Sign in to use this feature.

Years

Between: -

Subjects

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Journals

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Article Types

Countries / Regions

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Search Results (515)

Search Parameters:
Keywords = protease-activated receptor-2

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
16 pages, 745 KiB  
Review
Bidirectional Interplay Between Microglia and Mast Cells
by Szandra Lakatos and Judit Rosta
Int. J. Mol. Sci. 2025, 26(15), 7556; https://doi.org/10.3390/ijms26157556 - 5 Aug 2025
Abstract
Microglia, the brain’s resident innate immune cells, play a fundamental role in maintaining neural homeostasis and mediating responses to injury or infection. Upon activation, microglia undergo morphological and functional changes, including phenotypic switching between pro- and anti-inflammatory types and the release of different [...] Read more.
Microglia, the brain’s resident innate immune cells, play a fundamental role in maintaining neural homeostasis and mediating responses to injury or infection. Upon activation, microglia undergo morphological and functional changes, including phenotypic switching between pro- and anti-inflammatory types and the release of different inflammatory mediators. These processes contribute to neuroprotection and the pathogenesis of various central nervous system (CNS) disorders. Mast cells, although sparsely located in the brain, exert a significant influence on neuroinflammation through their interactions with microglia. Through degranulation and secretion of different mediators, mast cells disrupt the blood–brain barrier and modulate microglial responses, including alteration of microglial phenotypes. Notably, mast cell-derived factors, such as histamine, interleukins, and tryptase, activate microglia through various pathways including protease-activated receptor 2 and purinergic receptors. These interactions amplify inflammatory cascades via various signaling pathways. Previous studies have revealed an exceedingly complex crosstalk between mast cells and microglia suggesting a bidirectional regulation of CNS immunity, implicating their cooperation in both neurodegenerative progression and repair mechanisms. Here, we review some of the diverse communication pathways involved in this complex interplay. Understanding this crosstalk may offer novel insights into the cellular dynamics of neuroinflammation and highlight potential therapeutic targets for a variety of CNS disorders. Full article
Show Figures

Figure 1

17 pages, 1229 KiB  
Review
The Role of PAR2 in MASLD Progression and HCC Development
by Pietro Guerra, Patrizia Pontisso and Andrea Martini
Int. J. Mol. Sci. 2025, 26(15), 7076; https://doi.org/10.3390/ijms26157076 - 23 Jul 2025
Viewed by 223
Abstract
Metabolic dysfunction-associated steatotic liver disease (MASLD) has recently become the leading cause of chronic liver disease and can progress to hepatocellular carcinoma (HCC) through multiple pathogenic mechanisms. Protease-activated receptor 2 (PAR2) is a G-protein-coupled receptor activated by proteases such as trypsin, tryptase or [...] Read more.
Metabolic dysfunction-associated steatotic liver disease (MASLD) has recently become the leading cause of chronic liver disease and can progress to hepatocellular carcinoma (HCC) through multiple pathogenic mechanisms. Protease-activated receptor 2 (PAR2) is a G-protein-coupled receptor activated by proteases such as trypsin, tryptase or coagulation factors VII and Xa. Recent studies have shown that PAR2 expression is increased in the liver of patients with MASLD or liver fibrosis. Its activation is linked to metabolic dysfunction through several pathways, including SREBP1c activation, AMPK inhibition and Akt-induced insulin resistance. Inhibition of PAR2 has been effective in reducing MASLD progression in different animal models. Notably, PAR2 blockade has also been effective in more advanced stages of the disease by dampening chronic inflammation and fibrogenesis through the inhibition of hepatic stellate cell activation and of TGF-β and SerpinB3 production. PAR2 also plays a role in cancer development, promoting tumour proliferation, angiogenesis and expression of immune checkpoint inhibitors (like PD-L1, CD47 and CD24). Due to its multifaceted involvement in liver disease, PAR2 is emerging as a key therapeutic target in this clinical context. This review aims to summarise current knowledge on PAR2′s role in MASLD and its potential as a therapeutic target. Full article
(This article belongs to the Special Issue Obesity and Cancer Risk: Molecular Mechanisms and Perspectives)
Show Figures

Figure 1

16 pages, 3372 KiB  
Article
Soybean Trypsin Inhibitor Possesses Potency Against SARS-CoV-2 Infection by Blocking the Host Cell Surface Receptors ACE2, TMPRSS2, and CD147
by Wen-Liang Wu, Jaung-Geng Lin, Wen-Ping Jiang, Hsi-Pin Hung, Atsushi Inose and Guan-Jhong Huang
Int. J. Mol. Sci. 2025, 26(14), 6583; https://doi.org/10.3390/ijms26146583 - 9 Jul 2025
Viewed by 383
Abstract
Angiotensin-converting enzyme 2 (ACE2) is a cell-surface receptor that helps the body regulate blood pressure and endocrine secretions. Transmembrane serine protease 2 (TMPRSS2) is a cell surface protein expressed mainly by endothelial cells of the respiratory and digestive tract, which participates in the [...] Read more.
Angiotensin-converting enzyme 2 (ACE2) is a cell-surface receptor that helps the body regulate blood pressure and endocrine secretions. Transmembrane serine protease 2 (TMPRSS2) is a cell surface protein expressed mainly by endothelial cells of the respiratory and digestive tract, which participates in the cleavage of protein peptide bonds with serine as the active site. These two proteins have been studied to be highly associated with infection with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). Soybean trypsin inhibitor (SBTI) has special bioactivities such as anticarcinogenic and anti-inflammatory functions, which can be widely used in functional foods or drugs. Our study involved in vitro and in vivo experiments to elucidate the effect of SBTI on SARS-CoV-2 host invasion. First, it was confirmed that being under 250 μg/mL of SBTI was not toxic to HepG2, HEK293T, and Calu-3 cells. The animal study administered SBTI to mice once daily for 14 days. In the lungs, liver, and kidneys, the histopathologic findings of the SBTI group were not different from those of the control group, but the expression of ACE2, TMPRSS2, and CD147 was reduced. Thus, our findings suggest that the inhibition of ACE2, TMPRSS,2 and CD147 proteins by SBTI shows promise in potentially inhibiting SARS-CoV-2 infection. Full article
(This article belongs to the Special Issue New Advances in Bioactive Compounds in Health and Disease)
Show Figures

Graphical abstract

23 pages, 2062 KiB  
Review
Potential Compounds as Inhibitors of Staphylococcal Virulence Factors Involved in the Development of Thrombosis
by Anna Lichota, Krzysztof Gwozdzinski and Monika Sienkiewicz
Toxins 2025, 17(7), 340; https://doi.org/10.3390/toxins17070340 - 4 Jul 2025
Viewed by 422
Abstract
For many years, staphylococci have been detected mainly in infections of the skin and soft tissues, organs, bone inflammations, and generalized infections. Thromboembolic diseases have also become a serious plague of our times, which, as it turns out, are closely related to the [...] Read more.
For many years, staphylococci have been detected mainly in infections of the skin and soft tissues, organs, bone inflammations, and generalized infections. Thromboembolic diseases have also become a serious plague of our times, which, as it turns out, are closely related to the toxic effects of staphylococci. Staphylococcus aureus, because of the presence of many different kinds of virulence factors, is capable of manipulating the host’s innate and adaptive immune responses. These include toxins and cofactors that activate host zymogens and exoenzymes, as well as superantigens, which are highly inflammatory and cause leukocyte death. Coagulases and staphylokinases can control the host’s coagulation system. Nucleases and proteases inactivate various immune defense and surveillance proteins, including complement components, peptides and antibacterial proteins, and surface receptors that are important for leukocyte chemotaxis. On the other hand, secreted toxins and exoenzymes are proteins that disrupt the endothelial and epithelial barrier as a result of cell lysis and disintegration of linking proteins, which ultimately increases the risk of thromboembolism. In this review, we discuss various virulence factors and substances that may inhibit their activity. Full article
Show Figures

Graphical abstract

21 pages, 735 KiB  
Article
Characterizing zonulin and par2 Expression in Zonulin Transgenic and Zonulin Inhibition Mouse Models of Motility and Inflammation
by Enid E. Martinez, Jordan D. Philpott, Jinggang Lan, K. Marco Rodriguez Hovnanian and Alessio Fasano
Int. J. Mol. Sci. 2025, 26(13), 6381; https://doi.org/10.3390/ijms26136381 - 2 Jul 2025
Viewed by 330
Abstract
We aimed to examine the effect of zonulin and zonulin inhibition on gastrointestinal (GI) motility and the mRNA expression of zonulin and the protease-activated receptor 2 (par2), the primary receptor for zonulin, under conditions of inflammation by lipopolysaccharide (LPS) injection. The [...] Read more.
We aimed to examine the effect of zonulin and zonulin inhibition on gastrointestinal (GI) motility and the mRNA expression of zonulin and the protease-activated receptor 2 (par2), the primary receptor for zonulin, under conditions of inflammation by lipopolysaccharide (LPS) injection. The experimental models included zonulin transgenic mice (ztm), par2 knockout ztm (ztm-par2 −/−), ztm exposed to the zonulin inhibitor AT1001 (ztm-AT1001), and wildtype mouse controls. GI transit was measured by fluorescein isothiocyanate-dextran and mRNA expression by real-time quantitative polymerase chain reaction in whole, and in epithelial and non-epithelial tissues of all GI segments. There were no differences in the GI transit between mouse groups at baseline. After the LPS injection, ztm mice had an attenuated slowing of the GI transit compared to wildtype mice. The zonulin-inhibited mice had motility patterns similar to wildtype mice. zonulin upregulation was noted in GI segments of the ztm, ztm-par2 −/−, and ztm-AT1001 after the LPS injection. Differences in motility patterns between ztm and zonulin inhibition models despite zonulin expression in GI segments of all mouse groups supports that PAR2 is key for zonulin’s effect on motility under conditions of inflammation. However, the findings from the epithelial and non-epithelial compartments suggest that the pathway of activity is complex and likely indirect. Full article
(This article belongs to the Special Issue The Role of Tight Junction Proteins in Health and Disease)
Show Figures

Figure 1

15 pages, 1864 KiB  
Article
Administration of Purified Alpha-1 Antitrypsin in Salt-Loaded Hypertensive 129Sv Mice Attenuates the Expression of Inflammatory Associated Proteins in the Kidney
by Van-Anh L. Nguyen, Yunus E. Dogan, Niharika Bala, Erika S. Galban, Sihong Song and Abdel A. Alli
Biomolecules 2025, 15(7), 951; https://doi.org/10.3390/biom15070951 - 30 Jun 2025
Viewed by 384
Abstract
Background: Alpha-1 antitrypsin (AAT) is a multifunctional protease inhibitor that has been shown to have anti-inflammatory properties in various diseases. AAT has been reported to protect against renal injury via anti-apoptotic, anti-fibrotic, and anti-inflammatory effects. However, its role in mitigating renal inflammation and [...] Read more.
Background: Alpha-1 antitrypsin (AAT) is a multifunctional protease inhibitor that has been shown to have anti-inflammatory properties in various diseases. AAT has been reported to protect against renal injury via anti-apoptotic, anti-fibrotic, and anti-inflammatory effects. However, its role in mitigating renal inflammation and reducing high blood pressure induced by salt-loading has never been studied. Methods: In this study, we salt-loaded 129Sv mice to induce hypertension and then administered purified human AAT (hAAT) or the vehicle to investigate whether renal inflammation and associated inflammatory/signaling pathways are mitigated. Results: Western blotting and densitometric analysis showed administration of hAAT attenuated protein expression of kidney injury molecule-1 (KIM1), CD93, CD36, and the toll-like receptor 2 and 4 (TLR-2/4) in kidney lysates. Similarly, protein expression of two key inflammatory transcription factors, signal transducer and activator of transcription 3 (STAT3) and NF-Kappa B were shown to be attenuated in the kidneys of 129Sv mice that received hAAT. Conversely, hAAT treatment upregulated the expression of heat shock protein 70 (HSP70) and immunohistochemistry confirmed these findings. Conclusions: Purified hAAT administration may be efficacious in mitigating renal inflammation associated with the development of hypertension from salt-loading, potentially through a mechanism involving the reduction of pro-inflammatory and injury-associated proteins. Full article
(This article belongs to the Section Molecular Medicine)
Show Figures

Figure 1

19 pages, 1219 KiB  
Review
Carboxylesterase Factors Influencing the Therapeutic Activity of Common Antiviral Medications Used for SARS-CoV-2 Infection
by Yue Shen, William Eades, Linh Dinh and Bingfang Yan
Pharmaceutics 2025, 17(7), 832; https://doi.org/10.3390/pharmaceutics17070832 - 26 Jun 2025
Viewed by 584
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), the virus responsible for COVID-19, remains a major global health threat. The virus enters host cells by binding to the angiotensin-converting enzyme 2 (ACE2) receptor. Several small-molecule antiviral drugs, including molnupiravir, favipiravir, remdesivir, and nirmatrelvir have [...] Read more.
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), the virus responsible for COVID-19, remains a major global health threat. The virus enters host cells by binding to the angiotensin-converting enzyme 2 (ACE2) receptor. Several small-molecule antiviral drugs, including molnupiravir, favipiravir, remdesivir, and nirmatrelvir have been shown to inhibit SARS-CoV-2 replication and are approved for treating SARS-CoV-2 infections. Nirmatrelvir inhibits the viral main protease (Mpro), a key enzyme for processing polyproteins in viral replication. In contrast, molnupiravir, favipiravir, and remdesivir are prodrugs that target RNA-dependent RNA polymerase (RdRp), which is crucial for genome replication and subgenomic RNA production. However, undergoing extensive metabolism profoundly impacts their therapeutic effects. Carboxylesterases (CES) are a family of enzymes that play an essential role in the metabolism of many drugs, especially prodrugs that require activation through hydrolysis. Molnupiravir is activated by carboxylesterase-2 (CES2), while remdesivir is hydrolytically activated by CES1 but inhibits CES2. Nirmatrelvir and remdesivir are oxidized by the same cytochrome P450 (CYP) enzyme. Additionally, various transporters are involved in the uptake or efflux of these drugs and/or their metabolites. It is well established that drug-metabolizing enzymes and transporters are differentially expressed depending on the cell type, and these genes exhibit significant polymorphisms. In this review, we examine how CES-related cellular and genetic factors influence the therapeutic activities of these widely used COVID-19 medications. This article highlights implications for improving product design, targeted inhibition, and personalized medicine by exploring genetic variations and their impact on drug metabolism and efficacy. Full article
(This article belongs to the Special Issue ADME Properties in the Drug Delivery)
Show Figures

Figure 1

25 pages, 1445 KiB  
Review
The Role of Astrocytes in Synaptic Dysfunction and Memory Deficits in Alzheimer’s Disease
by Cristina A. Muñoz de León-López, Irene Navarro-Lobato and Zafar U. Khan
Biomolecules 2025, 15(7), 910; https://doi.org/10.3390/biom15070910 - 20 Jun 2025
Viewed by 822
Abstract
Astrocytes are the most abundant glial cells in the brain. They play critical roles in synapse formation and function, neurotransmitter release and uptake, the production of trophic factors, and energy supply for neuronal survival. In addition to producing proteases for amyloid-β degradation, astrocytes [...] Read more.
Astrocytes are the most abundant glial cells in the brain. They play critical roles in synapse formation and function, neurotransmitter release and uptake, the production of trophic factors, and energy supply for neuronal survival. In addition to producing proteases for amyloid-β degradation, astrocytes express various receptors, transporters, gliotransmitters, and other molecules that enable them to sense and respond to external signals. They are also implicated in amyloid-β clearance. In Alzheimer’s disease, excessive accumulation of amyloid-β induces the polarization of astrocytes into the A1 phenotype, promoting the release of inflammatory cytokines and mitochondrial reactive oxygen species, leading to alterations in astrocytic functions. Under such conditions, gliotransmitter release, glutamate neurotransmission, AMPA receptor trafficking, and both Hebbian and non-Hebbian forms of synaptic plasticity—biological activities essential for synaptic functions—are compromised. Moreover, astrocytes are essential for learning, memory, and synaptic plasticity, and alterations in their function are associated with memory deficits in Alzheimer’s disease. This review provides an overview of the current understanding of the defects in astrocytes that lead to altered synaptic functions, neuronal structural plasticity, and memory deficits in Alzheimer’s disease. Full article
(This article belongs to the Special Issue The Role of Astrocytes in Neurodegenerative Diseases)
Show Figures

Figure 1

20 pages, 2636 KiB  
Article
Effect of Potassium–Magnesium Sulfate on Intestinal Dissociation and Absorption Rate, Immune Function, and Expression of NLRP3 Inflammasome, Aquaporins and Ion Channels in Weaned Piglets
by Cui Zhu, Kaiyong Huang, Xiaolu Wen, Kaiguo Gao, Xuefen Yang, Zongyong Jiang, Shuting Cao and Li Wang
Animals 2025, 15(12), 1751; https://doi.org/10.3390/ani15121751 - 13 Jun 2025
Viewed by 456
Abstract
This study investigated the effects of potassium magnesium sulfate (PMS) on intestinal dissociation and absorption rate, immune function, and expression of the NOD-like receptor thermal domain-associated protein 3 (NLRP3) inflammasome, aquaporins (AQPs), and potassium and magnesium ion channels in weaned piglets. Experiment 1 [...] Read more.
This study investigated the effects of potassium magnesium sulfate (PMS) on intestinal dissociation and absorption rate, immune function, and expression of the NOD-like receptor thermal domain-associated protein 3 (NLRP3) inflammasome, aquaporins (AQPs), and potassium and magnesium ion channels in weaned piglets. Experiment 1 involved the assessment of the dissociation rate of PMS in pig digestive fluid and the absorption rate of PMS in the small intestine using an Ussing chamber in vitro. In Experiment 2, 216 healthy 21-day-old weaned piglets were selected and randomly assigned to six groups (0%, 0.15%, 0.30%, 0.45%, 0.60%, and 0.75% PMS), with each group 6 replicates of six piglets per replicate. The in vitro Ussing chamber results indicated that the absorption of K+ and Mg2+ in the jejunum and ileum was significantly higher than that in the duodenum (p < 0.05). The in vivo study demonstrated that the addition of PMS resulted in a linear increase in serum K+, IgG, and interleukin (IL)-2 levels while simultaneously reducing serum IL-1β levels (p < 0.05). Dietary PMS significantly elevated serum IL-10 and Mg2+ levels in feces (p < 0.05). Furthermore, supplementation with 0.60% or 0.75% PMS significantly downregulated the mRNA expression of NLRP3 in the jejunum (p < 0.05). Dietary PMS supplementation linearly reduced the mRNA expression levels of cysteine protease 1 (Caspase-1) and IL-1β in both the jejunum and colon as well as the mRNA expression levels of two-pore domain channel subfamily K member 5 (KCNK5) in these regions (p < 0.05). Notably, supplementation with 0.15% PMS significantly decreased the mRNA expression of transient receptor potential channel 6 (TRPM6) in the jejunum and significantly increased the expression of TRPM6 in the colon (p < 0.05). Dietary addition of 0.45% and 0.60% PMS significantly increased the mRNA expression of aquaporin 3 (AQP3) in the colon (p < 0.05), whereas 0.75% PMS significantly increased the mRNA expression of aquaporin 8 (AQP8) in both the jejunum and colon. Moreover, the expression levels of AQP3 and AQP8 were significantly negatively correlated with the diarrhea rate observed between days 29 and 42. In conclusion, dietary PMS supplementation improved immune function, inhibited the activation of intestinal NLRP3, and modulated the expression of water and ion channels in weaned piglets, thereby contributing to the maintenance of intestinal water and ion homeostasis, which could potentially alleviate post-weaning diarrhea in piglets. The recommended supplemental level of PMS in the corn-soybean basal diet for weaned piglets is 0.30%. Full article
(This article belongs to the Section Animal Nutrition)
Show Figures

Figure 1

24 pages, 1036 KiB  
Review
ADAM Proteases in Cancer: Biological Roles, Therapeutic Challenges, and Emerging Opportunities
by Sakshi Arora, Andrew M. Scott and Peter W. Janes
Cancers 2025, 17(10), 1703; https://doi.org/10.3390/cancers17101703 - 19 May 2025
Viewed by 1868
Abstract
ADAM (A Disintegrin and Metalloproteinase) family members are multifunctional transmembrane proteases that govern tumorigenesis and metastasis by cleaving membrane-bound substrates such as growth factors, cytokines, and cell adhesion molecules. Several ADAMs, including ADAM8, ADAM9, ADAM10, ADAM12, and ADAM17, are overexpressed in malignancies and [...] Read more.
ADAM (A Disintegrin and Metalloproteinase) family members are multifunctional transmembrane proteases that govern tumorigenesis and metastasis by cleaving membrane-bound substrates such as growth factors, cytokines, and cell adhesion molecules. Several ADAMs, including ADAM8, ADAM9, ADAM10, ADAM12, and ADAM17, are overexpressed in malignancies and are linked with a poor prognosis. These proteases contribute to tumour growth by regulating cell proliferation, cell fate, invasion, angiogenesis, and immune evasion. ADAM10 and ADAM17, especially, facilitate the shedding of critical developmental and growth factors and their receptors, as well as immuno-regulatory molecules, hence promoting tumour progression, immune escape, and resistance to therapy. Recent work has unveiled multiple regulatory pathways that modulate ADAM functions, which include trafficking, dimerization, and conformational modifications that affect substrate accessibility. These observations have rekindled efforts to produce selective ADAM inhibitors, avoiding the off-target consequences reported with early small molecule inhibitors targeting the enzyme active site, which is conserved also in matrix metalloproteinases (MMPs). Promising approaches tested in preclinical models and, in some cases, clinical settings include more selective small-molecule inhibitors, monoclonal antibodies, and antibody–drug conjugates designed to specifically target ADAMs. In this review, we will discuss the emerging roles of ADAMs in cancer biology, as well as the molecular processes that control their function. We further discuss the therapeutic potential of targeting ADAMs, with a focus on recent advances and future directions in the development of ADAM-specific cancer therapies. Full article
Show Figures

Figure 1

25 pages, 10876 KiB  
Article
The Influence of Anti-PAR 1 and Anti-ACE 2 Antibody Levels on the Course of Specific Glomerulonephritis Types
by Maciej Szymczak, Harald Heidecke, Marcelina Żabińska, Łucja Janek, Jakub Wronowicz, Krzysztof Kujawa, Kai Schulze-Forster, Karolina Marek-Bukowiec, Tomasz Gołębiowski and Mirosław Banasik
J. Clin. Med. 2025, 14(9), 3178; https://doi.org/10.3390/jcm14093178 - 4 May 2025
Viewed by 618
Abstract
Background: Anti-PAR 1 (protease-activated receptor 1) and anti-ACE 2 (angiotensin 2-converting enzyme 2) antibodies are a kind of non-HLA (human leukocyte antigens) antibodies postulated to be of significance in autoimmunological diseases and organ transplantation. Methods: We assessed anti-PAR 1 and anti-ACE 2 antibody [...] Read more.
Background: Anti-PAR 1 (protease-activated receptor 1) and anti-ACE 2 (angiotensin 2-converting enzyme 2) antibodies are a kind of non-HLA (human leukocyte antigens) antibodies postulated to be of significance in autoimmunological diseases and organ transplantation. Methods: We assessed anti-PAR 1 and anti-ACE 2 antibody levels in patients with membranous nephropathy n= 18, focal and segmental glomerulosclerosis (FSGS) n = 25, lupus nephritis (LN) n = 17, IgA nephropathy n = 14, mesangial proliferative (non-IgA) glomerulonephritis n = 6, c-ANCA (cytoplasmic anti-neutrophil cytoplasmic antibodies) vasculitis n = 40, p (perinuclear)-ANCA vasculitis n = 16, and compared them with a healthy control group n = 22. Next, we observed the clinical course of the patients (creatinine, total protein, and albumin) up to 2 years and correlated the results with the level of antibodies. Results: The anti-PAR 1 antibody level was lower in membranous nephropathy and FSGS compared to the control group. Anti-PAR 1 antibody levels were higher in secondary compared to primary glomerulonephritis. Both anti-PAR 1 and anti-ACE 2 antibody levels correlated positively (in focal and segmental glomerulosclerosis) or negatively (in lupus nephritis) with total protein and albumin at different time points of observation. Anti-PAR 1 and anti-ACE 2 antibody levels correlated also with creatinine level at one time point of observation in IgA nephropathy. Anti-PAR 1 and anti-ACE 2 antibodies correlated with each other in membranous nephropathy, FSGS, and p- and c-ANCA vasculitis (p < 0.05). Conclusions: The anti-PAR 1 antibody level was lower in membranous nephropathy and focal and segmental glomerulosclerosis compared to the control group. Anti-PAR 1 antibody levels tend to be higher in secondary compared to primary glomerulonephritis. Full article
(This article belongs to the Special Issue Newer Developments in the Field of Glomerular Diseases)
Show Figures

Graphical abstract

25 pages, 28238 KiB  
Article
Analysis of Kallikrein 6, Acetyl-α-Tubulin, and Aquaporin 1 and 2 Expression Patterns During Normal Human Nephrogenesis and in Congenital Anomalies of the Kidney and Urinary Tract (CAKUT)
by Nela Kelam, Marin Ogorevc, Ivona Gotovac, Ivana Kuzmić Prusac, Katarina Vukojević, Mirna Saraga-Babić and Snježana Mardešić
Genes 2025, 16(5), 499; https://doi.org/10.3390/genes16050499 - 27 Apr 2025
Viewed by 498
Abstract
Background/Objectives: The human kallikrein-related peptidase 6 (KLK6), a serine protease with trypsin-like properties, belongs to the 15-member kallikrein (KLK) gene family and is predominantly recognized for its role in oncogenesis, neurodegenerative disorders, and skin conditions. Aquaporins (AQPs) are integral membrane [...] Read more.
Background/Objectives: The human kallikrein-related peptidase 6 (KLK6), a serine protease with trypsin-like properties, belongs to the 15-member kallikrein (KLK) gene family and is predominantly recognized for its role in oncogenesis, neurodegenerative disorders, and skin conditions. Aquaporins (AQPs) are integral membrane proteins that facilitate water transport across cell membranes. AQP1 is constitutively active in the kidneys and plays a crucial role in reabsorbing filtered water, while AQP2 is regulated by vasopressin and is essential for maintaining body fluid homeostasis. The primary objective of the present study is to investigate the spatio-temporal expression patterns of KLK6, AQP1, and AQP2 throughout normal human nephrogenesis and congenital kidney and urinary tract (CAKUT) abnormalities: duplex kidneys, horseshoe kidneys, and dysplastic kidneys. Methods: An immunofluorescence analysis of KLK6, AQP1, and AQP2 was performed on 37 paraffin-embedded fetal kidney samples. The area percentage of KLK6 in the kidney cortex was calculated in normal developing samples during developmental phases 2, 3, and 4 and compared with CAKUT samples. Results: KLK6 exhibits distinct spatiotemporal expression patterns during human kidney development, with consistent localization in proximal tubules. Its subcellular positioning shifts from the basolateral cytoplasm in early phases to the apical cytoplasm in later stages, which may be strategically positioned to act on its substrate in either the peritubular space or the tubular fluid. KLK6 expression followed a quadratic trajectory, peaking at Ph4. This marked increase in the final developmental phase aligns with its strong expression in mature kidneys, suggesting a potential role in proximal tubule differentiation and functional maturation through facilitating extracellular matrix remodeling and activating proteinase-activated receptors, modulating the signaling pathways that are essential for tubular development. In duplex kidneys, structural abnormalities such as ureteral obstruction and hydronephrosis may upregulate KLK6 as part of a reparative response, while its downregulation could impair epithelial remodeling and cytoskeletal integrity, exacerbating dysplastic phenotypes. Conclusions: These findings highlight the potential of KLK6 involvement in normal kidney development and the pathology of CAKUT. Full article
(This article belongs to the Section Human Genomics and Genetic Diseases)
Show Figures

Figure 1

18 pages, 641 KiB  
Systematic Review
Identifying Molecular Probes for Fluorescence-Guided Surgery in Neuroblastoma: A Systematic Review
by Megan Hennessy, Jonathan J. Neville, Laura Privitera, Adam Sedgwick, John Anderson and Stefano Giuliani
Children 2025, 12(5), 550; https://doi.org/10.3390/children12050550 - 24 Apr 2025
Viewed by 721
Abstract
Background/Objectives: Targeted and non-targeted fluorescent molecular probes (FMPs) can be used intra-operatively to visualise tumour tissue. Multiple probes have been clinically approved for fluorescence-guided surgery (FGS) in adult oncology, and the translation of these technologies to paediatric neuroblastoma may provide novel strategies [...] Read more.
Background/Objectives: Targeted and non-targeted fluorescent molecular probes (FMPs) can be used intra-operatively to visualise tumour tissue. Multiple probes have been clinically approved for fluorescence-guided surgery (FGS) in adult oncology, and the translation of these technologies to paediatric neuroblastoma may provide novel strategies for optimising tumour resection whilst minimising morbidity. We aimed to identify clinically approved FMPs with potential utility for FGS in neuroblastoma. Methods: A systematic review of the literature was performed in accordance with the PRISMA guidelines (PROSPERO CRD42024541623). PubMed and Web of Science databases were searched to identify studies investigating clinically approved FGS probes and/or their targets in the context of neuroblastoma. Pre-clinical and clinical studies looking at human neuroblastoma were included. The primary outcomes were that the FGS probe was tested in patients with neuroblastoma, the probe selectively accumulated in neuroblastoma tissue, or that the target of the probe was selectively over-expressed in neuroblastoma tissue. Results: Forty-two studies were included. Four were clinical studies, and the remainder were pre-clinical studies using human neuroblastoma cell lines, human tumour tissue, or xenograft models using human neuroblastoma cells. The only FMP clinically evaluated in neuroblastoma is indocyanine green (ICG). FMP targets that have been investigated in neuroblastoma include poly-ADP ribose polymerase (PARP) (targeted by PARPiFL), endothelial growth factor receptor (EGFR) (targeted by Panitumumab-IRDye800CW, Cetuximab-IRDye800CW, Nimotuzumab-IRDye800CW and QRHKPRE-Cy5), vascular endothelial growth factor receptor (VEGFR) (targeted by Bevacizumab IRDye800CW), and proteases such as cathepsins and matrix metalloproteinases that activate the fluorescent signal of FMPs, such as LUM015 and AVB-620. Of the clinical studies included, all were found to have a high risk of bias. Conclusions: ICG is the only clinically approved fluorescent dye currently used for FGS in neuroblastoma; however, studies suggest that its ability to recognise neuroblastoma tissue is inconsistent. There are several clinically approved FMPs, or FMPs in clinical trials, that are used in adult oncology surgery that have targets expressed in neuroblastoma. Further research should validate these probes in neuroblastoma to enable their rapid translation into clinical practice. Full article
(This article belongs to the Section Pediatric Surgery)
Show Figures

Figure 1

12 pages, 3376 KiB  
Article
Role of Liver Kinase 1B in Platelet Activation and Host Defense During Klebsiella pneumoniae-Induced Pneumosepsis
by Osoul Chouchane, Valentine Léopold, Christine C. A. van Linge, Alex F. de Vos, Joris J. T. H. Roelofs, Cornelis van ‘t Veer and Tom van der Poll
Int. J. Mol. Sci. 2025, 26(8), 3714; https://doi.org/10.3390/ijms26083714 - 14 Apr 2025
Viewed by 593
Abstract
Pneumonia is the most common cause of sepsis, with Klebsiella pneumoniae frequently implicated as a causative pathogen. Platelets play a crucial role in host defense during sepsis, and their activation is essential for effective immune responses, which is at least in part induced [...] Read more.
Pneumonia is the most common cause of sepsis, with Klebsiella pneumoniae frequently implicated as a causative pathogen. Platelets play a crucial role in host defense during sepsis, and their activation is essential for effective immune responses, which is at least in part induced through activation of the collagen receptor glycoprotein (GP)VI. Platelets require energy for their activation, and Liver kinase B1 (LKB1) is a key regulator of energy metabolism. We sought to determine the role of LKB1 in platelet function and host response during K. pneumoniae-induced pneumosepsis. Platelet-specific-Lkb1-deficient mice were generated and compared to control littermates. Platelet counts were unaffected by Lkb1 deficiency in naïve mice. However, Lkb1-deficient platelets exhibited significant hyperreactivity to GPVI stimulation, an effect not observed after stimulation of the thrombin receptor protease-activated receptor 4. During K. pneumoniae infection, platelets of both Lkb1-deficient and control mice became equally hyporesponsive to GPVI stimulation, without differences between genotypes. Platelet-specific Lkb1 deficiency did not alter bacterial outgrowth or dissemination, inflammatory responses, or lung pathology. These findings suggest that while Lkb1 plays a role in regulating platelet activation in response to GPVI stimulation, it does not significantly impact platelet activation or the host response during pneumonia-induced sepsis. Full article
Show Figures

Figure 1

11 pages, 3525 KiB  
Article
Wound Healing in Human Skin Equivalents Reconstructed with Biopolymers Under Fine-Dust Exposure
by Taeeun Kim, Junwoo Lim, Jaehyun Jeong and Heewook Ryu
Polymers 2025, 17(7), 901; https://doi.org/10.3390/polym17070901 - 27 Mar 2025
Viewed by 894
Abstract
Airborne fine-dust pollution poses a significant threat to both respiratory and skin health; however, the skin’s wound-healing process in response to such exposure remains underexplored. Therefore, this study examined the effect of fine-dust-model compounds, specifically polycyclic aromatic hydrocarbons (PM10-PAHs) and trace-metal-containing [...] Read more.
Airborne fine-dust pollution poses a significant threat to both respiratory and skin health; however, the skin’s wound-healing process in response to such exposure remains underexplored. Therefore, this study examined the effect of fine-dust-model compounds, specifically polycyclic aromatic hydrocarbons (PM10-PAHs) and trace-metal-containing particles (PM10-Trace), on the wound-healing process using human skin equivalents reconstructed with collagen-based biomaterials and human skin cells. Our findings revealed that fine-dust exposure significantly delayed wound closure by 2–3 times compared with unexposed controls, impairing re-epithelialization. Live imaging of wound-healing dynamics revealed that trace-metal-containing particles had a more pronounced inhibitory effect than polycyclic aromatic hydrocarbons. Furthermore, fine-dust exposure elevated protease-activated receptor-1 (PAR1) expression by up to 161%, indicating significant physiological disruption. Additionally, fine-dust exposure triggered inflammation and oxidative stress, leading to structural and functional damage in the reconstructed skin. These results provide critical insights into how airborne pollutants disrupt skin repair mechanisms and highlight the need for targeted strategies to mitigate their harmful effects. Full article
(This article belongs to the Special Issue Biomedical Polymer Materials for Wound Healing)
Show Figures

Figure 1

Back to TopTop