
Show Figures

Figure 1

27 pages, 2674 KB  
Review
Small RNA and Epigenetic Control of Plant Immunity
by Sopan Ganpatrao Wagh, Akshay Milind Patil, Ghanshyam Bhaurao Patil, Sumeet Prabhakar Mankar, Khushboo Rastogi and Masamichi Nishiguchi
DNA 2025, 5(4), 47; https://doi.org/10.3390/dna5040047 - 1 Oct 2025
Viewed by 1935
Abstract
Plants have evolved a complex, multilayered immune system that integrates molecular recognition, signaling pathways, epigenetic regulation, and small RNA-mediated control. Recent studies have shown that DNA-level regulatory mechanisms, such as RNA-directed DNA methylation (RdDM), histone modifications, and chromatin remodeling, are critical for modulating [...] Read more.
Plants have evolved a complex, multilayered immune system that integrates molecular recognition, signaling pathways, epigenetic regulation, and small RNA-mediated control. Recent studies have shown that DNA-level regulatory mechanisms, such as RNA-directed DNA methylation (RdDM), histone modifications, and chromatin remodeling, are critical for modulating immune gene expression, allowing for rapid and accurate pathogen-defense responses. The epigenetic landscape not only maintains immunological homeostasis but also promotes stress-responsive transcription via stable chromatin modifications. These changes contribute to immunological priming, a process in which earlier exposure to pathogens or abiotic stress causes a heightened state of preparedness for future encounters. Small RNAs, including siRNAs, miRNAs, and phasiRNAs, are essential for gene silencing before and after transcription, fine-tuning immune responses, and inhibiting negative regulators. These RNA molecules interact closely with chromatin features, influencing histone acetylation/methylation (e.g., H3K4me3, H3K27me3) and guiding DNA methylation patterns. Epigenetically encoded immune memory can be stable across multiple generations, resulting in the transgenerational inheritance of stress resilience. Such memory effects have been observed in rice, tomato, maize, and Arabidopsis. This review summarizes new findings on short RNA biology, chromatin-level immunological control, and epigenetic memory in plant defense. Emerging technologies, such as ATAC-seq (Assay for Transposase-Accessible Chromatin using Sequencing), ChIP-seq (Chromatin Immunoprecipitation followed by Sequencing), bisulfite sequencing, and CRISPR/dCas9-based epigenome editing, are helping researchers comprehend these pathways. These developments hold an opportunity for establishing epigenetic breeding strategies that target the production of non-GMO, stress-resistant crops for sustainable agriculture. Full article
Show Figures

Figure 1

22 pages, 1934 KB  
Review
Deep Learning-Driven Intelligent Fluorescent Probes: Advancements in Molecular Design for Accurate Food Safety Detection
by Yongqiang Shi, Sisi Yang, Wenting Li, Yuqing Wu and Weiran Luo
Foods 2025, 14(17), 3114; https://doi.org/10.3390/foods14173114 - 5 Sep 2025
Cited by 1 | Viewed by 1701
Abstract
The complexity of global food supply chains challenges public health, requiring advanced detection technologies beyond traditional lab methods. Fluorescent sensing, known for its sensitivity and quick response, is promising for food safety but hindered by inefficient probe design and difficulties in analyzing complex [...] Read more.
The complexity of global food supply chains challenges public health, requiring advanced detection technologies beyond traditional lab methods. Fluorescent sensing, known for its sensitivity and quick response, is promising for food safety but hindered by inefficient probe design and difficulties in analyzing complex signals in food. Deep Learning (DL) offers solutions with its nonlinear modeling and pattern recognition capabilities. This review explores recent advancements in DL applications for fluorescent sensing. We explore deep learning methods for predicting fluorescent probe properties and generating fluorescent molecule structures, highlighting their role in accelerating high-performance probe development. We then offer a detailed discussion on the pivotal technologies of deep learning in the intelligent analysis of complex fluorescent signals. On this basis, we engage in a thorough reflection on the core challenges presently confronting the field and propose a forward-looking perspective on the future developmental trajectories of fluorescent sensing technology, offering a comprehensive and insightful roadmap for future research in this interdisciplinary domain. Full article
Show Figures

Figure 1

29 pages, 8644 KB  
Review
Recent Advances in Resistive Gas Sensors: Fundamentals, Material and Device Design, and Intelligent Applications
by Peiqingfeng Wang, Shusheng Xu, Xuerong Shi, Jiaqing Zhu, Haichao Xiong and Huimin Wen
Chemosensors 2025, 13(7), 224; https://doi.org/10.3390/chemosensors13070224 - 21 Jun 2025
Cited by 5 | Viewed by 3875
Abstract
Resistive gas sensors have attracted significant attention due to their simple architecture, low cost, and ease of integration, with widespread applications in environmental monitoring, industrial safety, and healthcare diagnostics. This review provides a comprehensive overview of recent advances in resistive gas sensors, focusing [...] Read more.
Resistive gas sensors have attracted significant attention due to their simple architecture, low cost, and ease of integration, with widespread applications in environmental monitoring, industrial safety, and healthcare diagnostics. This review provides a comprehensive overview of recent advances in resistive gas sensors, focusing on their fundamental working mechanisms, sensing material design, device architecture optimization, and intelligent system integration. These sensors primarily operate based on changes in electrical resistance induced by interactions between gas molecules and sensing materials, including physical adsorption, charge transfer, and surface redox reactions. In terms of materials, metal oxide semiconductors, conductive polymers, carbon-based nanomaterials, and their composites have demonstrated enhanced sensitivity and selectivity through strategies such as doping, surface functionalization, and heterojunction engineering, while also enabling reduced operating temperatures. Device-level innovations—such as microheater integration, self-heated nanowires, and multi-sensor arrays—have further improved response speed and energy efficiency. Moreover, the incorporation of artificial intelligence (AI) and Internet of Things (IoT) technologies has significantly advanced signal processing, pattern recognition, and long-term operational stability. Machine learning (ML) algorithms have enabled intelligent design of novel sensing materials, optimized multi-gas identification, and enhanced data reliability in complex environments. These synergistic developments are driving resistive gas sensors toward low-power, highly integrated, and multifunctional platforms, particularly in emerging applications such as wearable electronics, breath diagnostics, and smart city infrastructure. This review concludes with a perspective on future research directions, emphasizing the importance of improving material stability, interference resistance, standardized fabrication, and intelligent system integration for large-scale practical deployment. Full article
Show Figures

Figure 1

16 pages, 1632 KB  
Review
Innate Immune Sensing of Parapoxvirus Orf Virus and Viral Immune Evasion
by Basheer A. AlDaif and Stephen B. Fleming
Viruses 2025, 17(4), 587; https://doi.org/10.3390/v17040587 - 19 Apr 2025
Cited by 3 | Viewed by 1278
Abstract
Orf virus (ORFV) is the type species of Parapoxvirus of the Poxviridae family that induces cutaneous pustular skin lesions in sheep and goats, and causes zoonotic infections in humans. Pattern recognition receptors (PRRs) sense pathogen-associated molecular patterns (PAMPs), leading to the triggering of [...] Read more.
Orf virus (ORFV) is the type species of Parapoxvirus of the Poxviridae family that induces cutaneous pustular skin lesions in sheep and goats, and causes zoonotic infections in humans. Pattern recognition receptors (PRRs) sense pathogen-associated molecular patterns (PAMPs), leading to the triggering of the innate immune response through multiple signalling pathways involving type I interferons (IFNs). The major PAMPs generated during viral infection are nucleic acids, which are the most important molecules that are recognized by the host. The induction of type l IFNs leads to activation of the Janus kinase (JAK)-signal transducer activator of transcription (STAT) pathway, which results in the induction of hundreds of interferon-stimulated genes (ISGs), many of which encode proteins that have antiviral roles in eliminating virus infection and create an antiviral state. Genetic and functional analyses have revealed that ORFV, as found for other poxviruses, has evolved multiple immunomodulatory genes and strategies that manipulate the innate immune sensing response. Full article
(This article belongs to the Section Viral Immunology, Vaccines, and Antivirals)
Show Figures

Figure 1

Back to TopTop