Sign in to use this feature.

Years

Between: -

Subjects

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Journals

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

Article Types

Countries / Regions

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Search Results (1,690)

Search Parameters:
Keywords = direct protein binding

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
25 pages, 1775 KB  
Review
Advances in Mammalian Metallomics: New Insights into Metal Dynamics and Biological Significance
by Xin Tian, Yifan Teng, Yuhang Deng, Qian Zhang, Caihong Hu and Jie Feng
Int. J. Mol. Sci. 2025, 26(19), 9729; https://doi.org/10.3390/ijms26199729 (registering DOI) - 6 Oct 2025
Abstract
Mammalian metallomics, an advanced interdisciplinary field, explores the dynamic roles of metal elements within biological systems and their significance to life processes. While prior reviews have broadly covered metallomics across different systems, this review narrows the focus to mammals, offering new insights into [...] Read more.
Mammalian metallomics, an advanced interdisciplinary field, explores the dynamic roles of metal elements within biological systems and their significance to life processes. While prior reviews have broadly covered metallomics across different systems, this review narrows the focus to mammals, offering new insights into the physiological roles of metal elements, their complex absorption and transport mechanisms, and their intricate associations with diseases. We summarize the characteristics and applications of common metal detection technologies and elaborate on the dynamic landscape of the mammalian metallomics across different tissues and life stages. Furthermore, we elaborate on the physiological functions of the metals from three perspectives, metal-binding proteins, metal ions, and gut microorganisms, and highlight the potential of metallomics in clinical translation, including its diagnostic and therapeutic implications, alongside future directions centered on multi-omics integration. Overall, this review introduces several common metallomics technologies and synthesizes the findings of mammalian metallomics research from multiple perspectives, offering new insights for future related studies. Full article
(This article belongs to the Special Issue The Role of Trace Elements in Nutrition and Health)
Show Figures

Figure 1

29 pages, 15230 KB  
Article
Harpagide Confers Protection Against Acute Lung Injury Through Multi-Omics Dissection of Immune–Microenvironmental Crosstalk and Convergent Therapeutic Mechanisms
by Hong Wang, Jicheng Yang, Yusheng Zhang, Jie Wang, Shaoqi Song, Longhui Gao, Mei Liu, Zhiliang Chen and Xianyu Li
Pharmaceuticals 2025, 18(10), 1494; https://doi.org/10.3390/ph18101494 - 4 Oct 2025
Abstract
Background: Acute lung injury (ALI) and its severe form, acute respiratory distress syndrome (ARDS), remain major causes of morbidity and mortality, yet no targeted pharmacological therapy is available. Excessive neutrophil and macrophage infiltration drives reactive oxygen species (ROS) production and cytokine release, leading [...] Read more.
Background: Acute lung injury (ALI) and its severe form, acute respiratory distress syndrome (ARDS), remain major causes of morbidity and mortality, yet no targeted pharmacological therapy is available. Excessive neutrophil and macrophage infiltration drives reactive oxygen species (ROS) production and cytokine release, leading to alveolar–capillary barrier disruption and fatal respiratory failure. Methods: We applied an integrative multi-omics strategy combining single-cell transcriptomics, peripheral blood proteomics, and lung tissue proteomics in a lipopolysaccharide (LPS, 10 mg/kg)-induced mouse ALI model to identify key signaling pathways. Harpagide, an iridoid glycoside identified from our natural compound screen, was evaluated in vivo (40 and 80 mg/kg) and in vitro (0.1–1 mg/mL). Histopathology, oxidative stress markers (SOD, GSH, and MDA), cytokine levels (IL-6 and IL-1β), and signaling proteins (HIF-1α, p-PI3K, p-AKT, Nrf2, and HO-1) were quantitatively assessed. Direct target engagement was probed using surface plasmon resonance (SPR), the cellular thermal shift assay (CETSA), and 100 ns molecular dynamics (MD) simulations. Results: Multi-omics profiling revealed robust activation of HIF-1, PI3K/AKT, and glutathione-metabolism pathways following the LPS challenge, with HIF-1α, VEGFA, and AKT as core regulators. Harpagide treatment significantly reduced lung injury scores by ~45% (p < 0.01), collagen deposition by ~50%, and ROS accumulation by >60% relative to LPS (n = 6). The pro-inflammatory cytokines IL-6 and IL-1β were reduced by 55–70% at the protein level (p < 0.01). Harpagide dose-dependently suppressed HIF-1α and p-AKT expression while enhancing Nrf2 and HO-1 levels (p < 0.05). SPR confirmed direct binding of Harpagide to HIF-1α (KD = 8.73 µM), and the CETSA demonstrated enhanced thermal stability of HIF-1α. MD simulations revealed a stable binding conformation within the inhibitory/C-TAD region after 50 ns. Conclusions: This study reveals convergent immune–microenvironmental regulatory mechanisms across cellular and tissue levels in ALI and demonstrates the protective effects of Harpagide through multi-pathway modulation. These findings offer new insights into the pathogenesis of ALI and support the development of “one-drug, multilayer co-regulation” strategies for systemic inflammatory diseases. Full article
(This article belongs to the Section Pharmacology)
Show Figures

Graphical abstract

14 pages, 2044 KB  
Article
Molecular Characterization of Wilson’s Disease in Liver Transplant Patients: A Five-Year Single-Center Experience in Iran
by Zahra Beyzaei, Melika Majed, Seyed Mohsen Dehghani, Mohammad Hadi Imanieh, Ali Khazaee, Bita Geramizadeh and Ralf Weiskirchen
Diagnostics 2025, 15(19), 2504; https://doi.org/10.3390/diagnostics15192504 - 1 Oct 2025
Abstract
Background/Objectives: Wilson’s disease (WD) is an autosomal recessive disorder characterized by pathological copper accumulation, primarily in the liver and brain. Severe hepatic involvement can be effectively treated with liver transplantation (LT). Geographic variation in ATP7B mutations suggests the presence of regional patterns [...] Read more.
Background/Objectives: Wilson’s disease (WD) is an autosomal recessive disorder characterized by pathological copper accumulation, primarily in the liver and brain. Severe hepatic involvement can be effectively treated with liver transplantation (LT). Geographic variation in ATP7B mutations suggests the presence of regional patterns that may impact disease presentation and management. This study aims to investigate the genetic basis of WD in patients from a major LT center in Iran. Methods: A retrospective analysis was conducted on clinical, biochemical, and pathological data from patients suspected of WD who underwent evaluation for LT between May 2020 and June 2025 at Shiraz University of Medical Sciences. Genetic testing was carried out on 20 patients at the Shiraz Transplant Research Center (STRC). Direct mutation analysis of ATP7B was performed for all patients, and the results correlated with clinical and demographic information. Results: In total, 20 WD patients who underwent liver transplantation (15 males, 5 females) carried 25 pathogenic or likely pathogenic ATP7B variants, 21 of which were previously unreported. Fifteen patients were homozygous, and five were compound-heterozygous; all heterozygous combinations occurred in the offspring of second-degree consanguineous unions. Recurrent changes included p.L549V, p.V872E, and p.P992S/L, while two nonsense variants (p.E1293X, p.R1319X) predicted truncated proteins. Variants were distributed across copper-binding, transmembrane, phosphorylation, and ATP-binding domains, and in silico AlphaMissense scores indicate damaging effects for most novel substitutions. Post-LT follow-up showed biochemical normalization in the majority of recipients, with five deaths recorded during the study period. Conclusions: This single-center Iranian study reveals a highly heterogeneous ATP7B mutational landscape with a large proportion of novel population-specific variants and underscores the benefit of comprehensive gene sequencing for timely WD diagnosis and family counseling, particularly in regions with prevalent consanguinity. Full article
Show Figures

Figure 1

11 pages, 2044 KB  
Communication
CRISPR-Enhanced Colorimetric Aptasensor for Adenosine Triphosphate Detection Based on MoS2-Based Nanozymes
by Zhiqiang Zhu, Haojie Ma, Huashan Yao, Yuan Yuan, Xiangyang Miao and Shao Su
Biosensors 2025, 15(10), 651; https://doi.org/10.3390/bios15100651 - 1 Oct 2025
Abstract
As the direct energy source in organisms, accurate and simple detection of adenosine triphosphate (ATP) is of great significance. Herein, a colorimetric aptasensor for ATP determination was designed by integrating the CRISPR/Cas12a system with an aptamer, and with Prussian blue nanocube and gold [...] Read more.
As the direct energy source in organisms, accurate and simple detection of adenosine triphosphate (ATP) is of great significance. Herein, a colorimetric aptasensor for ATP determination was designed by integrating the CRISPR/Cas12a system with an aptamer, and with Prussian blue nanocube and gold nanoparticle co-functionalized MoS2 (MoS2-PBNCs-AuNPs) nanozymes. As expected, the introduced CRISPR/Cas12a system and aptamer could efficiently amplify the detection signal and improve the specific recognition ability, respectively. Meanwhile, the catalytic activity of the MoS2-PBNCs-AuNPs nanozymes can be regulated with the concentration of ATP. The high-affinity binding of ATP to the aptamer competitively inhibited aptamer-crRNA hybridization, causing fewer Cas12 proteins to be activated. As a result, the uncleaved single-stranded DNA (ssDNA) adsorbed onto the surface of nanozymes to effectively enhance their catalytic oxidation capability toward 3,3′,5,5′-tetramethylbenzidine (TMB). According to this phenomenon, this CRISPR-enhanced colorimetric aptasensor can detect down to 0.14 μM ATP with high selectivity, reproducibility, and stability. In addition, acceptable recoveries and low relative standard deviations of the aptasensor for ATP determination suggest that it is promising for application in early detection of clinical-related diseases. Full article
(This article belongs to the Special Issue CRISPR/Cas System-Based Biosensors)
Show Figures

Figure 1

23 pages, 9866 KB  
Article
Dysferlin Protein–Protein Interaction Pathways in the Organ of Corti and Spiral Ganglion Intersect with Alzheimer’s Protein Pathways
by Marian J. Drescher, Dennis G. Drescher, Khalid M. Khan, James S. Hatfield and Darshi Hemani
Int. J. Mol. Sci. 2025, 26(19), 9559; https://doi.org/10.3390/ijms26199559 - 30 Sep 2025
Abstract
Dysferlin direct protein–protein interactions (PPI) previously have been elucidated with surface plasmon resonance (SPR) and predicted to underlie membrane repair in mechanotransducing myofibrils. In mechanotransducing inner ear hair cells, dysferlin is detected with Z-stack confocal immunofluorescence in the stereocilia and their inserts in [...] Read more.
Dysferlin direct protein–protein interactions (PPI) previously have been elucidated with surface plasmon resonance (SPR) and predicted to underlie membrane repair in mechanotransducing myofibrils. In mechanotransducing inner ear hair cells, dysferlin is detected with Z-stack confocal immunofluorescence in the stereocilia and their inserts in the tectorial membrane (TM) co-localizing with FKBP8, consistent with the SPR determination of tight, positively Ca2+-dependent interaction. FKBP8, a direct binding partner of mechanotransducing TMC1, when overexpressed, evokes an elevation in anti-apoptotic BCL2, inhibition of ryanodine receptor (RYR) activity, and a consequent reduction in Ca2+ release. RYR3 has now been immunolocalized to the tip of the TM in close association with a third-row outer hair cell (OHC) stereociliary BCL2-positive insertion. Dysferlin, annexin A2, and Alzheimer’s proteins BACE1 and amyloid precursor protein (APP) are also accumulated in these stereociliary insertions. RYR2 and RYR1 have been immunolocalized to the TM core, in position to influence TM Ca2+. Dysferlin PPI pathways also intersect with AD protein pathways in the spiral ganglion (SG). Dysferlin segregates with FKBP8, BACE1, and RYR3 in the interiors of SG type I cell bodies. RYR1, RYR2, PSEN1, BCL2, and caspase 3 are primarily confined to plasma membrane sites. RYR3 pathways traverse the plasma membrane to the cell body interior. Western analysis of dysferlinopathy proteins links FKBP8 and BCL2 overexpression with RYR inhibition, indicative of dysferlin targets that are ameliorative in AD. Full article
(This article belongs to the Section Molecular Neurobiology)
Show Figures

Figure 1

29 pages, 2729 KB  
Review
Applications of Isothermal Titration Calorimetry in Studying Biomimetic Nanocarriers
by Martin Guerrero, Colby Braden and Yuping Bao
Biomolecules 2025, 15(10), 1349; https://doi.org/10.3390/biom15101349 - 23 Sep 2025
Viewed by 148
Abstract
Biomimetic nanocarriers, particularly membrane-based systems, have emerged as promising platforms for drug delivery. A thorough understanding of the molecular interactions that govern their assembly, stability, and cargo-loading efficiency is essential for optimizing their design and performance. Equally important are their interactions with biological [...] Read more.
Biomimetic nanocarriers, particularly membrane-based systems, have emerged as promising platforms for drug delivery. A thorough understanding of the molecular interactions that govern their assembly, stability, and cargo-loading efficiency is essential for optimizing their design and performance. Equally important are their interactions with biological components such as proteins, lipids, nucleotides, and cells, which significantly influence delivery efficacy. Among various techniques for characterizing these nanocarriers, isothermal titration calorimetry (ITC) has proven to be an invaluable tool to study their molecular interactions. ITC enables direct quantification of key thermodynamic parameters, such as binding affinity, stoichiometry, enthalpy, and entropy changes, without the need for molecular labeling or immobilization. This review highlights the application of ITC in the study of biomimetic nanocarriers, focusing on solid lipid nanoparticles, liposomes, extracellular vesicles, cell-derived vesicles and live cells. For each type of nanocarrier, the ITC applications in specific areas and the resulting information are discussed. For example, ITC was used to characterize drug interaction and protein adsorption for solid nanoparticles. In contrast, many aspects of liposomes were explored by ITC, including membrane solubilization and stabilization, peptide interactions, and macromolecule and protein adsorption. Overall, this review aims to provide a conceptual and practical framework for employing ITC in the investigation of biomimetic nanocarrier systems, facilitating their rational design and improved therapeutic performance. Furthermore, the discussion encourages further development of strategies to increase the application in cell-derived vesicles and live cells. Full article
(This article belongs to the Special Issue Advances in Nano-Based Drug Delivery: Unveiling the Next Frontier)
Show Figures

Graphical abstract

28 pages, 4648 KB  
Article
Allosteric Control Overcomes Steric Limitations for Neutralizing Antibodies Targeting Conserved Binding Epitopes of the SARS-CoV-2 Spike Protein: Exploring the Intersection of Binding, Allostery, and Immune Escape with a Multimodal Computational Approach
by Mohammed Alshahrani, Vedant Parikh, Brandon Foley and Gennady Verkhivker
Biomolecules 2025, 15(9), 1340; https://doi.org/10.3390/biom15091340 - 18 Sep 2025
Viewed by 310
Abstract
Understanding the atomistic basis of multi-layer mechanisms employed by broadly reactive neutralizing antibodies of the SARS-CoV-2 spike protein without directly blocking receptor engagement remains an important challenge in coronavirus immunology. Class 4 antibodies represent an intriguing case: they target a deeply conserved, cryptic [...] Read more.
Understanding the atomistic basis of multi-layer mechanisms employed by broadly reactive neutralizing antibodies of the SARS-CoV-2 spike protein without directly blocking receptor engagement remains an important challenge in coronavirus immunology. Class 4 antibodies represent an intriguing case: they target a deeply conserved, cryptic epitope on the receptor-binding domain yet exhibit variable neutralization potency across subgroups F1 (CR3022, EY6A, COVA1-16), F2 (DH1047), and F3 (S2X259). The molecular basis for this variability is not fully understood. Here, we employed a multi-modal computational approach integrating atomistic and coarse-grained molecular dynamics simulations, binding free energy calculations, mutational scanning, and dynamic network analysis to elucidate how these antibodies engage the receptor-binding domain (RBD) of the SARS-CoV-2 spike protein and influence its function. Our results reveal that neutralization efficacy arises from the interplay of direct interfacial interactions and allosteric effects. Group F1 antibodies (CR3022, EY6A, COVA1-16) primarily operate via classic allostery, modulating flexibility in RBD loop regions to indirectly interfere with the ACE2 receptor binding through long-range effects. Group F2 antibody DH1047 represents an intermediate mechanism, combining partial steric hindrance—through engagement of ACE2-critical residues T376, R408, V503, and Y508—with significant allosteric influence, facilitated by localized communication pathways linking the epitope to the receptor interface. Group F3 antibody S2X259 achieves potent neutralization through a synergistic mechanism involving direct competition with ACE2 and localized allosteric stabilization, albeit with potentially increased escape vulnerability. Dynamic network analysis identified a conserved “allosteric ring” within the RBD core that serves as a structural scaffold for long-range signal propagation, with antibody-specific extensions modulating communication to the ACE2 interface. These findings support a model where Class 4 neutralization strategies evolve through the refinement of peripheral allosteric connections rather than epitope redesign. This study establishes a robust computational framework for understanding the atomistic basis of neutralization activity and immune escape for Class 4 antibodies, highlighting how the interplay of binding energetics, conformational dynamics, and allosteric modulation governs their effectiveness against SARS-CoV-2. Full article
(This article belongs to the Special Issue Protein Biophysics)
Show Figures

Graphical abstract

16 pages, 2560 KB  
Article
Brassinin Induces H2S Signals and Improves Vascular Smooth Muscle Cell Functions
by Jazmin Fergani, Xiaoli Han, Zhuping Jin, Yanxi Pei, Sabine Montaut and Guangdong Yang
Molecules 2025, 30(18), 3775; https://doi.org/10.3390/molecules30183775 - 17 Sep 2025
Viewed by 308
Abstract
Brassinin, a sulfur-containing phytoalexin, exerts anticancer and anti-inflammatory effects. Hydrogen sulfide (H2S) is an important gasotransmitter with significant cardioprotective properties. The effects of brassinin on H2S signaling and vascular smooth muscle cell (SMC) functions remain unexplored. This study found [...] Read more.
Brassinin, a sulfur-containing phytoalexin, exerts anticancer and anti-inflammatory effects. Hydrogen sulfide (H2S) is an important gasotransmitter with significant cardioprotective properties. The effects of brassinin on H2S signaling and vascular smooth muscle cell (SMC) functions remain unexplored. This study found that brassinin protected against angiotensin II (Ang II)-induced SMC dysfunctions. These effects included the attenuation of excessive cell proliferation, migration, and oxidative stress; and upregulation of smooth muscle contractile protein expressions; and down-regulation of inflammatory gene expressions. Notably, brassinin did not directly release H2S under the tested conditions; instead, it stimulated endogenous H2S synthesis in cultured SMCs by inducing the expression of cystathionine gamma-lyase (CSE), a key H2S-generating enzyme. Further mechanistic investigations revealed that brassinin may bind to the transcription factor C/EBPβ and enhance its interaction with the CSE promoter, thereby upregulating CSE transcription. In conclusion, our findings demonstrate that brassinin protects against SMC dysfunction, at least in part, by activating H2S signaling rather than acting as a direct H2S donor. These results provide new insights into the potential of brassinin as a therapeutic agent for improving vascular health and preventing cardiovascular diseases. Full article
Show Figures

Graphical abstract

56 pages, 7184 KB  
Article
Curcumin Inhibits Protease Activated Receptor 2-Induced ERK Phosphorylation Calcium Mobilization and Anti-Apoptotic Signaling in Inflammation-Driven Colorectal Cancer Cells
by Rajashree Patnaik, Riah Varghese, Ahad Al-Kabani, Shirin Jannati and Yajnavalka Banerjee
Cells 2025, 14(18), 1451; https://doi.org/10.3390/cells14181451 - 16 Sep 2025
Viewed by 390
Abstract
Background: Chronic inflammation drives colorectal cancer (CRC) progression, with PAR-2, a G-protein coupled receptor, linking extracellular inflammatory signals to tumor-promoting pathways via ERK1/2 phosphorylation, calcium mobilization, TNF-α upregulation, and apoptosis suppression. While curcumin has notable anti-inflammatory and anti-cancer properties, its effects on PAR-2 [...] Read more.
Background: Chronic inflammation drives colorectal cancer (CRC) progression, with PAR-2, a G-protein coupled receptor, linking extracellular inflammatory signals to tumor-promoting pathways via ERK1/2 phosphorylation, calcium mobilization, TNF-α upregulation, and apoptosis suppression. While curcumin has notable anti-inflammatory and anti-cancer properties, its effects on PAR-2 signaling in inflammation-driven CRC remain underexplored. Objective: This study investigates how curcumin modulates PAR-2 expression and downstream oncogenic signaling in inflammation-driven CRC cells and explores its potential direct interaction with PAR-2 at the structural level. Methods: HT 29 and Caco-2 CRC cell lines were exposed to lipopolysaccharide (LPS) to induce an inflammatory phenotype, followed by treatment with curcumin at 50 µM and 100 µM. PAR-2 and PAR-1 expression, along with downstream markers including ERK1/2, p-ERK, TNF-α, caspase-8, cleaved caspase-8, caspase-3, Bcl 2, and Bax, were analyzed by Western blot and quantitative PCR. Calcium mobilization was assessed using Fluo-4 dye-based fluorescence imaging. Apoptosis was quantified using MTT viability assays, AO/EtBr dual staining, and Annexin V/PI flow cytometry. In parallel, AlphaFold-predicted structural models of PAR-2 were used to perform molecular docking with curcumin using CB-Dock2, to identify potential binding pockets and assess binding energetics. Results: Curcumin selectively downregulated PAR-2—but not PAR-1—at both transcript and protein levels in a dose-dependent manner. This downregulation was accompanied by suppression of ERK phosphorylation and calcium signaling, inhibition of TNF-α secretion, and reversal of the anti-apoptotic signaling axis (Bcl 2 downregulation and Bax and caspase-3/-8 upregulation). Functional assays confirmed enhanced apoptosis in curcumin-treated cells. Computational docking revealed a high-affinity binding interaction between curcumin and the transmembrane domain of PAR-2, supporting the hypothesis of direct G-Protein-Coupled Receptor (GPCR) modulation. Conclusions: Our findings reveal that curcumin targets the PAR-2/ERK/TNF-α axis and reactivates apoptotic pathways in inflammation-driven CRC, establishing it as a potent, mechanistically validated candidate for therapeutic repurposing in CRC. Full article
(This article belongs to the Collection Molecular and Cellular Mechanisms of Cancers: Colorectal Cancer)
Show Figures

Figure 1

26 pages, 4801 KB  
Article
Structural and Dynamic Insights into Acyl Carrier Protein upon Metal Binding and Acylation Revealed by NMR Spectroscopy and MD Simulations
by Chae Yeong Lee, Sungchan Jang, Hyunjoon Cho, Min-Cheol Jeong, Yoojin Oh and Yangmee Kim
Int. J. Mol. Sci. 2025, 26(18), 9005; https://doi.org/10.3390/ijms26189005 - 16 Sep 2025
Viewed by 273
Abstract
Protein dynamics are crucial for the acyl carrier protein (ACP) acting as a cofactor, communicating with various fatty acid synthesis (FAS) enzymes. Using a combination of NMR spectroscopy and molecular dynamics (MD) simulations, we demonstrate how the conformational flexibility of Escherichia coli ACP [...] Read more.
Protein dynamics are crucial for the acyl carrier protein (ACP) acting as a cofactor, communicating with various fatty acid synthesis (FAS) enzymes. Using a combination of NMR spectroscopy and molecular dynamics (MD) simulations, we demonstrate how the conformational flexibility of Escherichia coli ACP (EcACP) modulates metal binding and facilitates its molecular switches, thereby determining the pathway for different acyl chains. Our results show that Ca2+ binding greatly stabilizes the protein—boosting thermal stability by over 13 °C—and modulates its dynamic properties, affecting two acidic metal binding sites and the conformation of the hydrophobic cavity. Hydrogen–deuterium exchange and chemical denaturation experiments revealed that Ile11 and Ile72 are the key residues for the global folding of EcACP, stabilizing hydrophobic cavity. Backbone dynamics and MD simulation results indicate that longer acyl chains induce conformational adjustments, increasing flexibility in α3-helix and hydrophobic motifs, including Phe28 and Ile54. Furthermore, our findings highlight the conformational plasticity of EcACP, with key molecular switches, Leu42 and Leu46, adapting to accommodate various acyl chains and directing their pathway. These insights deepen our understanding of ACP flexibility and its functional role in FAS, offering a new strategy for designing inhibitors that target the dynamic nature of bacterial FAS pathways. Full article
(This article belongs to the Special Issue Advanced Research on Protein Structure and Protein Dynamics)
Show Figures

Graphical abstract

17 pages, 299 KB  
Review
Indications and Mechanisms of Action of the Main Treatment Modalities for Non-Melanoma Skin Cancer
by Marcio F. Chedid, Aline C. Tregnago, Floriano Riva, Lucas Prediger, Anisha Agarwal and Jane Mattei
Life 2025, 15(9), 1447; https://doi.org/10.3390/life15091447 - 16 Sep 2025
Viewed by 423
Abstract
Skin cancer is the most common cancer worldwide. The incidence of skin cancer has been increasing worldwide. Nearly 75% of all skin cancers are basal cell carcinomas (BCC), cutaneous squamous cell carcinoma (cSCC) represents approximately 20%, and those remaining are melanomas (4%) or [...] Read more.
Skin cancer is the most common cancer worldwide. The incidence of skin cancer has been increasing worldwide. Nearly 75% of all skin cancers are basal cell carcinomas (BCC), cutaneous squamous cell carcinoma (cSCC) represents approximately 20%, and those remaining are melanomas (4%) or other rare tumors (1%). Given the high cure rates and the ability to histologically confirm tumor clearance, surgical therapy is the gold standard for the treatment of skin cancer. Conventional surgery is the most employed technique for the removal of non-melanoma skin cancer (NMSCs). Mohs Micrographic Surgery (MMS) is the most precise surgical method for the treatment of non-melanoma skin cancer, allowing for 100% margin evaluation, being the gold-standard method for surgical treatment of non-melanoma skin cancer. Whenever it is possible to obtain wide margins (4 to 6 mm), cure rates vary from 70% to 99%. Imiquimod, a synthetic imidazoquinolinone amine, is a topical immune response modifier approved by the U.S. Food and Drug Administration (FDA) for the treatment of external anogenital warts, actinic keratosis (AK), and superficial basal cell carcinoma (sBCC). The efficacy of imiquimod is primarily attributed to its ability to modulate both innate and adaptive immune responses, as well as its direct effects on cancer cells. Imiquimod exerts its immunomodulatory effects by activating Toll-like receptors 7 and 8 (TLR7/8) on various immune cells, including dendritic cells, macrophages, and natural killer (NK) cells. Upon binding to these receptors, imiquimod triggers the MyD88-dependent signaling pathway, leading to the activation of nuclear factor kappa B (NF-κB) and interferon regulatory factors (IRFs). This cascade leads to the production of pro-inflammatory cytokines, including interferon-alpha (IFN-α), tumor necrosis factor-alpha (TNF-α), interleukin-12 (IL-12), and interleukin-6 (IL-6). These cytokines enhance local inflammation, recruit additional immune cells to the tumor site, and stimulate antigen presentation, thereby promoting an anti-tumor immune response. Radiation therapy (RTh) may be employed as a primary treatment to BCC. It may also be employed as an adjuvant treatment to surgery for SCC and aggressive subtypes of BCC. RTh triggers both direct and indirect DNA damage on cancer cells and generates reactive oxygen species (ROS) within cells. ROS trigger oxidative damage to DNA, proteins, and lipids, exacerbating the cellular stress and contributing to tumor cell death. Recently, immunotherapy emerged as a revolutionary treatment for all stages of SCC. Cemiplimab is a human programmed cell death 1 (PD-1)-blocking antibody that triggers a response to over 50% of patients with locally advanced and metastatic SCC. A randomized clinical trial (RCT) published in 2022 revealed that cemiplimab was highly effective in the neoadjuvant treatment of large SCCs. The drug promoted a significant tumor size decrease, enabling organ-sparing operations and a much better cosmetic effect. A few months ago, a RCT of cemiplimab on adjuvant therapy for locally aggressive SCC was published. Interestingly, cemiplimab was administered to patients with local or regional cutaneous squamous cell carcinoma after surgical resection and postoperative radiotherapy, at high risk for recurrence owing to nodal features, revealed that cemiplimab led to much lower risks both of locoregional recurrence and distant recurrence. Full article
24 pages, 4301 KB  
Article
Investigating SMR Peptide Interactions with Breast Cancer-Associated Proteins
by Ming-Bo Huang, Purushottam B. Tiwari, Aykut Üren, Martin N. Shelton, Dara Brena, Jennifer Y. Wu, Mahfuz B. Khan, Michael D. Powell, Jonathan K. Stiles, Erica L. Johnson, Fengxia Yan, Lily Yang and Vincent C. Bond
Int. J. Mol. Sci. 2025, 26(18), 8848; https://doi.org/10.3390/ijms26188848 - 11 Sep 2025
Viewed by 312
Abstract
Breast cancer (BC) is a major cause of cancer-related mortality. Mortalin and Vimentin—two proteins implicated in BC progression and metastasis—have been identified as binding partners of the Secretion Modification Region (SMR) peptide from the HIV Nef protein. These interactions disrupt exosome release and [...] Read more.
Breast cancer (BC) is a major cause of cancer-related mortality. Mortalin and Vimentin—two proteins implicated in BC progression and metastasis—have been identified as binding partners of the Secretion Modification Region (SMR) peptide from the HIV Nef protein. These interactions disrupt exosome release and offer novel therapeutic strategies. This study investigates the binding interactions between the SMR peptide, Mortalin, and Vimentin using surface plasmon resonance (SPR), co-immunoprecipitation (Co-IP), and Western blot assays. We also map the SMR binding sites on Mortalin through scanning peptide mapping and then identify a similar site on the Vimentin protein. Based on these data, we propose that the SMR peptide and its analogs interact with specific amino acid sequences in Mortalin and Vimentin, thereby disrupting cellular processes essential for Epithelial–Mesenchymal Transition (EMT) and tumor progression. SPR analysis revealed that the Nef protein exhibited the highest binding affinity to Vimentin (KD = 0.75 ± 1.1 nM) and Mortalin (KD = 3.16 ± 0.03 nM). The SMRwt peptide also demonstrated direct binding to both proteins with micromolar affinities (KD = 6.63 ± 0.74 µM for Vimentin; KD = 20.73 ± 2.33 µM for Mortalin), though the binding affinity was weaker than the full Nef protein. Co-IP experiments using MDA-MB-231, MCF-7, and BT474 BC cell lines confirmed that SMRwt, but not SMRmut, co-immunoprecipitated with Mortalin. Western blot analysis validated these interactions. Further, Mortalin peptide #56, derived from the substrate-binding domain, did not bind the SMR domain or inhibit Nef function. In contrast, peptides #61 and #62 from the C-terminal domain of Mortalin bound the SMR domain and effectively inhibited Nef activity. Notably, Mortalin peptide #61 inhibited SMRwt binding to both Mortalin and Vimentin, disrupting complex formation on the SPR sensor chip. These findings suggest that specific Mortalin-derived peptides can block SMR interactions, offering a potential therapeutic mechanism. Full article
(This article belongs to the Special Issue Molecular Research and Treatment of Breast Cancer: 3rd Edition)
Show Figures

Figure 1

14 pages, 2469 KB  
Article
WUSCHEL Transcription Factor Regulates Floral Development in ‘Jizaomi’ Grapevine
by Zedong Sun, Huan Xu, Wenxuan Shi, Jialin Fu, Pengfei Wen, Jinjun Liang and Pengfei Zhang
Horticulturae 2025, 11(9), 1099; https://doi.org/10.3390/horticulturae11091099 - 11 Sep 2025
Viewed by 354
Abstract
Carpel number has been recognized as a critical factor influencing fruit size, ultimately determining yield and economic efficiency. The WUSCHEL (WUS) protein is essential for maintaining stem cell homeostasis in the floral meristem. Its expression level directly influences the size of the floral [...] Read more.
Carpel number has been recognized as a critical factor influencing fruit size, ultimately determining yield and economic efficiency. The WUSCHEL (WUS) protein is essential for maintaining stem cell homeostasis in the floral meristem. Its expression level directly influences the size of the floral meristem (FM), thereby determining the number of floral organs in Arabidopsis thaliana, Solanum lycopersicum, and Cucumis sativus. While its role remained largely unexplored in grapevine (Vitis vinifera). This study cloned the VvWUS gene from the polycarpic grape cultivar ‘Jizaomi’. Transgenic tomato lines expressing VvWUS heterologously exhibited accelerated floral transition, enhanced carpel/floral organ initiation, and had significantly higher locule numbers relative to wild type. Furthermore, direct binding of VvWUS to the VvAGAMOUS (VvAG) promoter and activation of VvAG expression were demonstrated through yeast one-hybrid (Y1H) and dual-luciferase (LUC) assays. These findings elucidated the molecular function of VvWUS in grape carpel development, providing a foundational basis for molecular breeding strategies targeting large-berry grape varieties. Full article
Show Figures

Figure 1

18 pages, 2274 KB  
Article
Isoniazid-Derived Hydrazones Featuring Piperazine/Piperidine Rings: Design, Synthesis, and Investigation of Antitubercular Activity
by Esma Özcan, Siva Krishna Vagolu, Rasoul Tamhaev, Christian Lherbet, Lionel Mourey, Tone Tønjum, Miyase Gözde Gündüz and Şengül Dilem Doğan
Biomolecules 2025, 15(9), 1305; https://doi.org/10.3390/biom15091305 - 11 Sep 2025
Viewed by 583
Abstract
Isoniazid (isonicotinic acid hydrazide, INH) is a key drug used to treat tuberculosis (TB), which continues to be the world’s most lethal infectious disease. Nevertheless, the efficacy of INH has diminished because of the emergence of Mycobacterium tuberculosis (Mtb) strains that [...] Read more.
Isoniazid (isonicotinic acid hydrazide, INH) is a key drug used to treat tuberculosis (TB), which continues to be the world’s most lethal infectious disease. Nevertheless, the efficacy of INH has diminished because of the emergence of Mycobacterium tuberculosis (Mtb) strains that are resistant to INH. Our goal in this study was to modify INH to reduce this significant resistance chemically. We synthesized INH-based hydrazones (IP1IP13) through the reaction of INH with in-house obtained benzaldehydes carrying a piperidine or piperazine ring in refluxing ethanol. Upon confirmation of their proposed structures by various spectral techniques, IP1IP13 were evaluated for their antimycobacterial capacity against Mtb H37Rv strain and INH-resistant clinical isolates with katG and inhA mutations using the Microplate Alamar Blue Assay (MABA). The compounds were additionally tested for their cytotoxicity. The obtained data indicated that the compounds with moderately increased lipophilicity compared to INH (IP7IP13) were promising antitubercular drug candidates, exhibiting drug-like properties and negligible cytotoxicity. Out of these, IP11 (N′-(4-(4-cyclohexylpiperazin-1-yl)benzylidene)isonicotinohydrazide) emerged as the most promising derivative, demonstrating the lowest MIC values against all Mtb strains tested. Subsequently, the target molecules were evaluated for their capacity to inhibit enoyl acyl carrier protein reductase (InhA), the main target enzyme of INH. Except for IP11 demonstrating 81% InhA inhibition at a concentration of 50 μM, direct InhA inhibition was shown not to be the primary mechanism responsible for the antitubercular activity of the compounds. The binding mechanism of IP11 to InhA was analyzed through molecular docking and molecular dynamics simulations. Altogether, our research identified a novel approach to modify INH to address the challenges posed by the rising prevalence of drug-resistant Mtb strains. Full article
Show Figures

Graphical abstract

17 pages, 1063 KB  
Review
Dual Role of Transformer 2 Beta as Both a Developmental Necessity and a Disease Modulator
by Evan Swarup and Hongyu Qiu
Int. J. Mol. Sci. 2025, 26(18), 8805; https://doi.org/10.3390/ijms26188805 - 10 Sep 2025
Viewed by 264
Abstract
Transformer 2 beta (TRA2β) is a critical RNA-binding protein that regulates gene alternative splicing and is involved in cell cycle progression, neuronal differentiation, and cytoskeletal organization. It plays an essential role in embryonic development, particularly neurogenesis, where its deletion leads to severe cortical [...] Read more.
Transformer 2 beta (TRA2β) is a critical RNA-binding protein that regulates gene alternative splicing and is involved in cell cycle progression, neuronal differentiation, and cytoskeletal organization. It plays an essential role in embryonic development, particularly neurogenesis, where its deletion leads to severe cortical malformations and perinatal lethality. Dysregulation of TRA2β has been implicated in a range of diseases, including neurological, oncological, and immune-related disorders. Given its broad influence, TRA2β is a compelling candidate for targeted therapies and diagnostic biomarkers. This review highlights recent advances in our understanding of TRA2β regulation and its role in modulating alternative splicing across diverse cell types. It emphasizes TRA2β’s dual function as both a developmental regulator and a disease modulator and explores emerging insights into its therapeutic potential and future research directions. A deeper understanding of the cell-specific regulation of TRA2β may accelerate the development of innovative therapeutic strategies targeting this versatile protein. Full article
(This article belongs to the Section Molecular Pathology, Diagnostics, and Therapeutics)
Show Figures

Figure 1

Back to TopTop