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

Article Types

Countries / Regions

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Search Results (336)

Search Parameters:
Keywords = matrix-degrading enzyme

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
13 pages, 922 KB  
Review
Biofilms and Chronic Wounds: Pathogenesis and Treatment Options
by Annabel Z. Shen, Mohamad Taha, Mahmoud Ghannoum and Stephen K. Tyring
J. Clin. Med. 2025, 14(21), 7784; https://doi.org/10.3390/jcm14217784 (registering DOI) - 2 Nov 2025
Abstract
Introduction: Chronic wounds are a growing healthcare challenge, with infections being major complications that delay healing. Biofilms are structured microbial communities encased in extracellular polymeric substances. Biofilms confer antimicrobial resistance, promote inflammation, and protect pathogens from host defenses. These mechanisms make eradication difficult [...] Read more.
Introduction: Chronic wounds are a growing healthcare challenge, with infections being major complications that delay healing. Biofilms are structured microbial communities encased in extracellular polymeric substances. Biofilms confer antimicrobial resistance, promote inflammation, and protect pathogens from host defenses. These mechanisms make eradication difficult with standard therapies. Methods: A focused literature review was conducted using PubMed (2010–2025) to examine the role of biofilms in chronic wounds, diabetic foot ulcers (DFUs), and burn injuries, as well as conventional and emerging treatment strategies. Studies are included if they addressed microbial composition, host–microbe interactions, or therapeutic outcomes in clinical or translational models. Discussion: Biofilms are implicated in up to 60% of chronic wounds and more than half of burn wounds. In DFUs, both bacterial and fungal biofilms contribute to chronicity and impaired healing. Conventional treatments such as debridement and antiseptics reduce surface biofilm burden but rarely achieve full eradication. Emerging approaches include quorum sensing inhibitors, bacteriophage therapy, matrix-degrading enzymes, electroceutical dressings, antifungal strategies, and nanotechnology. They show promise when integrated with standard wound care. Conclusions: Biofilms are central to the pathogenesis of chronic wounds, DFUs, and burns. Integrating mechanism-based antibiofilm therapies with standard care represents a key research priority to improve healing outcomes. Full article
(This article belongs to the Section Dermatology)
Show Figures

Figure 1

16 pages, 1813 KB  
Article
The Effect of IL-17A and Combined Mechanical Injury on Meniscal Tissue Integrity In Vitro
by Greta Ahrens, Florian Gellhaus, Jan-Tobias Weitkamp, Peter Behrendt, François Cossais, Bernd Rolauffs, Alan J. Grodzinsky and Bodo Kurz
J. Clin. Med. 2025, 14(21), 7573; https://doi.org/10.3390/jcm14217573 - 25 Oct 2025
Viewed by 233
Abstract
Objectives: Meniscal integrity is crucial for knee joint stability and the prevention of osteoarthritis (OA) development. Recent studies suggested that mechanical overload and interleukin (IL)-17A may be important intertwined players in meniscal degeneration, but a direct impact of IL-17A on the meniscus [...] Read more.
Objectives: Meniscal integrity is crucial for knee joint stability and the prevention of osteoarthritis (OA) development. Recent studies suggested that mechanical overload and interleukin (IL)-17A may be important intertwined players in meniscal degeneration, but a direct impact of IL-17A on the meniscus has not been investigated. Therefore, the aim of this study was to analyze the effect of IL-17A on meniscal tissue with and without combined mechanical injury (MI). Methods: Meniscal explant disks (1 mm height, 3 mm diameter) were isolated from bovine menisci (preserving the native tibial superficial zone) and exposed to IL-17A [0–100 ng/mL] and/or MI (single compression, 50% strain, strain rate 1 mm/sec). After three days of incubation in a serum-free medium, the proteoglycan release (sGAG; DMMB assay), mRNA level of matrix-degrading enzymes (qRT-PCR), aggrecan degradation (NITEGE immunostaining), and cell death (histomorphometry of nuclear blebbing/apoptosis and condensed nuclei/unspecified cell death) were determined. Statistics: one- and two-way ANOVA with Tukey’s multiple comparisons or Kruskal–Wallis with post hoc testing. Results: IL-17A increased sGAG release in a dose-dependent significant manner. MI also induced the release of sGAG significantly, but the combination with IL-17A showed the highest levels. Both IL-17A and MI individually affected the mRNA levels for ADAMTS4 and MMP-13 slightly, but the combination of both particularly induced a significant increase in mRNA levels. Signals for the ADAMTS4-related aggrecan neoepitope NITEGE were elevated by IL-17A in superficial areas of the excised tissue and by MI in superficial and deeper areas. The combination of both stimuli intensified this signal further. MI increased the number of cells with condensed nuclei significantly and induced apoptosis in a small proportion of cells. IL-17A had no significant impact on the amount of condensed or apoptotic nuclei. Conclusions: Our findings emphasize an interaction between inflammatory cytokine IL-17A signaling and mechanical stress since IL-17A induced matrix degeneration in meniscal tissue, which intensified in combination with a trauma. The latter might create a post-traumatic environment that promotes meniscal degeneration and subsequently osteoarthritis progression. Full article
(This article belongs to the Section Orthopedics)
Show Figures

Figure 1

19 pages, 3310 KB  
Article
The Preparation and Evaluation of Carvacrol-Added Hyaluronic Acid for Early Osteoarthritis Treatment
by Yu-Ping Chen, Jhih-Ni Lin, Chia-Tien Chang, Yu-Ying Lin, Che-Yung Kuan, Yu-Chun Chen and Feng-Huei Lin
Antioxidants 2025, 14(10), 1265; https://doi.org/10.3390/antiox14101265 - 21 Oct 2025
Viewed by 551
Abstract
Osteoarthritis (OA) is a prevalent degenerative joint disease characterized by cartilage degradation, synovial inflammation, and subchondral bone remodeling, leading to chronic pain and reduced mobility. In early-stage OA, sustained oxidative stress and inflammation drive chondrocyte dysfunction and extracellular matrix (ECM) loss. Hyaluronic acid [...] Read more.
Osteoarthritis (OA) is a prevalent degenerative joint disease characterized by cartilage degradation, synovial inflammation, and subchondral bone remodeling, leading to chronic pain and reduced mobility. In early-stage OA, sustained oxidative stress and inflammation drive chondrocyte dysfunction and extracellular matrix (ECM) loss. Hyaluronic acid (HA), a key component of synovial fluid responsible for lubrication and viscoelasticity, is prone to enzymatic and oxidative degradation under inflammatory conditions, limiting its therapeutic effect. To address this, we developed an HA-based system incorporating the natural antioxidant and anti-inflammatory molecule carvacrol. The potential of this formulation was assessed in interleukin-1b-stimulated chondrocytes, which mimic the inflammatory environment of OA. The carvacrol-added HA combination upregulated antioxidant enzyme expression, attenuated pro-inflammatory signaling, and promoted ECM preservation by up regulating cartilage-specific markers and glycosaminoglycan production. In vivo efficacy was further evaluated in a rat model of monosodium iodoacetate-induced OA. HA-Carvacrol treatment alleviated pain-related behaviors and preserved cartilage structure, as confirmed by behavioral assessments and histological analyses. This dual-function formulation integrates the lubricating benefits of HA with the bioactivity of carvacrol, providing preclinical proof-of-concept evidence for its potential in early-stage OA. Full article
(This article belongs to the Special Issue Inflammation and Oxidative Stress in Articular Cartilage)
Show Figures

Graphical abstract

18 pages, 789 KB  
Review
Elastin in the Pathogenesis of Abdominal Aortic Aneurysm
by Dunpeng Cai and Shi-You Chen
Cells 2025, 14(20), 1597; https://doi.org/10.3390/cells14201597 - 14 Oct 2025
Viewed by 542
Abstract
Abdominal aortic aneurysms (AAAs) are progressive, life-threatening vascular disorders characterized by focal dilation of the abdominal aorta due to chronic weakening of the arterial wall. The condition often remains asymptomatic until rupture, which carries mortality rates exceeding 70–85%. Among the various etiological theories [...] Read more.
Abdominal aortic aneurysms (AAAs) are progressive, life-threatening vascular disorders characterized by focal dilation of the abdominal aorta due to chronic weakening of the arterial wall. The condition often remains asymptomatic until rupture, which carries mortality rates exceeding 70–85%. Among the various etiological theories of AAA development, degradation of the extracellular matrix (ECM) has emerged as the most widely accepted paradigm, with the breakdown of elastin representing a central and irreversible hallmark event. Elastin, a highly cross-linked and durable structural protein, provides elasticity and recoil to the aortic wall. In human AAA specimens, reduced elastin content, impaired cross-linking, and extensive fiber fragmentation are consistently observed, while experimental studies across multiple animal models confirm that elastin degradation directly correlates with aneurysm initiation, expansion, and rupture risk. Elastin loss is driven by a complex interplay of proteolytic enzymes coupled with inflammatory cell infiltration and oxidative stress. Furthermore, elastin-derived peptides perpetuate immune cell recruitment and matrix degradation, creating a vicious cycle of wall injury. Genetic and epigenetic factors, including variants in ECM regulators and dysregulation of non-coding RNAs, further modulate elastin homeostasis in AAA pathobiology. Clinically, biomarkers of elastin turnover and elastin-targeted molecular imaging techniques are emerging as tools for risk stratification. Therapeutically, novel strategies aimed at stabilizing elastin fibers, enhancing cross-linking, or delivering drugs directly to sites of elastin damage have shown promise in preclinical models and early translational studies. In parallel, regenerative approaches employing stem cells, exosomes, and bioengineered elastin scaffolds are under development to restore structural integrity. Collectively, these advances underscore the pivotal roles of elastin not only as a structural determinant of aneurysm development but also as a diagnostic and therapeutic target. This review summarizes and integrates recent discoveries on elastin biology in AAA, with a particular emphasis on molecular mechanisms of elastin degradation and the translational potential of elastin-centered interventions for the prevention and treatment of AAA. Full article
(This article belongs to the Special Issue Molecular Pathogenesis of Cardiovascular Diseases)
Show Figures

Figure 1

23 pages, 2203 KB  
Review
The Influence of Sirtuin 6 on Chondrocyte Senescence in Osteoarthritis Under Aging: Focusing on Mitochondrial Dysfunction and Oxidative Stress
by Huiying Zhao and Wei Wu
Antioxidants 2025, 14(10), 1228; https://doi.org/10.3390/antiox14101228 - 13 Oct 2025
Viewed by 958
Abstract
Osteoarthritis (OA) is one of the most common joint diseases worldwide, which is characterized by degenerative changes in articular cartilage and secondary osteophyte formation. Numerous factors influence OA, including aging, obesity, joint injury and chronic overloading. Among them, the senescence of chondrocytes is [...] Read more.
Osteoarthritis (OA) is one of the most common joint diseases worldwide, which is characterized by degenerative changes in articular cartilage and secondary osteophyte formation. Numerous factors influence OA, including aging, obesity, joint injury and chronic overloading. Among them, the senescence of chondrocytes is one of the key factors leading to OA. Chondrocyte senescence can trigger inflammatory responses, extracellular matrix (ECM) degradation, mitochondrial dysfunction and oxidative stress (OS), and autophagy. Sirtuin 6 (SIRT6), as a deacetylase related to aging, can regulate chondrocyte senescence and plays a certain part in OA. SIRT6 regulates the number and membrane integrity of mitochondria, alleviates excessive Reactive Oxygen Species (ROS) in mitochondria and reduces inflammation-mediated mitochondrial damage. In addition, SIRT6 can also regulate the activity of antioxidant enzymes, inhibit excessive ROS induced by inflammatory factors, and alleviate OS. However, as aging progresses, the activity of SIRT6 will decrease. Activating the activity of SIRT6 becomes a potential therapeutic target and has a certain alleviating effect on the development of OA. The supplementation of nicotinamide adenine dinucleotide (NAD+) precursors and SIRT6-specific activators can increase SIRT6, alleviate chondrocyte senescence, and reduce OA. This paper aims to focus on mitochondrial dysfunction and OS to explore SIRT6’s effects on OA chondrocytes’ senescence under aging and summarize the potential therapeutic targets for activating SIRT6 to provide assistance for the improvement of OA. Full article
(This article belongs to the Special Issue Inflammation and Oxidative Stress in Articular Cartilage)
Show Figures

Figure 1

16 pages, 4838 KB  
Article
Critical Requirement of Senescence-Associated CCN3 Expression in CD44-Positive Stem Cells for Osteoarthritis Progression
by Janvier Habumugisha, Ryuichiro Okuda, Kazuki Hirose, Miho Kuwahara, Ziyi Wang, Mitsuaki Ono, Hiroshi Kamioka, Satoshi Kubota and Takako Hattori
Int. J. Mol. Sci. 2025, 26(19), 9630; https://doi.org/10.3390/ijms26199630 - 2 Oct 2025
Viewed by 556
Abstract
Osteoarthritis (OA) is a degenerative joint disease characterized by progressive cartilage breakdown, synovial inflammation, and subchondral bone remodeling. Previous studies have shown that cellular communication network factor 3 (CCN3) expression increases with age in cartilage, and its overexpression promotes OA-like changes by inducing [...] Read more.
Osteoarthritis (OA) is a degenerative joint disease characterized by progressive cartilage breakdown, synovial inflammation, and subchondral bone remodeling. Previous studies have shown that cellular communication network factor 3 (CCN3) expression increases with age in cartilage, and its overexpression promotes OA-like changes by inducing senescence-associated secretory phenotypes. This study aimed to investigate the effect of Ccn3 knockout (KO) on OA development using a murine OA model. Destabilization of the medial meniscus (DMM) surgery was performed in wild-type (WT) and Ccn3-KO mice. Histological scoring and staining were used to assess cartilage degeneration and proteoglycan loss. Gene and protein expressions of catabolic enzyme (Mmp9), hypertrophic chondrocyte marker (Col10a1), senescence marker, and cyclin-dependent kinase inhibitor 1A (Cdkn1a) were evaluated. Single-cell RNA sequencing (scRNA-seq) data from WT and Sox9-deficient cartilage were reanalyzed to identify Ccn3+ progenitor populations. Immunofluorescence staining assessed CD44 and Ki67 expression in articular cartilage. The effects of Ccn3 knockdown on IL-1β-induced Mmp13 and Adamts5 expression in chondrocytes were examined in vitro. Ccn3 KO mice exhibited reduced cartilage degradation and catabolic gene expression compared with WT mice post-DMM. scRNA-seq revealed enriched Ccn3-Cd44 double-positive cells in osteoblast progenitor, synovial mesenchymal stem cell, and mesenchymal stem cell clusters. Immunofluorescence showed increased CCN3+/CD44+ cells in femoral and tibial cartilage and meniscus. Ki67+ cells were significantly increased in DMM-treated Ccn3 KO cartilage, mostly CD44+. In vitro Ccn3 knockdown attenuated IL-1β-induced Mmp13 and Adamts5 expressions in chondrocytes. Ccn3 contributes to OA pathogenesis by promoting matrix degradation, inducing hypertrophic changes, and restricting progenitor cell proliferation, highlighting Ccn3 as a potential therapeutic target for OA. Full article
(This article belongs to the Special Issue Advanced Molecular Mechanism of Pathogenesis of Osteoarthritis)
Show Figures

Figure 1

15 pages, 769 KB  
Article
Antioxidant, Anti-Melanogenic, and Anti-Aging Activities of the Aqueous–Ethanolic Dry Extract of Rosa lucieae with Phytochemical Profiling
by Yun Gyeong Park, Ji-Yul Kim, Seok-Chun Ko, Kyung Woo Kim, Dongwoo Yang, Du-Min Jo, Hyo-Geun Lee, Jeong Min Lee, Mi-Jin Yim, Chul Hwan Kim, Dae-Sung Lee, Hyun-Soo Kim and Gun-Woo Oh
Antioxidants 2025, 14(10), 1177; https://doi.org/10.3390/antiox14101177 - 26 Sep 2025
Viewed by 584
Abstract
In this study, the cosmeceutical potential of a 70% ethanol extract of Rosa lucieae was investigated as a multifunctional bioactive ingredient. The extract was systematically evaluated for its antioxidant, anti-melanogenic, and anti-aging properties, and was comprehensively phytochemically profiled using ultra-high-performance liquid chromatography–quadrupole time-of-flight [...] Read more.
In this study, the cosmeceutical potential of a 70% ethanol extract of Rosa lucieae was investigated as a multifunctional bioactive ingredient. The extract was systematically evaluated for its antioxidant, anti-melanogenic, and anti-aging properties, and was comprehensively phytochemically profiled using ultra-high-performance liquid chromatography–quadrupole time-of-flight mass spectrometry. The analysis tentatively identified 21 metabolites, including phenolic acids (gallic acid, ellagic acid, and corilagin), flavonoids (catechin, rutin, quercetin, hyperoside, and quercitrin), and glycosidic derivatives (e.g., phlorizin), several of which are well-documented for their skin-protective effects. Quantitative measurements confirmed high polyphenol and flavonoid contents, correlating with strong radical-scavenging and reducing capacities in α-diphenyl-β-picrylhydrazyl, 2,2′-azino-bis(3-ethylbenzothiazoline-6-sulfonic acid, as well as ferric ion reducing antioxidant power assays. Moreover, the extract inhibited tyrosinase activity and 3,4-dihydroxyphenylalanine oxidation, thereby suppressing melanin biosynthesis. In addition, marked inhibitory effects against collagenase, elastase, and hyaluronidase were observed; these enzymes are critically involved in extracellular matrix degradation and skin aging. Taken together, these results indicate that the biological activities of R. lucieae are supported by a diverse polyphenol- and flavonoid-rich chemical profile, highlighting the potential of this plant as a natural multifunctional ingredient for cosmeceutical, nutraceutical, functional food, and preventive healthcare applications. Full article
Show Figures

Figure 1

13 pages, 1097 KB  
Article
Astragalus membranaceus Extract Attenuates Inflammatory Cytokines and Matrix-Degrading Enzymes in Human Chondrocytes: A Novel Nutraceutical Strategy for Joint Health
by Alessia Mariano, Rosario Russo, Anna Scotto d’Abusco and Fabiana Superti
Curr. Issues Mol. Biol. 2025, 47(9), 731; https://doi.org/10.3390/cimb47090731 - 9 Sep 2025
Viewed by 872
Abstract
The dried root extract of Astragalus membranaceus, also known as Astragali radix, is widely used in traditional Chinese medicine for its multiple health benefits and well-established safety profile. Astragalus root extract exhibits several bioactive properties, including anti-inflammatory, antioxidant, antiviral and hepatoprotective [...] Read more.
The dried root extract of Astragalus membranaceus, also known as Astragali radix, is widely used in traditional Chinese medicine for its multiple health benefits and well-established safety profile. Astragalus root extract exhibits several bioactive properties, including anti-inflammatory, antioxidant, antiviral and hepatoprotective effects. Due to its unique features, it is being investigated in a novel application as a complementary remedy in the management of joint disorders. In this study, we evaluated the effect of Astragalus membranaceus hydroalcoholic root extract (0.01 and 0.1 mg/mL) in vitro on the HTB-94 cell line, a well-known model for studying inflammatory pathways in human chondrocytes. The mRNA modulation levels were measured by quantitative real-time polymerase chain reaction (qRT-PCR), while the protein secretion levels were assessed using an Enzyme-Linked Immunosorbent Assay (ELISA). Results obtained demonstrated that this extract is able to decrease the tumor necrosis factor-α (TNF-α)-induced inflammatory response by downregulating both the mRNA expression and release of the pro-inflammatory mediators Interleukin-6 (IL-6), Interleukin-1β (IL-1β) and Interelukin-8 (IL-8), as well as matrix metalloproteases, including Matrix Metalloprotease-3 (MMP-3), Matrix Metalloprotease-13 (MMP-13) and A disintegrin, and metalloproteinase with thrombospondin motifs 5 (ADAMTS-5). Moreover, the interleukin and matrix metalloprotease production was also assessed in non-TNF-α-stimulated cells, revealing that the extract did not alter the basal levels of these mediators. Finally, our findings highlight the potential benefits of Astragalus membranaceus extract, both in terms of its favorable safety profile and its efficacy mitigating joint inflammatory responses. These results support the potential of this extract as a nutraceutical agent for joint health support. Full article
(This article belongs to the Special Issue Role of Natural Products in Inflammatory Diseases)
Show Figures

Figure 1

17 pages, 3777 KB  
Article
Heparanase-Neutralizing Monoclonal Antibody (mAb A54) Attenuates Tumor Growth and Metastasis
by Uri Barash, Malik Farhoud, Maali Odeh, Eliezer Huberman, Liang Wu and Israel Vlodavsky
Cells 2025, 14(17), 1379; https://doi.org/10.3390/cells14171379 - 4 Sep 2025
Viewed by 974
Abstract
Heparanase is the only human enzyme responsible for heparan sulfate (HS) breakdown, an activity that remodels the extracellular matrix (ECM) and strongly drives cancer metastasis and angiogenesis. Compelling evidence implies that heparanase promotes essentially all aspects of the tumorigenic process, namely, tumor initiation, [...] Read more.
Heparanase is the only human enzyme responsible for heparan sulfate (HS) breakdown, an activity that remodels the extracellular matrix (ECM) and strongly drives cancer metastasis and angiogenesis. Compelling evidence implies that heparanase promotes essentially all aspects of the tumorigenic process, namely, tumor initiation, vascularization, growth, metastasis, and chemoresistance. A key mechanism by which heparanase accelerates cancer progression is by enabling the release and bioavailability of HS-bound growth factors, chemokines, and cytokines, residing in the tumor microenvironment and supporting tumor growth and metastasis. The currently available heparanase inhibitors are mostly HS/heparin-like compounds that lack specificity and exert multiple off-target side effects. To date, only four such compounds have progressed to clinical trials, and none have been approved for clinical use. We have generated and characterized an anti-heparanase monoclonal antibody (A54 mAb) that specifically inhibits heparanase enzymatic activity (ECM degradation assay) and cellular uptake. Importantly, A54 mAb attenuates xenograft tumor growth and metastasis (myeloma, glioma, pancreatic, and breast carcinomas) primarily when administered (syngeneic or immunocompromised mice) in combination with conventional anti-cancer drugs. Co-crystallization of the A54 Fab fragment and the heparanase enzyme revealed that the interaction between the two proteins takes place adjacent to the enzyme HS/heparin binding domain II (HBDII; Pro271-Ala276), likely hindering heparanase from interacting with HS substrates via steric occlusion of the active site cleft. Collectively, we have generated and characterized a novel mAb that specifically neutralizes heparanase enzymatic activity and attenuates its pro-tumorigenic effects in preclinical models, paving the way for its clinical examination against cancer, inflammation, and other diseases. Full article
Show Figures

Figure 1

12 pages, 1147 KB  
Communication
Synergistic Protective Effects of Oleaster Fruit and Sophora japonica L. Fruit Extracts Against IL-1β-Induced Inflammation in Human Chondrocytes
by Hana Lee, Jinyeong Lim, Myeonghwan Oh and Junsoo Lee
Foods 2025, 14(17), 3099; https://doi.org/10.3390/foods14173099 - 4 Sep 2025
Viewed by 942
Abstract
Osteoarthritis (OA) is a degenerative joint disease characterized by chronic inflammation and progressive cartilage breakdown. This study investigated the synergistic protective effects of oleaster fruit extract (OE) and Sophora japonica L. fruit extract (SJE) against interleukin-1β (IL-1β) -induced inflammation in human chondrocytes. OE [...] Read more.
Osteoarthritis (OA) is a degenerative joint disease characterized by chronic inflammation and progressive cartilage breakdown. This study investigated the synergistic protective effects of oleaster fruit extract (OE) and Sophora japonica L. fruit extract (SJE) against interleukin-1β (IL-1β) -induced inflammation in human chondrocytes. OE and SJE were tested individually and in combination at various ratios (3:1, 2:1, 1:1, 1:2, and 1:3). Among the tested mixtures, the 3:1 OE:SJE ratio exhibited the most pronounced synergistic effect. When used individually, the concentrations required to achieve 90% cell viability were 313.6 µg/mL for OE and 4135.8 µg/mL for SJE. In contrast, the same viability level was achieved with a combined treatment of 26.4 µg/mL OE and 8.8 µg/mL SJE, yielding a combination index of 0.25, indicative of strong synergy. The 3:1 OE:SJE combination also significantly suppressed inflammatory mediators, including nitric oxide, interleukin-6, and tumor necrosis factor-α. This treatment also led to the downregulation of matrix-degrading enzymes, such as matrix metalloproteinase (MMP)-9 and MMP-13, and it promoted the preservation of hyaluronan, a key extracellular matrix component. These findings suggest that the 3:1 OE:SJE combination exerts a synergistic protective effect by modulating both inflammatory and catabolic pathways and may represent a promising therapeutic strategy for OA prevention or treatment. Full article
(This article belongs to the Section Food Nutrition)
Show Figures

Graphical abstract

31 pages, 4629 KB  
Article
Mandragora autumnalis: Phytochemical Composition, Antioxidant and Anti-Cancerous Bioactivities on Triple-Negative Breast Cancer Cells
by Ghosoon Albahri, Adnan Badran, Heba Hellany, Serine Baydoun, Rola Abdallah, Mohamad Alame, Akram Hijazi, Marc Maresca and Elias Baydoun
Int. J. Mol. Sci. 2025, 26(17), 8506; https://doi.org/10.3390/ijms26178506 - 1 Sep 2025
Viewed by 838
Abstract
Breast cancer is a common and chronic condition, and despite improvements in diagnosis, treatment, and prevention, the number of cases of breast cancer is rising annually. New therapeutic drugs that target specific checkpoints should be created to fight breast cancer. Mandragora autumnalis possesses [...] Read more.
Breast cancer is a common and chronic condition, and despite improvements in diagnosis, treatment, and prevention, the number of cases of breast cancer is rising annually. New therapeutic drugs that target specific checkpoints should be created to fight breast cancer. Mandragora autumnalis possesses substantial cultural value as a herb and is regarded as one of the most significant medicinal plants; however, little is known about its anticancerous biological activity and chemopreventive molecular pathways against the triple-negative breast cancer (MDA-MB-231) cell line. In this study, the antioxidant, anticancer, and underlying molecular mechanisms of the Mandragora autumnalis ethanolic leaves extract (MAE) were evaluated, and its phytochemical composition was determined. Results indicated that MAE diminished the viability of MDA-MB-231 cells in a concentration- and time-dependent manner. Although MAE exhibited 55% radical scavenging activity at higher concentrations in the 2,2-diphenyl-1-picrylhydrazyl (DPPH) assay, the attenuation of its cytotoxic effects in MDA-MB-231 cells with N-acetylcysteine (NAC) co-treatment suggests a potential role of oxidative stress. Additionally, MAE caused an increase in the tumor suppressor p53. Moreover, this extract caused a significant decrease in the expression of Ki-67 (a cellular proliferation marker), MMP-9 (matrix metalloproteinase-9, an enzyme involved in extracellular matrix degradation and metastasis), and STAT-3 (a transcription factor regulating cell growth and survival). Also, MAE altered cell cycle, cell migration, angiogenesis, invasion, aggregation, and adhesion to suppress cellular processes linked to metastasis. All of our research points to MAE’s potential to function as an anticancer agent and opens up new possibilities for the development of innovative triple-negative breast cancer treatments. Full article
Show Figures

Graphical abstract

34 pages, 1707 KB  
Review
Mimicking Gastric Cancer Collagen Reorganization with Decellularized ECM-Based Scaffolds
by Néstor Corro, Sebastián Alarcón, Ángel Astroza, Roxana González-Stegmaier and Carolina Añazco
Biology 2025, 14(8), 1067; https://doi.org/10.3390/biology14081067 - 16 Aug 2025
Viewed by 1491
Abstract
The tumor microenvironment (TME) has a substantial impact on the progression of gastric cancer. Collagen, the most abundant protein in the extracellular matrix (ECM), forms a dense physical barrier that regulates anti-tumor immunity in the TME. It is a significant regulator of the [...] Read more.
The tumor microenvironment (TME) has a substantial impact on the progression of gastric cancer. Collagen, the most abundant protein in the extracellular matrix (ECM), forms a dense physical barrier that regulates anti-tumor immunity in the TME. It is a significant regulator of the signaling pathways of cancer cells, which are responsible for migration, proliferation, and metabolism. ECM proteins, particularly remodeling enzymes and collagens, can be modified to increase stiffness and alter the mechanical properties of the stroma. This, in turn, increases the invasive potential of tumor cells and resistance to immunotherapy. Given the dynamic nature of collagen, novel therapeutic strategies have emerged that target both collagen biosynthesis and degradation, processes that are essential for addressing ECM stiffening. This review delineates the upregulation of the expression and deposition of collagen, as well as the biological functions, assembly, and reorganization that contribute to the dissemination of this aggressive malignancy. Furthermore, the review emphasizes the importance of creating 3D in vitro models that incorporate innovative biomaterials that avoid the difficulties of traditional 2D culture in accurately simulating real-world conditions that effectively replicate the distinctive collagen microenvironment. Ultimately, it investigates the use of decellularized ECM-derived biomaterials as tumor models that are designed to precisely replicate the mechanisms associated with the progression of stomach cancer. Full article
(This article belongs to the Special Issue Tumor Biomechanics and Mechanobiology)
Show Figures

Graphical abstract

21 pages, 10081 KB  
Article
Melanoma–Keratinocyte Crosstalk Participates in Melanoma Progression with Mechanisms Partially Overlapping with Those of Cancer-Associated Fibroblasts
by Ramona Marrapodi, Daniela Kovacs, Emilia Migliano, Silvia Caputo, Federica Papaccio, Tiziano Pallara, Carlo Cota and Barbara Bellei
Int. J. Mol. Sci. 2025, 26(16), 7901; https://doi.org/10.3390/ijms26167901 - 15 Aug 2025
Viewed by 847
Abstract
The Tumour Microenvironment (TME) is pivotal for melanoma progression and contributes to therapy resistance. While dermal cell involvement is well established, the role of epidermal cells remains less defined. To explore the contribution of Normal Human Keratinocytes (NHKs) to melanoma biology, we investigated [...] Read more.
The Tumour Microenvironment (TME) is pivotal for melanoma progression and contributes to therapy resistance. While dermal cell involvement is well established, the role of epidermal cells remains less defined. To explore the contribution of Normal Human Keratinocytes (NHKs) to melanoma biology, we investigated the modification of gene and protein expression of NHKs exposed to melanoma-conditioned medium or maintained in a co-culture system. The analysis focused on pathways related to proliferation, inflammation, Extracellular Matrix (ECM) remodelling, and cell adhesion. Due to the well-documented melanoma–fibroblast crosstalk, Normal Human Fibroblasts (NHFs) and Cancer-Associated Fibroblasts (CAFs) were used as comparative references. Keratinocyte gene expression changes under the influence of melanoma secretome only partially overlapped with those of NHFs and CAFs, indicating cell-type-specific responses. Exposure to melanoma-conditioned medium induced the upregulation of bFGF, CXCL-16, TIMP-2, and E-cadherin in NHKs, alongside downregulating TGF-β and MMP-9. Although bFGF is a recognized pro-tumorigenic factor, the modulation of CXCL-16, TIMP-2, and TGF-β may reflect a protective response. Notably, under co-culture conditions, NHKs exhibited a pronounced pro-inflammatory and ECM-remodelling phenotype, characterized by elevated production of cytokines (IL-1α, IL-1β, and IL-8) and ECM-degrading enzymes (MMP-7, 9, 12, and 13), indicative of a pro-tumoral feature. Collectively, these findings underscore an active role for NHKs in melanoma initiation and progression. Full article
Show Figures

Figure 1

10 pages, 2056 KB  
Article
Complete Loss of Cramp Promotes Experimental Osteoarthritis with Enhanced Chondrocyte Apoptosis in Mice
by Moon-Chang Choi, Jiwon Jo and Junghee Park
Int. J. Mol. Sci. 2025, 26(16), 7874; https://doi.org/10.3390/ijms26167874 - 15 Aug 2025
Viewed by 641
Abstract
Osteoarthritis (OA) is the most prevalent form of joint arthritis, frequently associated with aging, mechanical wear, and inflammation. Our previous work demonstrated that cathelicidin-related antimicrobial peptide (Cramp) is upregulated in mouse OA cartilage, and that transient knockdown (KD) of Cramp in cultured chondrocytes [...] Read more.
Osteoarthritis (OA) is the most prevalent form of joint arthritis, frequently associated with aging, mechanical wear, and inflammation. Our previous work demonstrated that cathelicidin-related antimicrobial peptide (Cramp) is upregulated in mouse OA cartilage, and that transient knockdown (KD) of Cramp in cultured chondrocytes decreases IL-1β-induced expression of matrix-degrading enzymes. The aim of this study was to determine the in vivo role of Cramp in OA pathogenesis using whole-body Cramp knockout (KO) mice. Normal skeletal development and growth plate morphology were assessed in E18.5d embryos and 2-week-old mice, respectively. Expression profiles of catabolic and anabolic genes were analyzed in primary chondrocytes derived from Cramp KO mice. OA in mouse knee joints was induced using intra-articular monosodium iodoacetate (MIA) injections or surgical destabilization of the medial meniscus (DMM). We observed that Cramp loss does not impact normal skeletal development. In contrast to our expectations, complete Cramp deficiency in chondrocytes failed to decrease catabolic gene expression upon IL-1β stimulation. Instead, genetic deletion of Cramp significantly worsened OA cartilage degradation in both MIA- and DMM-induced models. The detrimental phenotype observed in Cramp-deficient mice results from enhanced chondrocyte apoptosis. Therefore, even minimal Cramp expression appears essential for maintaining catabolic balance and preventing chondrocyte apoptosis in OA cartilage. Collectively, our data indicate that Cramp may exert multifaceted effects on OA pathogenesis by modulating catabolic pathways and apoptosis. Full article
(This article belongs to the Special Issue Elucidating How Chondrocytes Maintain Cartilage Stability)
Show Figures

Figure 1

15 pages, 2605 KB  
Article
Dual-Compartment Anti-Inflammatory and Chondroprotective Effects of Intra-Articular Hydrolyzed Collagen in Experimental Osteoarthritis
by Mustafa Dinç, Ömer Cevdet Soydemir, Recep Karasu, Aysun Saricetin and Hunkar Cagdas Bayrak
Medicina 2025, 61(8), 1461; https://doi.org/10.3390/medicina61081461 - 14 Aug 2025
Viewed by 994
Abstract
Background and Objectives: Osteoarthritis (OA) is a degenerative joint disease involving inflammation, oxidative stress, and extracellular matrix (ECM) degradation, leading to cartilage damage and joint dysfunction. This study aimed to evaluate the chondroprotective effects of intra-articular hydrolyzed collagen in a rat model [...] Read more.
Background and Objectives: Osteoarthritis (OA) is a degenerative joint disease involving inflammation, oxidative stress, and extracellular matrix (ECM) degradation, leading to cartilage damage and joint dysfunction. This study aimed to evaluate the chondroprotective effects of intra-articular hydrolyzed collagen in a rat model of knee OA using a dual-compartment biochemical and histological approach. Materials and Methods: Twenty male Sprague-Dawley rats underwent ACL transection to induce osteoarthritis and were randomly assigned to receive intra-articular hydrolyzed collagen or saline once weekly for three weeks. At six weeks, knee joints were evaluated histologically using the Mankin score. Synovial fluid and cartilage homogenates were analyzed via enzyme-linked immunosorbent assay (ELISA) for cytokines, cartilage degradation markers, and oxidative stress indicators. Results: The collagen-treated group demonstrated significantly lower Mankin scores. Levels of pro-inflammatory cytokines, interleukin-1 beta (IL-1β), interleukin-6 (IL-6), and tumor necrosis factor-alpha (TNF-α), as well as cartilage degradation markers, matrix metalloproteinase-13 (MMP-13), C-terminal crosslinked telopeptide of type II collagen (CTX-II), and cartilage oligomeric matrix protein (COMP), were significantly reduced (p < 0.05). Additionally, oxidative stress indicators including inducible nitric oxide synthase (iNOS), total oxidant status (TOS), and oxidative stress index (OSI) were decreased, while total antioxidant status (TAS) was increased in both synovial fluid and cartilage homogenates (p < 0.05). Conclusions: Intra-articular hydrolyzed collagen reduced inflammation, oxidative stress, and extracellular matrix (ECM) degradation, indicating potential chondroprotective effects across both synovial and cartilage compartments. Full article
(This article belongs to the Section Orthopedics)
Show Figures

Figure 1

Back to TopTop