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
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
remove_circle_outline

Search Results (2,183)

Search Parameters:
Keywords = fibrosis pathway

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
15 pages, 1079 KB  
Review
P2Y2 Receptor Signaling in Health and Disease
by Fatemeh Salarpour and Jean Sévigny
Int. J. Mol. Sci. 2025, 26(19), 9815; https://doi.org/10.3390/ijms26199815 (registering DOI) - 9 Oct 2025
Abstract
P2Y2 receptors are a subclass of G protein-coupled receptors activated by the extracellular nucleotides ATP and UTP. These receptors are widely expressed in multiple tissues—including the brain, lungs, heart, and kidneys—and play pivotal roles in inflammation, wound healing, and cell migration. Through [...] Read more.
P2Y2 receptors are a subclass of G protein-coupled receptors activated by the extracellular nucleotides ATP and UTP. These receptors are widely expressed in multiple tissues—including the brain, lungs, heart, and kidneys—and play pivotal roles in inflammation, wound healing, and cell migration. Through coupling with various G proteins, P2Y2 receptors initiate diverse intracellular signaling pathways that mediate calcium mobilization, cytokine release, and cytoskeletal reorganization. Recent studies highlight their dual roles in health and disease. In physiological contexts, P2Y2 receptors contribute to immune modulation and tissue repair. In pathological conditions, they are implicated in Alzheimer’s disease by promoting non-amyloidogenic processing of amyloid precursor protein and in dry eye disease by enhancing mucin secretion while modulating ocular inflammation. They also influence chloride secretion and mucosal hydration in cystic fibrosis and contribute to inflammatory regulation and epithelial repair in inflammatory bowel disease. Additionally, P2Y2 receptors modulate breast cancer progression by regulating cell adhesion, migration, and matrix remodeling. Their involvement in blood pressure regulation via epithelial sodium channel modulation and their facilitative role in HIV-1 entry further underscore their clinical significance. These multifaceted functions position P2Y2 receptors as promising therapeutic targets for diverse diseases, warranting further investigation for translational applications. Full article
Show Figures

Figure 1

22 pages, 2533 KB  
Article
DST-3, a Novel Modified Cryptotanshinone, Protects Against Pulmonary Fibrosis via Inhibiting STAT3/Smad Signaling Pathway and Improves Bioavailability
by Ruoqing Guan, Xiangjun He, Yuxing Dai, Guangye Huang, Zhaoyun Xue, Jianwen Chen and Peiqing Liu
Pharmaceutics 2025, 17(10), 1307; https://doi.org/10.3390/pharmaceutics17101307 - 8 Oct 2025
Viewed by 26
Abstract
Background/Objectives: Idiopathic pulmonary fibrosis (IPF) is a fatal lung disease characterized by progressive loss of lung function and poor prognosis. Cryptotanshinone (CTS), a small-molecule compound extracted from Salvia miltiorrhiza, possesses diverse pharmacological activities but suffers from poor oral bioavailability, which restricts its [...] Read more.
Background/Objectives: Idiopathic pulmonary fibrosis (IPF) is a fatal lung disease characterized by progressive loss of lung function and poor prognosis. Cryptotanshinone (CTS), a small-molecule compound extracted from Salvia miltiorrhiza, possesses diverse pharmacological activities but suffers from poor oral bioavailability, which restricts its clinical development, particularly in pulmonary fibrosis. DST-3, a newly synthesized derivative of CTS, was designed to overcome these limitations. Methods: The antifibrotic effects of DST-3 were investigated in a bleomycin-induced pulmonary fibrosis model in C57BL/6 mice through lung function assessment, histopathological evaluation, hydroxyproline quantification, and cytokine profiling. In vitro, TGF-β1-stimulated MRC5 fibroblasts were employed to explore the mechanism of action, focusing on STAT3/Smad signaling via Western blotting and molecular binding assays. Furthermore, a validated HPLC–MS/MS method was developed for DST-3, and its pharmacokinetic profile was characterized in Sprague–Dawley rats and compared with that of CTS. Results: DST-3 markedly attenuated pulmonary fibrosis in vivo, as evidenced by improved lung function, reduced collagen deposition, and decreased proinflammatory cytokine levels. In vitro, DST-3 inhibited TGF-β1-induced fibroblast activation by directly binding to STAT3 and suppressing STAT3/Smad signaling. Pharmacokinetic analysis demonstrated that, compared with CTS, DST-3 exhibited more rapid absorption, a higher peak plasma concentration, a greater area under the curve (AUC), improved hepatic metabolic stability, and enhanced lung tissue exposure. Conclusions: Our study demonstrates that DST-3 exerts potent antifibrotic effects in vivo and in vitro, primarily through STAT3 pathway inhibition. Its improved pharmacokinetic characteristics further support its potential as a promising candidate for the treatment of pulmonary fibrosis. Full article
(This article belongs to the Section Pharmacokinetics and Pharmacodynamics)
Show Figures

Graphical abstract

20 pages, 983 KB  
Review
Lipid Peroxidation in Diabetic Kidney Disease: Mechanism and Natural Solution
by Yuxin Dong and Yanqing Tong
Int. J. Mol. Sci. 2025, 26(19), 9764; https://doi.org/10.3390/ijms26199764 - 7 Oct 2025
Viewed by 214
Abstract
Diabetic kidney disease (DKD), as one of the most serious microvascular complications of diabetes, is the main cause of end-stage renal disease in the world. Lipid peroxidation plays a crucial role in the development and progression of DKD. Under conditions of high glucose [...] Read more.
Diabetic kidney disease (DKD), as one of the most serious microvascular complications of diabetes, is the main cause of end-stage renal disease in the world. Lipid peroxidation plays a crucial role in the development and progression of DKD. Under conditions of high glucose and insulin resistance, renal lipid metabolism disorders result in abnormal accumulation of polyunsaturated fatty acids (PUFAs), which undergo lipid peroxidation via free radical chain reactions to generate reactive aldehydes. These substances not only directly damage the cell structure but can also be used as signaling molecules that activate pathways related to inflammation, fibrosis, and ferroptosis, eventually leading to glomerular sclerosis and tubulointerstitial fibrosis. Natural products have shown considerable application prospects in the treatment of DKD due to their multi-functional properties, including anti-inflammatory, antioxidant, and lipid-metabolism-regulating effects. To elucidate this, we conducted a systematic review of the literature available in electronic databases (including PubMed, Web of Science, and Scopus, and Google Scholar) from January 2000 to May 2025. This study further discusses the therapeutic effect and mechanism of natural products targeting lipid peroxidation in DKD. The results indicate that natural products are promising anti-lipid peroxidation drugs. Further clinical trials will be necessary to verify the safety and effectiveness of these natural compounds in clinical applications, thereby laying the foundation for developing novel treatment strategies for DKD. Full article
(This article belongs to the Section Molecular Biology)
Show Figures

Figure 1

25 pages, 988 KB  
Review
The NO Pathway as a Target in Patients with Stable and Advanced Heart Failure: An Additional Arrow in Our Quiver!
by Saverio D’Elia, Carmine Gentile, Achille Solimene, Rosa Franzese, Ettore Luisi, Antonio Caiazzo, Luigi Marotta, Simona Covino, Francesco Natale, Francesco S. Loffredo, Paolo Golino and Giovanni Cimmino
Biomolecules 2025, 15(10), 1420; https://doi.org/10.3390/biom15101420 - 6 Oct 2025
Viewed by 314
Abstract
The nitric oxide (NO) pathway is a fundamental regulator of vascular tone, myocardial function, and inflammation. In heart failure (HF), especially in advanced stages, dysregulation of NO–soluble guanylate cyclase (sGC)–cyclic guanosine monophosphate (cGMP) signaling contributes to endothelial dysfunction, increased vascular resistance, myocardial fibrosis, [...] Read more.
The nitric oxide (NO) pathway is a fundamental regulator of vascular tone, myocardial function, and inflammation. In heart failure (HF), especially in advanced stages, dysregulation of NO–soluble guanylate cyclase (sGC)–cyclic guanosine monophosphate (cGMP) signaling contributes to endothelial dysfunction, increased vascular resistance, myocardial fibrosis, and impaired cardiac performance. Chronic inflammation further reduces NO bioavailability, exacerbating HF progression This review synthesizes current knowledge on the role of the NO pathway in HF pathophysiology, with a focus on stable and advanced HF. Special attention is given to patient subgroups with comorbidities such as chronic kidney disease, where modulation of NO signaling may be particularly beneficial. We also evaluate therapeutic strategies targeting NO bioavailability and sGC stimulation. Evidence shows that impaired NO signaling promotes systemic and pulmonary vasoconstriction, elevates ventricular afterload, and worsens cardiac remodeling. Pharmacological agents that restore NO levels or activate downstream effectors such as sGC improve vasodilation, reduce fibrosis, and enhance myocardial relaxation. These effects are especially relevant in advanced HF patients and those with renal impairment, who often exhibit limited responses to conventional therapies. The NO pathway represents a promising therapeutic target in both stable and advanced HF. Modulating this pathway could improve outcomes, particularly in complex populations with multiple comorbidities, highlighting the need for further clinical research and tailored treatments. Full article
Show Figures

Figure 1

17 pages, 13481 KB  
Article
Luopan Mountain Pig Bone Marrow Mesenchymal Stem Cells Promote Liver Regeneration in D-Galactosamine-Induced Acute Liver Failure Rats by Regulating the PTEN-PI3K/Akt/mTOR Pathway
by Minjuan Li, Zhongfa Wang, Xingxing Yan, Yanchen Liu, Yunan He, Bianying Zhang and Weijun Guan
Biology 2025, 14(10), 1363; https://doi.org/10.3390/biology14101363 - 5 Oct 2025
Viewed by 110
Abstract
Treatment for acute liver failure (ALF) is constrained by shortages of liver transplant donors and immune rejection. Porcine bone marrow mesenchymal stem cells (pBMSCs) demonstrate clinical potential in xenotransplantation due to their abundant availability, low immunogenicity, and strong proliferative activity. This study is [...] Read more.
Treatment for acute liver failure (ALF) is constrained by shortages of liver transplant donors and immune rejection. Porcine bone marrow mesenchymal stem cells (pBMSCs) demonstrate clinical potential in xenotransplantation due to their abundant availability, low immunogenicity, and strong proliferative activity. This study is the first to investigate the reparative effects and mechanisms of pBMSCs derived from Luopan Mountain pigs in a D-galactosamine (D-GalN)-induced ALF rat model. The results demonstrated that tail-vein transplantation of pBMSCs significantly improved survival rates in ALF rats; reduced serum ALT, AST, and TBIL levels; enhanced hepatic glycogen metabolism; and mitigated histopathological liver damage. Additionally, pBMSC transplantation upregulated serum HGF, IGF-1, and VEGF levels while inhibiting hepatocyte apoptosis. Mechanistic studies indicate that pBMSCs promote liver function recovery and regeneration by activating the PI3K/Akt/mTOR signaling pathway and suppressing its key negative regulator, PTEN, by regulating the expression of key genes involved in inflammation, fibrosis, proliferation, and apoptosis. This study provides crucial experimental evidence for the use of pBMSCs in treating acute liver failure (ALF) and lays the groundwork for its clinical translation in the field of xenotransplantation. Full article
(This article belongs to the Section Cell Biology)
Show Figures

Figure 1

31 pages, 2025 KB  
Review
Emerging Radioligands as Tools to Track Multi-Organ Senescence
by Anna Gagliardi, Silvia Migliari, Alessandra Guercio, Giorgio Baldari, Tiziano Graziani, Veronica Cervati, Livia Ruffini and Maura Scarlattei
Diagnostics 2025, 15(19), 2518; https://doi.org/10.3390/diagnostics15192518 - 4 Oct 2025
Viewed by 268
Abstract
Senescence is a dynamic, multifaceted process implicated in tissue aging, organ dysfunction, and intricately associated with numerous chronic diseases. As senescent cells accumulate, they drive inflammation, fibrosis, and metabolic disruption through the senescence-associated secretory phenotype (SASP). Despite its clinical relevance, senescence remains challenging [...] Read more.
Senescence is a dynamic, multifaceted process implicated in tissue aging, organ dysfunction, and intricately associated with numerous chronic diseases. As senescent cells accumulate, they drive inflammation, fibrosis, and metabolic disruption through the senescence-associated secretory phenotype (SASP). Despite its clinical relevance, senescence remains challenging to detect non-invasively due to its heterogeneous nature and the lack of universal biomarkers. Recent advances in the development of specific imaging probes for positron emission tomography (PET) enable in vivo visualization of senescence-associated pathways across key organs, such as the lung, heart, kidney, and metabolic processes. For instance, [18F]FPyGal, a β-galactosidase-targeted tracer, has demonstrated selective accumulation in senescent cells in both preclinical and early clinical studies, while FAP-targeted radioligands are emerging as tools for imaging fibrotic remodeling in the lung, liver, kidney, and myocardium. This review examines a new generation of PET radioligands targeting hallmark features of senescence, with the potential to track and measure the process, the ability to be translated into clinical interventions for early diagnosis, and longitudinal monitoring of senescence-driven pathologies. By integrating organ-specific imaging biomarkers with molecular insights, PET probes are poised to transform our ability to manage and treat age-related diseases through personalized approaches. Full article
(This article belongs to the Section Medical Imaging and Theranostics)
Show Figures

Figure 1

15 pages, 993 KB  
Review
Antioxidants in Cardiovascular Health: Implications for Disease Modeling Using Cardiac Organoids
by Gracious R. Ross and Ivor J. Benjamin
Antioxidants 2025, 14(10), 1202; https://doi.org/10.3390/antiox14101202 - 3 Oct 2025
Viewed by 388
Abstract
Cardiovascular disease remains the leading cause of mortality worldwide, and at its molecular core lies a silent disruptor: oxidative stress. This imbalance between reactive oxygen species (ROS) and antioxidant defenses not only damages cellular components but also orchestrates a cascade of pathological events [...] Read more.
Cardiovascular disease remains the leading cause of mortality worldwide, and at its molecular core lies a silent disruptor: oxidative stress. This imbalance between reactive oxygen species (ROS) and antioxidant defenses not only damages cellular components but also orchestrates a cascade of pathological events across diverse cardiac cell types. In cardiomyocytes, ROS overload impairs contractility and survival, contributing to heart failure and infarction. Cardiac fibroblasts respond by promoting fibrosis through excessive collagen deposition. Macrophages intensify inflammatory responses, such as atherosclerosis, via ROS-mediated lipid oxidation—acting both as mediators of damage and targets for antioxidant intervention. This review examines how oxidative stress affects cardiac cell types and evaluates antioxidant-based therapeutic strategies. Therapeutic approaches include natural antioxidants (e.g., polyphenols and vitamins) and synthetic agents (e.g., enzyme modulators), which show promise in experimental models by improving myocardial remodeling. However, clinical trials reveal inconsistent outcomes, underscoring translational challenges (e.g., clinical biomarkers). Emerging strategies—such as targeted antioxidant delivery, activation of endogenous pathways, and disease modeling using 3D organoids—aim to enhance efficacy. In conclusion, we spotlight innovative technologies—like lab-grown heart tissue models—that help scientists better understand how oxidative stress affects heart health. These tools are bridging the gap between early-stage research and personalized medicine, opening new possibilities for diagnosing and treating heart disease more effectively. Full article
Show Figures

Figure 1

12 pages, 256 KB  
Review
The Role of Bleomycin Sclerotherapy in Venous Malformation Management: A Narrative Review
by Aikaterini Bini, Christos Topalidis, Triantafyllia Koletsa, Athanasios Papas, Efterpi Demiri and Leonidas Pavlidis
Life 2025, 15(10), 1553; https://doi.org/10.3390/life15101553 - 3 Oct 2025
Viewed by 315
Abstract
Venous malformations (VMs) are rare, non-involuting, slow-flow, congenital anomalies of vascular morphogenesis, presenting as dilated venous channels with reduced perivascular cell coverage. The treatment may be conservative or surgical, including laser therapy. The management of small superficial VMs typically involves surgical excision. In [...] Read more.
Venous malformations (VMs) are rare, non-involuting, slow-flow, congenital anomalies of vascular morphogenesis, presenting as dilated venous channels with reduced perivascular cell coverage. The treatment may be conservative or surgical, including laser therapy. The management of small superficial VMs typically involves surgical excision. In larger or deeper VMs, the intralesional–endovascular injection of the sclerosing agent bleomycin is the gold standard, as it eliminates the dysplastic venous vessels by inducing fibrosis and therefore promotes regression of the lesion. This review explores the current literature regarding the role of bleomycin in venous malformation management, emphasizing the molecular pathways involved, the efficacy of sclerotherapy with bleomycin and its complications and the associated management challenges. It evaluates the clinical and histological features of venous malformations, alongside diagnostic methodologies and treatment strategies, drawing on the most recent bibliographic data. The literature was systematically reviewed using the PubMed database, offering insights into future research directions and highlighting innovative treatment approaches. Full article
(This article belongs to the Special Issue Trends in Clinical Research 2025)
26 pages, 735 KB  
Review
Protective Effects of PACAP in Diabetic Complications: Retinopathy, Nephropathy and Neuropathy
by Dora Reglodi, Andrea Tamas, Inez Bosnyak, Tamas Atlasz, Edina Szabo, Lina Li, Gabriella Horvath, Balazs Opper, Peter Kiss, Liliana Lucas, Grazia Maugeri, Agata Grazia D’Amico, Velia D’Agata, Eszter Fabian, Gyongyver Reman and Alexandra Vaczy
Int. J. Mol. Sci. 2025, 26(19), 9650; https://doi.org/10.3390/ijms26199650 - 3 Oct 2025
Viewed by 194
Abstract
Pituitary adenylate cyclase-activating polypeptide (PACAP) is a neuropeptide exerting, among others, strong trophic and protective effects. It plays a role in several physiological functions, including glucose homeostasis. The protective effects of PACAP are mainly mediated via its specific PAC1 receptor by stimulating anti-inflammatory, [...] Read more.
Pituitary adenylate cyclase-activating polypeptide (PACAP) is a neuropeptide exerting, among others, strong trophic and protective effects. It plays a role in several physiological functions, including glucose homeostasis. The protective effects of PACAP are mainly mediated via its specific PAC1 receptor by stimulating anti-inflammatory, anti-apoptotic and antioxidant pathways. The aim of the present review is to summarize data on the protective effects of PACAP in the three major complications of diabetes, retinopathy, nephropathy and neuropathy, as well as some other complications. In type 1 and type 2 diabetic retinopathy models and in glucose-exposed cells of the eye, PACAP counteracted the degeneration of retinal layers and inhibited apoptosis and factors leading to abnormal vessel growth. In models of nephropathy, kidney morphology was better retained after PACAP administration, with decreased apoptosis and fibrosis. In diabetic neuropathy, PACAP protected against axonal–myelin lesions and less activation in pain processing centers. This neuropeptide has several other beneficial effects in diabetes-induced complications like altered vascular response, cognitive deficits and atherosclerosis. The promising therapeutic effects of PACAP in several pathological conditions have encouraged researchers to design PACAP-related drugs and to develop ways to enhance tissue delivery. These intentions are expected to result in overcoming the hurdles preventing PACAP from being introduced into therapeutic treatments, including diabetes-related conditions. Full article
Show Figures

Figure 1

26 pages, 1799 KB  
Review
Mechanotransduction-Epigenetic Coupling in Pulmonary Regeneration: Multifunctional Bioscaffolds as Emerging Tools
by Jing Wang and Anmin Xu
Pharmaceuticals 2025, 18(10), 1487; https://doi.org/10.3390/ph18101487 - 2 Oct 2025
Viewed by 235
Abstract
Pulmonary fibrosis (PF) is a progressive and fatal lung disease characterized by irreversible alveolar destruction and pathological extracellular matrix (ECM) deposition. Currently approved agents (pirfenidone and nintedanib) slow functional decline but do not reverse established fibrosis or restore functional alveoli. Multifunctional bioscaffolds present [...] Read more.
Pulmonary fibrosis (PF) is a progressive and fatal lung disease characterized by irreversible alveolar destruction and pathological extracellular matrix (ECM) deposition. Currently approved agents (pirfenidone and nintedanib) slow functional decline but do not reverse established fibrosis or restore functional alveoli. Multifunctional bioscaffolds present a promising therapeutic strategy through targeted modulation of critical cellular processes, including proliferation, migration, and differentiation. This review synthesizes recent advances in scaffold-based interventions for PF, with a focus on their dual mechano-epigenetic regulatory functions. We delineate how scaffold properties (elastic modulus, stiffness gradients, dynamic mechanical cues) direct cell fate decisions via mechanotransduction pathways, exemplified by focal adhesion–cytoskeleton coupling. Critically, we highlight how pathological mechanical inputs establish and perpetuate self-reinforcing epigenetic barriers to regeneration through aberrant chromatin states. Furthermore, we examine scaffolds as platforms for precision epigenetic drug delivery, particularly controlled release of inhibitors targeting DNA methyltransferases (DNMTi) and histone deacetylases (HDACi) to disrupt this mechano-reinforced barrier. Evidence from PF murine models and ex vivo lung slice cultures demonstrate scaffold-mediated remodeling of the fibrotic niche, with key studies reporting substantial reductions in collagen deposition and significant increases in alveolar epithelial cell markers following intervention. These quantitative outcomes highlight enhanced alveolar epithelial plasticity and upregulating antifibrotic gene networks. Emerging integration of stimuli-responsive biomaterials, CRISPR/dCas9-based epigenetic editors, and AI-driven design to enhance scaffold functionality is discussed. Collectively, multifunctional bioscaffolds hold significant potential for clinical translation by uniquely co-targeting mechanotransduction and epigenetic reprogramming. Future work will need to resolve persistent challenges, including the erasure of pathological mechanical memory and precise spatiotemporal control of epigenetic modifiers in vivo, to unlock their full therapeutic potential. Full article
(This article belongs to the Section Pharmacology)
Show Figures

Figure 1

16 pages, 1307 KB  
Article
Dual Endothelin Receptor Inhibition with Bosentan Does Not Prevent the Early Formation of Post-Traumatic Joint Contracture in a Rat Model
by Erik Wegner, Dennis Warnke, Victoria Buschmann, Benedikt Hild, Alexander Pirkl, Ulrike Ritz, Austin Harper, Erol Gercek, Philipp Drees and Andreas Baranowski
J. Clin. Med. 2025, 14(19), 6975; https://doi.org/10.3390/jcm14196975 - 1 Oct 2025
Viewed by 226
Abstract
Background: Post-traumatic joint contracture (PTJC) remains one of the most prevalent and challenging complications arising from musculoskeletal trauma or surgical intervention. Conventional treatment modalities are largely reactive and address symptoms after onset, yet provide limited efficacy once contracture has developed. In contrast, pharmacological [...] Read more.
Background: Post-traumatic joint contracture (PTJC) remains one of the most prevalent and challenging complications arising from musculoskeletal trauma or surgical intervention. Conventional treatment modalities are largely reactive and address symptoms after onset, yet provide limited efficacy once contracture has developed. In contrast, pharmacological strategies targeting the underlying inflammatory and fibrotic pathways offer a promising strategy for preventing the development of PTJC altogether. Methods: A total of 26 male Sprague Dawley rats underwent standardized knee trauma followed by immobilization for a duration of two weeks. Rats were randomized into two groups. The experimental group (n = 13) received bosentan at a dosage of 50 mg/kg twice daily throughout the immobilization period. The control group (n = 13) received a placebo instead. Joint mobility was quantitatively assessed by measuring the contracture angle (CA) and resistance to extension. In addition, posterior joint capsule tissues were harvested for histological analysis and subjected to quantitative PCR (qPCR) to quantify the expression of profibrotic genes, including α-Sma, Il-6, Tgf-β1, Nfκ-b, Ctgf. Results: Bosentan had no relevant effect on the biomechanics of the contracture compared to the placebo group. The contracture angle was comparable between the groups (86.8° ± 14.1°, 84.8° ± 11.1°). Similarly, the force required to achieve knee joint extension was comparable between the groups. Gene expression analysis also provided no evidence of reduced expression of pro-inflammatory or profibrotic genes. Histological assessments revealed no change in the absolute or relative number of myofibroblasts, or in the number of vessels, in the posterior joint capsules of the rats treated with bosentan. Compared to the control group, the number of myofibroblasts significantly increased in both the bosentan and control groups (p < 0.001, one-way ANOVA). Conclusions: Bosentan’s purported antifibrotic properties do not appear to confer a preventative effect on the development of PTJC. These findings suggest that, despite its potential in modulating fibrosis, bosentan does not mitigate the progression of the fibrotic condition. Furthermore, the involvement of endothelin-1 (ET-1) in the pathophysiology of PTJC remains yet to be fully understood, warranting further investigation. Full article
(This article belongs to the Section Orthopedics)
Show Figures

Figure 1

16 pages, 629 KB  
Review
Alcohol-Induced Oxidative Stress and Gut–Liver–Brain Crosstalk: Expanding the Paradigm from ALD to MetALD
by Jeong-Yoon Lee, Young-Min Jee, Keungmo Yang and Tom Ryu
Antioxidants 2025, 14(10), 1196; https://doi.org/10.3390/antiox14101196 - 1 Oct 2025
Viewed by 530
Abstract
Alcohol-associated liver disease (ALD) includes a spectrum from steatosis and steatohepatitis to cirrhosis and hepatocellular carcinoma driven by oxidative stress, immune activation, and systemic inflammation. Ethanol metabolism through alcohol dehydrogenase, aldehyde dehydrogenase, and cytochrome P450 2E1 generates reactive oxygen and nitrogen species, leading [...] Read more.
Alcohol-associated liver disease (ALD) includes a spectrum from steatosis and steatohepatitis to cirrhosis and hepatocellular carcinoma driven by oxidative stress, immune activation, and systemic inflammation. Ethanol metabolism through alcohol dehydrogenase, aldehyde dehydrogenase, and cytochrome P450 2E1 generates reactive oxygen and nitrogen species, leading to mitochondrial dysfunction, hepatocellular injury, and activation of inflammatory and fibrogenic pathways. Beyond hepatic effects, ALD engages the gut–liver–brain axis, where microbial dysbiosis, blood–brain barrier disruption, and neuroinflammation contribute to cognitive impairment and cerebrovascular risk. The emerging concept, metabolic dysfunction-associated steatotic liver disease and increased alcohol intake (MetALD), presents the synergistic impact of alcohol and metabolic comorbidities, enhancing oxidative injury and fibrosis. This review summarizes key mechanisms connecting oxidative stress to multisystem pathology and highlights the need for precision therapies targeting redox imbalance, immune dysregulation, and gut–brain–liver interactions to improve outcomes in ALD and MetALD. Full article
(This article belongs to the Special Issue Alcohol-Induced Oxidative Stress in Health and Disease, 2nd Edition)
Show Figures

Figure 1

23 pages, 2407 KB  
Review
YKL-40 in Virus-Associated Liver Disease: A Translational Biomarker Linking Fibrosis, Hepatocarcinogenesis, and Liver Transplantation
by Jadranka Pavicic Saric, Dinka Lulic, Dunja Rogic, Stipislav Jadrijevic, Danko Mikulic, Tajana Filipec Kanizaj, Nikola Prpic, Laura Karla Bozic, Ivona Adamovic, Iva Bacak Kocman, Zrinka Sarec, Gorjana Erceg, Mirta Adanic, Petra Ozegovic Zuljan, Filip Jadrijevic and Ileana Lulic
Int. J. Mol. Sci. 2025, 26(19), 9584; https://doi.org/10.3390/ijms26199584 - 1 Oct 2025
Viewed by 276
Abstract
Virus-associated hepatocellular carcinoma (HCC) remains a major global health burden despite effective antiviral therapies. Chronic infection with hepatitis B (HBV), hepatitis C (HCV), and hepatitis D (HDV) promotes malignant transformation through overlapping pathways of fibrosis, immune dysregulation, and microenvironmental remodeling. YKL-40, a glycoprotein [...] Read more.
Virus-associated hepatocellular carcinoma (HCC) remains a major global health burden despite effective antiviral therapies. Chronic infection with hepatitis B (HBV), hepatitis C (HCV), and hepatitis D (HDV) promotes malignant transformation through overlapping pathways of fibrosis, immune dysregulation, and microenvironmental remodeling. YKL-40, a glycoprotein secreted by hepatic stellate cells, hepatocytes under stress, macrophages, and endothelial cells, has emerged as a marker that reflects stromal activation rather than direct hepatocyte injury. Its expression is reinforced by profibrotic and angiogenic circuits, and circulating concentrations correlate with advanced fibrosis, residual risk after viral suppression, and oncologic outcomes. This review synthesizes current evidence on YKL-40 across HBV, HCV, and HDV cohorts, with emphasis on its role in bridging molecular mechanisms to clinical applications. We examine its utility in non-invasive fibrosis assessment, longitudinal monitoring after antiviral therapy, and prognostic modeling in HCC. Particular attention is given to its potential in the liver transplant pathway, where YKL-40 may refine eligibility beyond morphology, inform bridging therapy response, and predict post-transplant recurrence or graft fibrosis. Remaining challenges include its lack of disease specificity, assay variability, and limited multicenter validation. Future integration of YKL-40 into multimarker, algorithm-based frameworks could enable risk-adaptive strategies that align surveillance and transplant decisions with the evolving biology of virus-associated liver disease. Full article
Show Figures

Graphical abstract

35 pages, 1106 KB  
Review
Integrating Novel Biomarkers into Clinical Practice: A Practical Framework for Diagnosis and Management of Cardiorenal Syndrome
by Georgios Aletras, Maria Bachlitzanaki, Maria Stratinaki, Emmanuel Lamprogiannakis, Ioannis Petrakis, Emmanuel Foukarakis, Yannis Pantazis, Michael Hamilos and Kostas Stylianou
Life 2025, 15(10), 1540; https://doi.org/10.3390/life15101540 - 1 Oct 2025
Viewed by 435
Abstract
Cardiorenal syndrome (CRS) reflects the intricate and bidirectional interplay between cardiac and renal dysfunction, commonly resulting in diagnostic uncertainty, therapeutic dilemmas and poor outcomes. While traditional biomarkers like serum creatinine (Cr) and natriuretic peptides remain widely used, their limitations in specificity, timing and [...] Read more.
Cardiorenal syndrome (CRS) reflects the intricate and bidirectional interplay between cardiac and renal dysfunction, commonly resulting in diagnostic uncertainty, therapeutic dilemmas and poor outcomes. While traditional biomarkers like serum creatinine (Cr) and natriuretic peptides remain widely used, their limitations in specificity, timing and contextual interpretation often hinder optimal management. This narrative review synthesizes the current evidence on established and emerging biomarkers in CRS, with emphasis on their clinical relevance, integration into real-world practice, and potential to inform precision therapy. Markers of glomerular filtration rate beyond creatinine—such as cystatin C—offer more accurate assessment in frail or sarcopenic patients, while tubular injury markers such as NGAL, KIM-1, and urinary L-FABP (uL-FABP) provide early signals of structural renal damage. The FDA-approved NephroCheck® test—based on TIMP-2 and IGFBP7— enables risk stratification for imminent AKI up to 24 h before functional decline. Congestion-related markers such as CA125 and bio-adrenomedullin outperform natriuretic peptides in certain CRS phenotypes, particularly in right-sided heart failure or renally impaired patients. Fibrosis and inflammation markers (galectin-3, sST2, GDF-15) add prognostic insights, especially when combined with NT-proBNP or troponin. Rather than presenting biomarkers in isolation, this review proposes a framework that links them to specific clinical contexts—such as suspected decongestion-related renal worsening or persistent congestion despite therapy—to support actionable interpretation. A tailored, scenario-based, multi-marker strategy may enhance diagnostic precision and treatment safety in CRS. Future research should prioritize prospective biomarker-guided trials and standardized pathways for clinical integration. Full article
(This article belongs to the Special Issue Cardiorenal Disease: Pathogenesis, Diagnosis, and Treatments)
Show Figures

Figure 1

21 pages, 2663 KB  
Review
Pharmacological Potential and Mechanisms of Bisbenzylisoquinoline Alkaloids from Lotus Seed Embryos
by Yan Liu, Cong Wang, Qiong Liao and Canwei Du
Biomolecules 2025, 15(10), 1377; https://doi.org/10.3390/biom15101377 - 28 Sep 2025
Viewed by 355
Abstract
Lotus seed embryos, a key component in traditional Chinese medicine, have attracted growing scientific interest due to their wide-ranging therapeutic potential. Among the bioactive compounds found in lotus seed embryos, three bisbenzylisoquinoline alkaloids—liensinine, isoliensinine, and neferine—stand out for their diverse pharmacological activities. These [...] Read more.
Lotus seed embryos, a key component in traditional Chinese medicine, have attracted growing scientific interest due to their wide-ranging therapeutic potential. Among the bioactive compounds found in lotus seed embryos, three bisbenzylisoquinoline alkaloids—liensinine, isoliensinine, and neferine—stand out for their diverse pharmacological activities. These alkaloids are known to exhibit significant antitumor, anti-inflammatory, antihypertensive, neuroprotective, and antifibrotic effects, which make them promising candidates for the treatment of various chronic and acute diseases. Recent studies have highlighted their ability to modulate key signaling pathways involved in cancer progression, inflammation, fibrosis, and neurodegeneration. The precise mechanisms underlying their actions include modulation of oxidative stress, inhibition of pro-inflammatory cytokines, regulation of apoptosis, and modulation of cellular metabolism. This review aims to provide an in-depth overview of the pharmacological relevance of these alkaloids, focusing on their mechanisms of action and their therapeutic potential across different disease models. By synthesizing current evidence from preclinical studies, this review also lays a solid scientific foundation for future research, supporting the rational design and development of lotus-derived compounds for clinical application. Full article
Show Figures

Figure 1

Back to TopTop