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

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
remove_circle_outline

Search Results (695)

Search Parameters:
Keywords = canonical activation pathway

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
25 pages, 3642 KiB  
Article
A Novel Steroidogenic Action of Anti-Müllerian Hormone in Teleosts: Evidence from the European Sea Bass Male (Dicentrarchus labrax)
by Alessia Mascoli, Cinta Zapater, Soledad Ibañez, Mateus Contar Adolfi, Manfred Schartl and Ana Gómez
Int. J. Mol. Sci. 2025, 26(15), 7554; https://doi.org/10.3390/ijms26157554 (registering DOI) - 5 Aug 2025
Abstract
The Anti-Müllerian hormone (AMH) is widely recognized for promoting Müllerian duct regression in higher vertebrates and regulating key reproductive functions like steroidogenesis, folliculogenesis, and Leydig cell development. In teleost fish, which lack Müllerian ducts, Amh primarily influences male reproductive functions, including sex determination, [...] Read more.
The Anti-Müllerian hormone (AMH) is widely recognized for promoting Müllerian duct regression in higher vertebrates and regulating key reproductive functions like steroidogenesis, folliculogenesis, and Leydig cell development. In teleost fish, which lack Müllerian ducts, Amh primarily influences male reproductive functions, including sex determination, testis differentiation, and germ cell proliferation. In adult fish, Amh supports gonad development and spermatogenesis, but its role in teleost gonadal physiology remains largely underexplored. This study reveals a novel steroidogenic function in the European sea bass (Dicentrarchus labrax) using in vitro testis culture, in vivo plasmid injection, and cell-based transactivation assays. The Amh-induced significant increase in androgen levels was also confirmed in Japanese medaka (Oryzias latipes) treated with recombinant sea bass Amh. Beyond activating the canonical Smad pathway, Amh also triggered the cAMP/PKA signalling pathway via its cognate type II receptor, Amhr2. Inhibitors of these pathways independently and synergistically counteracted Amh-induced CRE-Luc activity, indicating pathway crosstalk. Moreover, inhibition of the cAMP pathway suppressed Amh-induced androgen production in testis cultures, emphasizing the crucial role of protein kinase A in mediating Amh steroidogenic action. These findings uncover a novel steroidogenic function of Amh in teleosts and highlight its broader role in male reproductive physiology. Full article
(This article belongs to the Special Issue Molecular Research in Animal Reproduction)
Show Figures

Figure 1

13 pages, 1809 KiB  
Perspective
Specific Low/Endogenous Replication Stress Response Protects Genomic Stability via Controlled ROS Production in an Adaptive Way and Is Dysregulated in Transformed Cells
by Bernard S. Lopez
Cells 2025, 14(15), 1183; https://doi.org/10.3390/cells14151183 - 31 Jul 2025
Viewed by 168
Abstract
Cells are assaulted daily by stresses that jeopardize genome integrity. Primary human cells adapt their response to the intensity of replication stress (RS) in a diphasic manner: below a stress threshold, the canonical DNA damage response (cDDR) is not activated, but a noncanonical [...] Read more.
Cells are assaulted daily by stresses that jeopardize genome integrity. Primary human cells adapt their response to the intensity of replication stress (RS) in a diphasic manner: below a stress threshold, the canonical DNA damage response (cDDR) is not activated, but a noncanonical cellular response, low-level stress-DDR (LoL-DDR), has recently been described. LoL-DDR prevents the accumulation of premutagenic oxidized bases (8-oxoguanine) through the production of ROS in an adaptive way. The production of RS-induced ROS (RIR) is tightly controlled: RIR are excluded from the nucleus and are produced by the NADPH oxidases DUOX1/DUOX2, which are controlled by NF-κB and PARP1; then, RIR activate the FOXO1-detoxifying pathway. Increasing the intensity of RS suppresses RIR via p53 and ATM. Notably, LoL-DDR is dysregulated in cancer cell lines, in which RIR are not produced by NADPH oxidases, are not detoxified under high-level stress, and favor the accumulation of 8-oxoguanine. LoL-DDR dysregulation occurred at an early stage of cancer progression in an in vitro model. Since, conversely, ROS trigger RS, this establishes a vicious cycle that continuously jeopardizes genome integrity, fueling tumorigenesis. These data reveal a novel type of ROS-controlled DNA damage response and demonstrate the fine-tuning of the cellular response to stress. The effects on genomic stability and carcinogenesis are discussed here. Full article
Show Figures

Figure 1

13 pages, 2596 KiB  
Article
Bark Extracts of Chamaecyparis obtusa (Siebold & Zucc.) Endl. Attenuate LPS-Induced Inflammatory Responses in RAW264.7 Macrophages
by Bo-Ae Kim, Ji-A Byeon, Young-Ah Jang and Yong-Jin Kwon
Plants 2025, 14(15), 2346; https://doi.org/10.3390/plants14152346 - 29 Jul 2025
Viewed by 295
Abstract
Chamaecyparis obtusa (Siebold & Zucc.) Endl. (C. obtusa) is an evergreen conifer native to temperate regions such as South Korea and Japan, traditionally used for its anti-inflammatory properties. However, the molecular mechanisms underlying the anti-inflammatory effects of C. obtusa bark extracts [...] Read more.
Chamaecyparis obtusa (Siebold & Zucc.) Endl. (C. obtusa) is an evergreen conifer native to temperate regions such as South Korea and Japan, traditionally used for its anti-inflammatory properties. However, the molecular mechanisms underlying the anti-inflammatory effects of C. obtusa bark extracts remain poorly understood. In this study, I compared the biological activities of C. obtusa bark extracts prepared using boiling water (COWB) and 70% ethanol (COEB), and investigated their anti-inflammatory mechanisms in lipopolysaccharide (LPS)-stimulated RAW264.7 macrophages. COEB significantly suppressed both mRNA and protein expression of inducible nitric oxide synthase (iNOS) and cyclooxygenase-2 (COX-2), along with decreased production of their respective inflammatory mediators, nitric oxide (NO) and prostaglandin E2 (PGE2). Additionally, COEB selectively downregulated interleukin (IL)-1β expression, without affecting tumor necrosis factor-α (TNF-α), and unexpectedly upregulated IL-6. Notably, COEB did not inhibit the LPS-induced activation of major inflammatory signaling pathways, including mitogen-activated protein kinase (MAPK), nuclear factor-kappa B (NF-κB), and Janus kinase/signal transducer and activator of transcription (JAK/STAT). These findings suggest that COEB exerts anti-inflammatory effects by modulating key inflammatory mediators independently of canonical signaling pathways and may offer a novel therapeutic strategy for controlling inflammation. Full article
(This article belongs to the Section Phytochemistry)
Show Figures

Figure 1

23 pages, 1789 KiB  
Review
Multi-Enzyme Synergy and Allosteric Regulation in the Shikimate Pathway: Biocatalytic Platforms for Industrial Applications
by Sara Khan and David D. Boehr
Catalysts 2025, 15(8), 718; https://doi.org/10.3390/catal15080718 - 28 Jul 2025
Viewed by 392
Abstract
The shikimate pathway is the fundamental metabolic route for aromatic amino acid biosynthesis in bacteria, plants, and fungi, but is absent in mammals. This review explores how multi-enzyme synergy and allosteric regulation coordinate metabolic flux through this pathway by focusing on three key [...] Read more.
The shikimate pathway is the fundamental metabolic route for aromatic amino acid biosynthesis in bacteria, plants, and fungi, but is absent in mammals. This review explores how multi-enzyme synergy and allosteric regulation coordinate metabolic flux through this pathway by focusing on three key enzymes: 3-deoxy-d-arabino-heptulosonate-7-phosphate synthase, chorismate mutase, and tryptophan synthase. We examine the structural diversity and distribution of these enzymes across evolutionary domains, highlighting conserved catalytic mechanisms alongside species-specific regulatory adaptations. The review covers directed evolution strategies that have transformed naturally regulated enzymes into standalone biocatalysts with enhanced activity and expanded substrate scope, enabling synthesis of non-canonical amino acids and complex organic molecules. Industrial applications demonstrate the pathway’s potential for sustainable production of pharmaceuticals, polymer precursors, and specialty chemicals through engineered microbial platforms. Additionally, we discuss the therapeutic potential of inhibitors targeting pathogenic organisms, particularly their mechanisms of action and antimicrobial efficacy. This comprehensive review establishes the shikimate pathway as a paradigmatic system where understanding allosteric networks enables the rational design of biocatalytic platforms, providing blueprints for biotechnological innovation and demonstrating how evolutionary constraints can be overcome through protein engineering to create superior industrial biocatalysts. Full article
Show Figures

Graphical abstract

36 pages, 5612 KiB  
Review
The Multifaceted Role of p53 in Cancer Molecular Biology: Insights for Precision Diagnosis and Therapeutic Breakthroughs
by Bolong Xu, Ayitila Maimaitijiang, Dawuti Nuerbiyamu, Zhengding Su and Wenfang Li
Biomolecules 2025, 15(8), 1088; https://doi.org/10.3390/biom15081088 - 27 Jul 2025
Viewed by 488
Abstract
The protein p53, often referred to as the “guardian of the genome,” is essential for preserving cellular balance and preventing cancerous transformations. As one of the most commonly altered genes in human cancers, its impaired function is associated with tumor initiation, development, and [...] Read more.
The protein p53, often referred to as the “guardian of the genome,” is essential for preserving cellular balance and preventing cancerous transformations. As one of the most commonly altered genes in human cancers, its impaired function is associated with tumor initiation, development, and resistance to treatment. Exploring the diverse roles of p53, which include regulating the cell cycle, repairing DNA, inducing apoptosis, reprogramming metabolism, and modulating immunity, provides valuable insights into cancer mechanisms and potential treatments. This review integrates recent findings on p53′s dual nature, functioning as both a tumor suppressor and an oncogenic promoter, depending on the context. Wild-type p53 suppresses tumors by inducing cell cycle arrest or apoptosis in response to genotoxic stress, while mutated variants often lose these functions or gain novel pro-oncogenic activities. Emerging evidence highlights p53′s involvement in non-canonical pathways, such as regulating tumor microenvironment interactions, metabolic flexibility, and immune evasion mechanisms. For instance, p53 modulates immune checkpoint expression and influences the efficacy of immunotherapies, including PD-1/PD-L1 blockade. Furthermore, advancements in precision diagnostics, such as liquid biopsy-based detection of p53 mutations and AI-driven bioinformatics tools, enable early cancer identification and stratification of patients likely to benefit from targeted therapies. Therapeutic strategies targeting p53 pathways are rapidly evolving. Small molecules restoring wild-type p53 activity or disrupting mutant p53 interactions, such as APR-246 and MDM2 inhibitors, show promise in clinical trials. Combination approaches integrating gene editing with synthetic lethal strategies aim to exploit p53-dependent vulnerabilities. Additionally, leveraging p53′s immunomodulatory effects through vaccine development or adjuvants may enhance immunotherapy responses. In conclusion, deciphering p53′s complex biology underscores its unparalleled potential as a biomarker and therapeutic target. Integrating multi-omics analyses, functional genomic screens, and real-world clinical data will accelerate the translation of p53-focused research into precision oncology breakthroughs, ultimately improving patient outcomes. Full article
(This article belongs to the Special Issue DNA Damage and Repair in Cancer Treatment)
Show Figures

Figure 1

15 pages, 1961 KiB  
Article
Age-Dependent Immune Defense Against Beauveria bassiana in Long- and Short-Lived Drosophila Populations
by Elnaz Bagheri, Han Yin, Arnie Lynn C. Bengo, Kshama Ekanath Rai, Taryn Conyers, Robert Courville, Mansour Abdoli, Molly K. Burke and Parvin Shahrestani
J. Fungi 2025, 11(8), 556; https://doi.org/10.3390/jof11080556 - 27 Jul 2025
Viewed by 326
Abstract
Aging in sexually reproducing organisms is shaped by the declining force of natural selection after reproduction begins. In Drosophila melanogaster, experimental evolution shows that altering the age of reproduction shifts the timing of aging. Using the Drosophila experimental evolution population (DEEP) resource, [...] Read more.
Aging in sexually reproducing organisms is shaped by the declining force of natural selection after reproduction begins. In Drosophila melanogaster, experimental evolution shows that altering the age of reproduction shifts the timing of aging. Using the Drosophila experimental evolution population (DEEP) resource, which includes long- and short- lived populations evolved under distinct reproductive schedules, we investigated how immune defense against Beauveria bassiana changes with age and evolved lifespan. We tested survival post-infection at multiple ages and examined genomic differentiation for immune-related genes. Both population types showed age-related declines in immune defense. Long-lived populations consistently exhibited age-specific defense when both long- and short-lived populations were tested. Genomic comparisons revealed thousands of differentiated loci, yet no enrichment for canonical immune genes or overlap with gene sets from studies of direct selection for immunity. These results suggest that enhanced immune defense can evolve alongside extended lifespan, likely via general physiological robustness rather than traditional immune pathways. A more detailed analysis may reveal that selection for lifespan favors tolerance-based mechanisms that reduce infection damage without triggering immune activation, in contrast to direct selection for resistance. Our findings demonstrate the utility of experimentally evolved populations for dissecting the genetic architecture of aging and immune defense to inform strategies to mitigate age-related costs associated with immune activation. Full article
(This article belongs to the Special Issue Advances in Research on Entomopathogenic Fungi)
Show Figures

Figure 1

11 pages, 1161 KiB  
Commentary
The Role of Nuclear Phosphoinositides in the p53-MDM2 Nexus
by Jeong Hyo Lee, Muhammad Khalil Salah, Xiangqin Chen, Nickolas Vladimir Kucherenko, Vincent L. Cryns and Richard A. Anderson
Cells 2025, 14(15), 1126; https://doi.org/10.3390/cells14151126 - 22 Jul 2025
Viewed by 333
Abstract
Recent insights into the p53-MDM2 nexus have advanced deeper understanding of their regulation and potent impact on cancer heterogeneity. The roles of nuclear phosphoinositide (PIPns) in modulating this pathway are emerging as a key mechanism. Here, we dissect the molecular mechanisms [...] Read more.
Recent insights into the p53-MDM2 nexus have advanced deeper understanding of their regulation and potent impact on cancer heterogeneity. The roles of nuclear phosphoinositide (PIPns) in modulating this pathway are emerging as a key mechanism. Here, we dissect the molecular mechanisms by which nuclear PIPns stabilize p53 through the recruitment of small heat shock proteins (sHSPs), activate the nuclear phosphatidylinositol 3-kinase (PI3K)-AKT signaling cascade, and modulate MDM2 function to regulate the p53-MDM2 interaction. We propose potential mechanisms by which nuclear PIPns coordinate signaling with nuclear p53, AKT, and MDM2. Ultimately, we highlight that nuclear PIPns serve as a ‘third messenger’ within the p53-MDM2 axis, expanding the current framework of non-canonical nuclear signaling in cancer biology. Full article
(This article belongs to the Section Cell Microenvironment)
Show Figures

Figure 1

28 pages, 944 KiB  
Review
Amphiregulin in Fibrotic Diseases and Cancer
by Tae Rim Kim, Beomseok Son, Chun Geun Lee and Han-Oh Park
Int. J. Mol. Sci. 2025, 26(14), 6945; https://doi.org/10.3390/ijms26146945 - 19 Jul 2025
Viewed by 417
Abstract
Fibrotic disorders pose a significant global health burden due to limited treatment options, creating an urgent need for novel therapeutic strategies. Amphiregulin (AREG), a low-affinity ligand for the epidermal growth factor receptor (EGFR), has emerged as a key mediator of fibrogenesis through dual [...] Read more.
Fibrotic disorders pose a significant global health burden due to limited treatment options, creating an urgent need for novel therapeutic strategies. Amphiregulin (AREG), a low-affinity ligand for the epidermal growth factor receptor (EGFR), has emerged as a key mediator of fibrogenesis through dual signaling pathways. Unlike high-affinity EGFR ligands, AREG induces sustained signaling that activates downstream effectors and promotes the integrin-mediated activation of transforming growth factor (TGF)-β. This enables both canonical and non-canonical EGFR signaling pathways that contribute to fibrosis. Elevated AREG expression correlates with disease severity across multiple organs, including the lungs, kidneys, liver, and heart. The therapeutic targeting of AREG has shown promising antifibrotic and anticancer effects, suggesting a dual-benefit strategy. The increasing recognition of the shared mechanisms between fibrosis and cancer further supports the development of unified treatment approaches. The inhibition of AREG has been shown to sensitize fibrotic tumor microenvironments to chemotherapy, enhancing combination therapy efficacy. Targeted therapies, such as Self-Assembled-Micelle inhibitory RNA (SAMiRNA)-AREG, have demonstrated enhanced specificity and favorable safety profiles in preclinical studies and early clinical trials. Personalized treatment based on AREG expression may improve clinical outcomes, establishing AREG as a promising precision medicine target for both fibrotic and malignant diseases. This review aims to provide a comprehensive understanding of AREG biology and evaluate its therapeutic potential in fibrosis and cancer. Full article
Show Figures

Figure 1

22 pages, 8519 KiB  
Article
RNA-Seq Analysis of MCF-7 Breast Cancer Cells Treated with Methyl Gallate Isolated from the Rhizomes of Nymphaea Odorata L. Shows Upregulation of Apoptosis, Autophagy, and Unfolded Protein Canonical Pathways
by Nishikant A. Raut, Pinal N. Kanabar, Mark Maienschein-Cline, Nina S. Los, Zarema Arbieva, Temitope O. Lawal, Shitalben Patel and Gail B. Mahady
Molecules 2025, 30(14), 3022; https://doi.org/10.3390/molecules30143022 - 18 Jul 2025
Viewed by 330
Abstract
The effects of a methanol extract of Nymphaea odorata (MeNO) rhizomes, its fractions and the active compound (methyl gallate, MeG) were investigated in estrogen receptor-positive (ER+) breast cancer cell lines MCF-7 and T47-D:A18, as well as ER-negative line SKBr3. Cell viability and cytotoxicity [...] Read more.
The effects of a methanol extract of Nymphaea odorata (MeNO) rhizomes, its fractions and the active compound (methyl gallate, MeG) were investigated in estrogen receptor-positive (ER+) breast cancer cell lines MCF-7 and T47-D:A18, as well as ER-negative line SKBr3. Cell viability and cytotoxicity were determined using CellTiter-Glo® 2.0 assays at concentrations ranging from 1 to 100 μg/mL. Caspase activity and apoptosis were determined using Caspase-Glo® 3/7, Caspase-Glo® 8, and ApoTox-Glo™ triplex assays, as well as qPCR. Total RNA was isolated from MCF-7 cells treated with MeG. RNA-seq libraries were prepared using a Universal Plus mRNASeq kit, and sequencing was performed on a NovaSeq 6000. MeNO inhibited the growth of MCF-7 cells with an IC50 of 14.1 μg/mL, as well as T47-D:A18 (IC50 of 25.6 μg/mL) and SKBr3 cells (IC50 of 35.5 μg/mL). Bioassay-guided fractionation of MeNO in MCF-7 cells identified the active fraction containing one compound, namely methyl gallate (MeG). MeG had an IC50 of 8.6 μg/mL in MCF-7 cells. Transcriptomic analysis of MeG-treated MCF-7 cells showed differential expression of 10,634 genes, with 5643 upregulated and 4991 downregulated (FDR < 0.05). Ingenuity pathway analysis revealed the involvement of 43 canonical pathways, with the top upregulated pathways including apoptosis, autophagy, and the unfolded protein response pathways. Full article
Show Figures

Graphical abstract

13 pages, 1002 KiB  
Perspective
Sensing the Stiffness: Cellular Mechano-Sensing at the Implant Interface
by Patricia S. Pardo, Delia Danila, Raja Devesh Kumar Misra and Aladin M. Boriek
Cells 2025, 14(14), 1101; https://doi.org/10.3390/cells14141101 - 17 Jul 2025
Viewed by 385
Abstract
In this perspective, we highlight the relevance of the FA-Hippo signaling pathway and its regulation of the Yes-associated protein (YAP) and the transcriptional coactivator with a PDZ-binding domain (TAZ) as main players in the process of implants integration. The modulation and responses of [...] Read more.
In this perspective, we highlight the relevance of the FA-Hippo signaling pathway and its regulation of the Yes-associated protein (YAP) and the transcriptional coactivator with a PDZ-binding domain (TAZ) as main players in the process of implants integration. The modulation and responses of YAP/TAZ triggered by substrate and ECM stiffness are of particular interest in the construction of materials used for medical implants. YAP/TAZ nuclear localization and activity respond to the substrate stiffness by several mechanisms that involve the canonical and non-canonical Hippo signaling and independently of the Hippo cascade. YAP/TAZ regulate the expression of genes involved in several mechanisms of relevance for implant integration such as the proliferation and differentiation of cell precursors and the immune response to the implant. The influence of substrate stiffness on the regulation of the immune response is not completely understood and the progress in this field can contribute to the designing of an adequate implant design. Though the use of nano-biomaterials has been proved to contribute to implant success, the relationship between grain size and stiffness of the material has not been explored in the biomedical field; filling these gaps in the knowledge of biomaterials will highly contribute to the design of biomaterials that could take advantage of the cells sensing and response to the stiffness at the implant interface. Full article
(This article belongs to the Section Cellular Biophysics)
Show Figures

Graphical abstract

23 pages, 2596 KiB  
Article
Integrated Behavioral and Proteomic Characterization of MPP+-Induced Early Neurodegeneration and Parkinsonism in Zebrafish Larvae
by Adolfo Luis Almeida Maleski, Felipe Assumpção da Cunha e Silva, Marcela Bermudez Echeverry and Carlos Alberto-Silva
Int. J. Mol. Sci. 2025, 26(14), 6762; https://doi.org/10.3390/ijms26146762 - 15 Jul 2025
Viewed by 315
Abstract
Zebrafish (Danio rerio) combine accessible behavioral phenotypes with conserved neurochemical pathways and molecular features of vertebrate brain function, positioning them as a powerful model for investigating early neurodegenerative processes and screening neuroprotective strategies. In this context, integrated behavioral and proteomic analyses [...] Read more.
Zebrafish (Danio rerio) combine accessible behavioral phenotypes with conserved neurochemical pathways and molecular features of vertebrate brain function, positioning them as a powerful model for investigating early neurodegenerative processes and screening neuroprotective strategies. In this context, integrated behavioral and proteomic analyses provide valuable insights into the initial pathophysiological events shared by conditions such as Parkinson’s disease and related disorders—including mitochondrial dysfunction, oxidative stress, and synaptic impairment—which emerge before overt neuronal loss and offer a crucial window to understand disease progression and evaluate therapeutic candidates prior to irreversible damage. To investigate this early window of dysfunction, zebrafish larvae were exposed to 500 μM 1-methyl-4-phenylpyridinium (MPP+) from 1 to 5 days post-fertilization and evaluated through integrated behavioral and label-free proteomic analyses. MPP+-treated larvae exhibited hypokinesia, characterized by significantly reduced total distance traveled, fewer movement bursts, prolonged immobility, and a near-complete absence of light-evoked responses—mirroring features of early Parkinsonian-like motor dysfunction. Label-free proteomic profiling revealed 40 differentially expressed proteins related to mitochondrial metabolism, redox regulation, proteasomal activity, and synaptic organization. Enrichment analysis indicated broad molecular alterations, including pathways such as mitochondrial translation and vesicle-mediated transport. A focused subset of Parkinsonism-related proteins—such as DJ-1 (PARK7), succinate dehydrogenase (SDHA), and multiple 26S proteasome subunits—exhibited coordinated dysregulation, as visualized through protein–protein interaction mapping. The upregulation of proteasome components and antioxidant proteins suggests an early-stage stress response, while the downregulation of mitochondrial enzymes and synaptic regulators reflects canonical PD-related neurodegeneration. Together, these findings provide a comprehensive functional and molecular characterization of MPP+-induced neurotoxicity in zebrafish larvae, supporting its use as a relevant in vivo system to investigate early-stage Parkinson’s disease mechanisms and shared neurodegenerative pathways, as well as for screening candidate therapeutics in a developmentally responsive context. Full article
(This article belongs to the Special Issue Zebrafish Model for Neurological Research)
Show Figures

Graphical abstract

20 pages, 2891 KiB  
Review
MAPK, PI3K/Akt Pathways, and GSK-3β Activity in Severe Acute Heart Failure in Intensive Care Patients: An Updated Review
by Massimo Meco, Enrico Giustiniano, Fulvio Nisi, Pierluigi Zulli and Emiliano Agosteo
J. Cardiovasc. Dev. Dis. 2025, 12(7), 266; https://doi.org/10.3390/jcdd12070266 - 10 Jul 2025
Viewed by 634
Abstract
Acute heart failure (AHF) is a clinical syndrome characterized by the sudden onset or rapid worsening of heart failure signs and symptoms, frequently triggered by myocardial ischemia, pressure overload, or cardiotoxic injury. A central component of its pathophysiology is the activation of intracellular [...] Read more.
Acute heart failure (AHF) is a clinical syndrome characterized by the sudden onset or rapid worsening of heart failure signs and symptoms, frequently triggered by myocardial ischemia, pressure overload, or cardiotoxic injury. A central component of its pathophysiology is the activation of intracellular signal transduction cascades that translate extracellular stress into cellular responses. Among these, the mitogen-activated protein kinase (MAPK) pathways have received considerable attention due to their roles in mediating inflammation, apoptosis, hypertrophy, and adverse cardiac remodeling. The canonical MAPK cascades—including extracellular signal-regulated kinases (ERK1/2), p38 MAPK, and c-Jun N-terminal kinases (JNK)—are activated by upstream stimuli such as angiotensin II (Ang II), aldosterone, endothelin-1 (ET-1), and sustained catecholamine release. Additionally, emerging evidence highlights the role of receptor-mediated signaling, cellular stress, and myeloid cell-driven coagulation events in linking MAPK activation to fibrotic remodeling following myocardial infarction. The phosphatidylinositol 3-kinase (PI3K)/Akt signaling cascade plays a central role in regulating cardiomyocyte survival, hypertrophy, energy metabolism, and inflammation. Activation of the PI3K/Akt pathway has been shown to confer cardioprotective effects by enhancing anti-apoptotic and pro-survival signaling; however, aberrant or sustained activation may contribute to maladaptive remodeling and progressive cardiac dysfunction. In the context of AHF, understanding the dual role of this pathway is crucial, as it functions both as a marker of compensatory adaptation and as a potential therapeutic target. Recent reviews and preclinical studies have linked PI3K/Akt activation with reduced myocardial apoptosis and attenuation of pro-inflammatory cascades that exacerbate heart failure. Among the multiple signaling pathways involved, glycogen synthase kinase-3β (GSK-3β) has emerged as a key regulator of apoptosis, inflammation, metabolic homeostasis, and cardiac remodeling. Recent studies underscore its dual function as both a negative regulator of pathological hypertrophy and a modulator of cell survival, making it a compelling therapeutic candidate in acute cardiac settings. While earlier investigations focused primarily on chronic heart failure and long-term remodeling, growing evidence now supports a critical role for GSK-3β dysregulation in acute myocardial stress and injury. This comprehensive review discusses recent advances in our understanding of the MAPK signaling pathway, the PI3K/Akt cascade, and GSK-3β activity in AHF, with a particular emphasis on mechanistic insights, preclinical models, and emerging therapeutic targets. Full article
(This article belongs to the Topic Molecular and Cellular Mechanisms of Heart Disease)
Show Figures

Figure 1

26 pages, 3644 KiB  
Article
Temporal Shifts in Hormone Signaling Networks Orchestrate Soybean Floral Development Under Field Conditions: An RNA-Seq Study
by Eszter Virág, Géza Hegedűs, Ágnes Nagy, József Péter Pallos and Barbara Kutasy
Int. J. Mol. Sci. 2025, 26(13), 6455; https://doi.org/10.3390/ijms26136455 - 4 Jul 2025
Viewed by 341
Abstract
Floral ontogeny in soybean (Glycine max) is governed by multilayered regulatory hierarchies that integrate phytohormonal cues with precisely choreographed gene-expression programs. Yet, the transcriptomic architecture underpinning this continuum remains only partially resolved. Here, we generated a strand-specific, high-depth temporal transcriptome atlas [...] Read more.
Floral ontogeny in soybean (Glycine max) is governed by multilayered regulatory hierarchies that integrate phytohormonal cues with precisely choreographed gene-expression programs. Yet, the transcriptomic architecture underpinning this continuum remains only partially resolved. Here, we generated a strand-specific, high-depth temporal transcriptome atlas of soybean inflorescences spanning four morphologically defined stadiums (Stadium 0–Stadium 3). We detected transcriptional activity for 60,889 loci; pairwise stadium contrasts revealed 4000–7000 differentially expressed genes, with the most extensive reprogramming coinciding with the onset of anthesis (Stadium 2). Unsupervised clustering delineated ~600 genes peaking at the pre-anthesis phase (Stadium 1), a cohort enriched for transcriptional regulators and floral organ-identity determinants. Stadium-resolved gene-set enrichment and KEGG mapping uncovered dynamic modulation of canonical hormone-signaling pathways—including auxin, cytokinin, gibberellin, abscisic acid, ethylene, jasmonate, and salicylate circuits—reflecting shifting developmental priorities. Forty-five MADS-box transcription factor genes were expressed; notably, JOINTLESS was strongly induced at anthesis, while the root-predominant factor GmNMH7 exhibited unexpected floral expression, implicating a hitherto unappreciated role in reproductive development. Quantitative RT-PCR of representative loci corroborated RNA-seq measurements. This high-resolution atlas refines our understanding of the hormonal and genetic circuitry of soybean floral morphogenesis, furnishing molecular targets for engineering flowering time and inflorescence architecture under fluctuating environmental conditions. Full article
Show Figures

Figure 1

22 pages, 3029 KiB  
Article
Epigenetic Remodeling of Regulatory Regions by Indicaxanthin Suggests a Shift in Cell Identity Programs in Colorectal Cancer Cells
by Maria Antonietta Ragusa, Carla Gentile, Aldo Nicosia, Salvatore Costa, Sara Volpes, Laura Greco, Flores Naselli and Fabio Caradonna
Int. J. Mol. Sci. 2025, 26(13), 6072; https://doi.org/10.3390/ijms26136072 - 24 Jun 2025
Viewed by 368
Abstract
Aberrant DNA methylation is a hallmark of colorectal cancer (CRC), contributing to tumor progression through the silencing of tumor suppressor genes and activation of oncogenes. Indicaxanthin (IND), a dietary betalain pigment from Opuntia ficus indica, has shown antiproliferative effects in CRC models, [...] Read more.
Aberrant DNA methylation is a hallmark of colorectal cancer (CRC), contributing to tumor progression through the silencing of tumor suppressor genes and activation of oncogenes. Indicaxanthin (IND), a dietary betalain pigment from Opuntia ficus indica, has shown antiproliferative effects in CRC models, yet its epigenetic impact remains unexplored. In this study, we investigated the effects of IND on the methylome of Caco-2 cells using Reduced Representation Bisulfite Sequencing (RRBS). IND induced a global hypermethylation profile, particularly at gene promoters and CpG islands. Among the differentially methylated genes, 60% were protein-coding, and 10% encoded transcription factors, including PAX5 and TFAP4, both hypermethylated at active enhancers. Functional enrichment analysis revealed pathways beyond canonical intestinal functions, suggesting altered cell identity and plasticity. Transcription factor targets (SOX10, NFKB1, AHR, ARNT) were significantly enriched among the affected genes, several of which are involved in transdifferentiation processes. Methylation changes also indicated potential reprogramming toward epithelial cell types from pulmonary or neuroectodermal origin. Moreover, IND induced selective hypomethylation of Alu elements on chromosome 21 and hypermethylation of rDNA loci, hinting at suppressed ribosomal biogenesis. Overall, these findings highlight the epigenetic remodeling potential of IND and its possible role in modulating cell fate and metabolism in CRC cells. Full article
(This article belongs to the Special Issue Fundamental and Translational Insights into Colorectal Cancer)
Show Figures

Figure 1

17 pages, 3818 KiB  
Article
Multi-Target Protective Effects of β-Caryophyllene (BCP) at the Intersection of Neuroinflammation and Neurodegeneration
by Caterina Ricardi, Anna Mazzierli, Stefano Guglielmo, Nicola Origlia, Francesca Gado, Clementina Manera, Grazia Chiellini and Beatrice Polini
Int. J. Mol. Sci. 2025, 26(13), 6027; https://doi.org/10.3390/ijms26136027 - 23 Jun 2025
Viewed by 431
Abstract
Recent advances in cannabinoid-based therapies identified the natural CB2 receptor agonist β-caryophyllene (BCP) as a promising anti-inflammatory and neuroprotective agent. To further explore its therapeutic potential on the management of neurodegenerative disorders, in the present study we investigated the ability of BCP to [...] Read more.
Recent advances in cannabinoid-based therapies identified the natural CB2 receptor agonist β-caryophyllene (BCP) as a promising anti-inflammatory and neuroprotective agent. To further explore its therapeutic potential on the management of neurodegenerative disorders, in the present study we investigated the ability of BCP to prevent neuroinflammation and promote neuroprotection by using both in vitro and ex vivo models of β-amyloid induced neurotoxicity. Our data showed that BCP significantly protected human microglial HMC3 cells from Aβ25-35-induced cytotoxicity, reducing the release of pro-inflammatory cytokines (TNF-α, IL-6) while enhancing IL-10 secretion. These effects were associated with a reduced activation of the NF-κB pathway, which emerged as a central mediator of BCP action. Notably, the use of CB2R- or PPARγ-selective antagonists revealed that the observed NF-κB inhibition by BCP may involve the coordinated activation of both canonical (e.g., CB2R) and non-canonical (e.g., PPARγ) receptors. Moreover, BCP restored the expression of SIRT1, PGC-1α, and BDNF, indicating the involvement of neurotrophic pathways. Clear neuroprotective properties for BCP have been highlighted in Aβ1-42-treated brain slice preparations, where BCP demonstrated the rescue of both the amyloid-dependent depression of BDNF expression and long-term synaptic potentiation (LTP) impairment. Overall, our results suggest that BCP constitutes an attractive natural molecule for the treatment of Aβ-induced neuroinflammation and synaptic dysfunction, warranting further exploration for its clinical application. Full article
(This article belongs to the Section Molecular Neurobiology)
Show Figures

Figure 1

Back to TopTop