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

Article Types

Countries / Regions

remove_circle_outline
remove_circle_outline
remove_circle_outline

Search Results (736)

Search Parameters:
Keywords = protein phosphatase inhibition

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
30 pages, 9213 KiB  
Article
Resveratrol Impairs Insulin Signaling in Hepatic Cells via Activation of PKC and PTP1B Pathways
by Karla D. Hernández-González, Monica A. Vinchira-Lamprea, Judith Hernandez-Aranda and J. Alberto Olivares-Reyes
Int. J. Mol. Sci. 2025, 26(15), 7434; https://doi.org/10.3390/ijms26157434 - 1 Aug 2025
Viewed by 363
Abstract
Resveratrol (RSV), a polyphenol found in a variety of berries and wines, is known for its anti-inflammatory, anticancer, and antioxidant properties. It has been suggested that RSV may play a role in the regulation of metabolic disorders, including diabetes and insulin resistance. However, [...] Read more.
Resveratrol (RSV), a polyphenol found in a variety of berries and wines, is known for its anti-inflammatory, anticancer, and antioxidant properties. It has been suggested that RSV may play a role in the regulation of metabolic disorders, including diabetes and insulin resistance. However, in recent years, it has been reported to completely inhibit Akt kinase function in liver cells. Akt is a central protein involved in the metabolic function of insulin and is regulated by the phosphatidylinositol-3-kinase (PI3K) pathway. In this study, we examined the effect of RSV on insulin-induced insulin receptor (IR) phosphorylation and proteins involved in the PI3K/Akt pathway in a hepatic cell model, clone 9 (C9), and in hepatoma cells, Hepa 1-6 (H1-6). In both cell lines, RSV inhibited tyrosine phosphorylation of IR and insulin-induced activation of Akt. We also evaluated the effect of RSV on the activation of protein tyrosine phosphatase 1B (PTP1B), which is associated with IR dephosphorylation, and found that RSV increased PTP1B-Tyr152 phosphorylation in a time- and concentration-dependent manner. Furthermore, we found that the protein kinase C (PKC) inhibitors BIM and Gö6976 prevented the inhibition of Akt phosphorylation by RSV and increased the phosphorylation of Ser/Thr residues in IR, suggesting that PKC is involved in the inhibition of the insulin pathway by RSV. Thus, classical PKC isoforms impair the PI3K/Akt pathway at the IR and GSK3 and GS downstream levels; however, IRS-Tyr632 phosphorylation remains unaffected. These results suggest that RSV can lead to insulin resistance by activating PTP1B and PKC, consequently affecting glucose homeostasis in hepatic cells. Full article
(This article belongs to the Special Issue The Molecular and Cellular Aspects of Insulin Resistance)
Show Figures

Figure 1

13 pages, 1600 KiB  
Article
LIMK2-1 Is a Phosphorylation-Dependent Inhibitor of Protein Phosphatase-1 Catalytic Subunit and Myosin Phosphatase Holoenzyme
by Andrea Kiss, Emese Tóth, Zsófia Bodogán, Mohamad Mahfood, Zoltán Kónya and Ferenc Erdődi
Int. J. Mol. Sci. 2025, 26(15), 7347; https://doi.org/10.3390/ijms26157347 - 30 Jul 2025
Viewed by 159
Abstract
The C-kinase-activated protein phosphatase-1 (PP1) inhibitor of 17 kDa (CPI-17) is a specific inhibitor of the PP1 catalytic subunit (PP1c) and the myosin phosphatase (MP) holoenzyme. CPI-17 requires the phosphorylation of Thr38 in the peptide segment 35ARV(P)TVKYDRREL46 for inhibitory activity. CPI-17 [...] Read more.
The C-kinase-activated protein phosphatase-1 (PP1) inhibitor of 17 kDa (CPI-17) is a specific inhibitor of the PP1 catalytic subunit (PP1c) and the myosin phosphatase (MP) holoenzyme. CPI-17 requires the phosphorylation of Thr38 in the peptide segment 35ARV(P)TVKYDRREL46 for inhibitory activity. CPI-17 regulates myosin phosphorylation in smooth muscle contraction and the tumorigenic transformation of several cell lines via the inhibition of MP. A phosphospecific antibody (anti-CPI-17pThr38) against the phosphorylation peptide was used to determine the phosphorylation levels in cells. We found that phospho-CPI-17 and its closely related proteins are not present in HeLa and MCF7 cells after inducing phosphorylation by inhibiting phosphatases with calyculin A. In contrast, cross-reactions of proteins in the 40–220 kDa range with anti-CPI-17pThr38 were apparent. Searching the protein database for similarities to the CPI-17 phosphorylation sequence revealed several proteins with 42–75% sequence identities. The LIMK2-1 isoform emerged as a possible PP1 inhibitor. Experiments with Flag-LIMK2-1 overexpressed in tsA201 cells proved that LIMK2-1 interacts with PP1c isoforms and is phosphorylated predominantly by protein kinase C. Phosphorylated LIMK2-1 inhibits PP1c and the MP holoenzyme with similar potencies (IC50 ~28–47 nM). In conclusion, our results suggest that LIMK2-1 is a novel phosphorylation-dependent inhibitor of PP1c and MP and may function as a CPI-17-like phosphatase inhibitor in cells where CPI-17 is present but not phosphorylated upon phosphatase inhibition. Full article
(This article belongs to the Special Issue 25th Anniversary of IJMS: Updates and Advances in Macromolecules)
Show Figures

Figure 1

21 pages, 8891 KiB  
Article
Urolithin A Attenuates Periodontitis in Mice via Dual Anti-Inflammatory and Osteoclastogenesis Inhibition: A Natural Metabolite-Based Therapeutic Strategy
by Yishu Xia, Danni Wu, Linyi Zhou, Xinyu Wu and Jianzhi Chen
Molecules 2025, 30(13), 2881; https://doi.org/10.3390/molecules30132881 - 7 Jul 2025
Viewed by 391
Abstract
Periodontitis is an inflammatory disease that affects the periodontal supporting tissues. Its cardinal clinical manifestations encompass gingival inflammation, periodontal pocket formation, and alveolar bone resorption. Urolithin A (UA), a gut microbiota-derived metabolite of ellagitannins, is known for its anti-inflammatory and osseous-protective properties. Nonetheless, [...] Read more.
Periodontitis is an inflammatory disease that affects the periodontal supporting tissues. Its cardinal clinical manifestations encompass gingival inflammation, periodontal pocket formation, and alveolar bone resorption. Urolithin A (UA), a gut microbiota-derived metabolite of ellagitannins, is known for its anti-inflammatory and osseous-protective properties. Nonetheless, the impact of UA on periodontitis remains unknown. To investigate the preventive effect of UA, we employed a lipopolysaccharide (LPS)-induced inflammation model in RAW 264.7 mouse macrophages, a receptor activator of nuclear factor-κB ligand (RANKL)-induced osteoclast differentiation model, and a ligature-induced periodontitis model in mice. The expression of inflammatory factors (tumor necrosis factor-α, TNF-α; interleukin-6, IL-6) was analyzed to assess anti-inflammatory efficacy. Bone loss in mice with periodontitis was assessed through histological and imaging techniques, including haematoxylin and eosin staining to evaluate alveolar bone morphology, Masson’s trichrome staining to visualize collagen fiber distribution, and micro-computed tomography scanning to quantify bone structural parameters. Additionally, we investigated the underlying mechanisms by examining osteoclast activity through tartrate-resistant acid phosphatase staining and the expression levels of proteins RANKL and osteoprotegerin (OPG). We found that UA reduced IL-6 and TNF-α levels in vitro and in vivo, inhibited osteoclast differentiation, and decreased the RANKL/OPG ratio in periodontitis mice. Full article
(This article belongs to the Section Medicinal Chemistry)
Show Figures

Figure 1

49 pages, 5285 KiB  
Review
Insights into Natural Products from Marine-Derived Fungi with Antimycobacterial Properties: Opportunities and Challenges
by Muhammad Azhari, Novi Merliani, Marlia Singgih, Masayoshi Arai and Elin Julianti
Mar. Drugs 2025, 23(7), 279; https://doi.org/10.3390/md23070279 - 3 Jul 2025
Viewed by 704
Abstract
Tuberculosis (TB) poses a persistent global health threat exacerbated by the emergence of drug-resistant strains; hence, there is a continuous quest for novel antimicrobial agents. Despite efforts to develop effective therapies, existing treatments require a relatively long duration of therapy to eradicate the [...] Read more.
Tuberculosis (TB) poses a persistent global health threat exacerbated by the emergence of drug-resistant strains; hence, there is a continuous quest for novel antimicrobial agents. Despite efforts to develop effective therapies, existing treatments require a relatively long duration of therapy to eradicate the pathogen due to its virulence factors, pathogenesis patterns, and ability to enter dormant states. This can lead to a higher risk of treatment failure due to poor patient adherence to the complex regimen. As a result, considerable research is necessary to identify alternative antituberculosis agents. The marine environment, particularly marine-derived fungi, has recently gained interest due to its potential as an abundant source of bioactive natural products. This review covers 19 genera of marine-derived fungi and 139 metabolites, 131 of which exhibit antimycobacterial activity. The integrated dataset pinpoints the fungal genera and chemical classes that most frequently yield potent antimycobacterial hits while simultaneously exposing critical gaps, such as the minimal evaluation of compounds against dormant bacilli and the presence of underexplored ecological niches and fungal genera. Several compounds exhibit potent activity through uncommon mechanisms, including the inhibition of mycobacterial protein tyrosine phosphatases (MptpB/MptpA), protein kinase PknG, ATP synthase and the disruption of mycobacterial DNA via G-quadruplex stabilization. Structure–activity relationship (SAR) trends are highlighted for the most potent agents, illuminating how specific functional groups underpin target engagement and potency. This review also briefly proposes a dereplication strategy and approaches for toxicity mitigation in the exploration of marine-derived fungi’s natural products. Through this analysis, we offer insights into the potency and challenges of marine-derived fungi’s natural products as hit compounds or scaffolds for further antimycobacterial research. Full article
Show Figures

Figure 1

24 pages, 2487 KiB  
Review
Targeting WEE1 Kinase for Breast Cancer Therapeutics: An Update
by Zhao Zhang, Ritika Harish, Naveed Elahi, Sawanjit Saini, Aamir Telia, Manjit Kundlas, Allexes Koroleva, Israel N. Umoh, Manpreet Lota, Meha Bilkhu, Aladdin Kawaiah, Manogna R. Allala, Armelle Leukeu, Emmanuel Nebuwa, Nadiya Sharifi, Anthony W. Ashton, Xuanmao Jiao and Richard G. Pestell
Int. J. Mol. Sci. 2025, 26(12), 5701; https://doi.org/10.3390/ijms26125701 - 13 Jun 2025
Viewed by 1664
Abstract
WEE1 kinase is a crucial cell cycle regulatory protein that controls the timing of mitotic entry. WEE1, via inhibition of Cyclin-dependent Kinase 1 (CDK1) and Cyclin-dependent Kinase 2 (CDK2), governs the G2-M checkpoint by inhibiting entry into mitosis. The state of balance between [...] Read more.
WEE1 kinase is a crucial cell cycle regulatory protein that controls the timing of mitotic entry. WEE1, via inhibition of Cyclin-dependent Kinase 1 (CDK1) and Cyclin-dependent Kinase 2 (CDK2), governs the G2-M checkpoint by inhibiting entry into mitosis. The state of balance between WEE family kinases and CDC25C phosphatases restricts CDK1/CycB activity. The WEE kinase family consists of WEE1, PKMYT1, and WEE2 (WEE1B). WEE1 and PKMYT1 regulate entry into mitosis during cell cycle progression, whereas WEE2 governs cell cycle progression during meiosis. Recent studies have identified WEE1 as a potential therapeutic target in several cancers, including therapy-resistant triple-negative breast cancer. Adavosertib’s clinical promise was challenged by inter-individual variations in response and side effects. Because of these promising preclinical outcomes, other WEE1 kinase inhibitors (Azenosertib, SC0191, IMP7068, PD0407824, PD0166285, WEE1-IN-5, Zedoresertib, WEE1-IN-8, and ATRN-1051) are being developed, with several currently being evaluated in clinical trials or as an adjuvant to chemotherapies. Preclinical studies show WEE1 inhibitors induce MHC class 1 antigens and STING when given as combination therapies, suggesting potential additional therapeutic opportunities. Reliable predictors of clinical responses based on mechanistic insights remain an important unmet need. Herein, we review the role of WEE1 inhibition therapy in breast cancer. Full article
(This article belongs to the Special Issue Molecular Research and Treatment of Breast Cancer: 3rd Edition)
Show Figures

Figure 1

21 pages, 1079 KiB  
Article
Toxicological Responses of Juvenile Gilthead Seabream to Enniatin B and Fumonisin B1
by Flávia V. Mello, Cheila Pereira, Busenur Özkan, Ana Luísa Maulvault, Florbela Soares, Pedro Pousão-Ferreira, José O. Fernandes, Sara C. Cunha, António Marques and Patrícia Anacleto
Int. J. Mol. Sci. 2025, 26(12), 5676; https://doi.org/10.3390/ijms26125676 - 13 Jun 2025
Viewed by 590
Abstract
The replacement of ingredients from animal sources with plant-based ingredients is increasing the risk of contamination by mycotoxins in aquafeeds, potentially causing detrimental effects on fish welfare. However, limited research has been carried out so far on the impact of mycotoxins on fish [...] Read more.
The replacement of ingredients from animal sources with plant-based ingredients is increasing the risk of contamination by mycotoxins in aquafeeds, potentially causing detrimental effects on fish welfare. However, limited research has been carried out so far on the impact of mycotoxins on fish health. Hence, the aim of this study was to assess the toxicological effects of the dietary emerging (enniatin B, ENNB) and regulated (fumonisin B1, FB1) mycotoxins (150 µg/kg) in different tissues of juvenile gilthead seabream (Sparus aurata) after 28 days of dietary exposure. Fitness indexes, plasma metabolites, and biomarkers of oxidative stress, metabolism, cellular, and neurotoxic damage were assessed. The exposure to each mycotoxin was sufficient to cause distinct effects in fish tissues. ENNB appears to be the most harmful mycotoxin to S. aurata, inducing changes on alkaline phosphatase and lipase activities in plasma, as well as protein and lipid degradation in liver. Increased lipid degradation was also induced in the brain by FB1 alone or combined with ENNB, whereas the exposure to the mixture inhibited acetylcholinesterase activity. Overall, this study contributes by highlighting the toxicological attributes of ENNB, thus reinforcing the need to include this mycotoxin in future legislation. Full article
(This article belongs to the Special Issue Toxicity Mechanism of Emerging Pollutants: 2nd Edition)
Show Figures

Figure 1

20 pages, 7854 KiB  
Article
Shear Stress Regulates Osteogenic Differentiation of Human Dental Pulp Stem Cells via the p38 Pathway
by Hnin Yu Lwin, Watcharaphol Tiskratok, Maythwe Kyawsoewin, Jeeranan Manokawinchoke, Chutimon Termkwanchareon, Nuttapol Limjeerajarus, Chalida Nakalekha Limjeerajarus, Hiroshi Egusa, Thanaphum Osathanon and Phoonsuk Limraksasin
Int. J. Mol. Sci. 2025, 26(12), 5667; https://doi.org/10.3390/ijms26125667 - 13 Jun 2025
Viewed by 798
Abstract
This study aimed to investigate the effects of shear stress on osteogenic differentiation of human dental pulp stem cells (hDPSCs). The hDPSCs were subjected to shear stress for 24 h before osteogenic induction for 21 days. The mRNA expression of osteogenic markers such [...] Read more.
This study aimed to investigate the effects of shear stress on osteogenic differentiation of human dental pulp stem cells (hDPSCs). The hDPSCs were subjected to shear stress for 24 h before osteogenic induction for 21 days. The mRNA expression of osteogenic markers such as RUNX2, OSX, ALP, COL1A1, OCN, and OPN was evaluated by real-time RT-PCR. Alkaline Phosphatase (ALP) activity and Alizarin Red S (ARS) staining were investigated to confirm osteogenic differentiation and mineralization of hDPSCs, respectively. The protein expression of osterix was shown by immunofluorescence staining and Western blotting. RNA sequencing was performed to investigate how shear stress affects the osteogenic differentiation of hDPSCs, which was validated through p38 inhibitor (SB203580) treatment. Real-time RT-PCR revealed that shear stress enhanced osteogenic marker-gene expression. The increased osterix protein expression was detected on Day 14 in the shear-stress loading group compared to the static group. Shear stress enhanced ALP activity and mineralization, observed on Days 14 and 21. A volcano plot exhibited up- and downregulated genes, while the p38 inhibitor markedly inhibited osteogenic differentiation of hDPSCs triggered by shear stress. In conclusion, shear stress promotes the osteogenic differentiation of hDPSCs through the p38 mitogen-activated protein kinase signaling pathway. Full article
(This article belongs to the Special Issue Molecular Mechanisms of Bone Injury and Bone Tissue Regeneration)
Show Figures

Graphical abstract

14 pages, 1413 KiB  
Review
Advances in the Exploration of Coordination Complexes of Vanadium in the Realm of Alzheimer’s Disease: A Mini Review
by Jesús Antonio Cruz-Navarro, Luis Humberto Delgado-Rangel, Ricardo Malpica-Calderón, Arturo T. Sánchez-Mora, Hugo Ponce-Bolaños, Andrés Felipe González-Oñate, Jorge Alí-Torres, Raúl Colorado-Peralta, Daniel Canseco-Gonzalez, Viviana Reyes-Márquez and David Morales-Morales
Molecules 2025, 30(12), 2547; https://doi.org/10.3390/molecules30122547 - 11 Jun 2025
Viewed by 587
Abstract
Alzheimer’s disease (AD) is a progressive neurodegenerative disorder characterized by cognitive decline, memory loss and limited therapeutic options. Metal-based drugs have emerged as promising alternatives in the search for effective treatments, and vanadium coordination complexes have shown significant potential due to their neuroprotective [...] Read more.
Alzheimer’s disease (AD) is a progressive neurodegenerative disorder characterized by cognitive decline, memory loss and limited therapeutic options. Metal-based drugs have emerged as promising alternatives in the search for effective treatments, and vanadium coordination complexes have shown significant potential due to their neuroprotective and anti-aggregant properties. This review explores the advances in the development of vanadium-based metallodrugs for AD, focusing on their ability to modulate amyloid-beta (Aβ) aggregation, oxidative stress, and neuroinflammation. Recent in vitro and in vivo studies highlight the efficacy of oxovanadium (IV) and peroxovanadium (V) complexes in inhibiting Aβ fibril formation and reducing neuronal toxicity. Additionally, the interaction of vanadium complexes with key biological targets, such as peroxisome proliferator-activated receptor gamma (PPARγ) and protein-tyrosine phosphatase 1B (PTP1B), suggests a multifaceted therapeutic approach. While these findings underscore the potential of vanadium compounds as innovative treatments for AD, further research is needed to optimize their bioavailability, selectivity, and safety for clinical applications. Full article
Show Figures

Graphical abstract

14 pages, 2678 KiB  
Article
Doping of Hollow Urchin-like MnO2 Nanoparticles in Beta-Tricalcium Phosphate Scaffold Promotes Stem Cell Osteogenic Differentiation
by Enze Qian, Ahmed Eltawila and Yunqing Kang
Int. J. Mol. Sci. 2025, 26(11), 5092; https://doi.org/10.3390/ijms26115092 - 26 May 2025
Viewed by 367
Abstract
Effective osteogenesis for bone regeneration is still considerably challenging for a porous β-tricalcium phosphate (β-TCP) scaffold to achieve. To overcome this challenge, hollow manganese dioxide (H-MnO2) nanoparticles with an urchin-like shell structure were prepared and added in the porous β-TCP scaffold. [...] Read more.
Effective osteogenesis for bone regeneration is still considerably challenging for a porous β-tricalcium phosphate (β-TCP) scaffold to achieve. To overcome this challenge, hollow manganese dioxide (H-MnO2) nanoparticles with an urchin-like shell structure were prepared and added in the porous β-TCP scaffold. A template-casting method was used to prepare the porous H-MnO2/β-TCP scaffolds. As a control, solid manganese dioxide (S-MnO2) nanoparticles were also added into β-TCP scaffolds. Human bone mesenchymal stem cells (hBMSC) were seeded in the porous scaffolds and characterized through cell viability assay and alkaline phosphatase (ALP) assay. Results from in vitro protein loading and releasing experiments showed that H-MnO2 can load significantly higher proteins and release more proteins compared to S-MnO2 nanoparticles. When they were doped into β-TCP, MnO2 nanoparticles did not significantly change the surface wettability and mechanical properties of porous β-TCP scaffolds. In vitro cell viability results showed that MnO2 nanoparticles promoted cell proliferation in a low dose, but inhibited cell growth when the added concentration went beyond 0.5%. At a range of lower than 0.5%, H-MnO2 doped β-TCP scaffolds promoted the early osteogenesis of hBMSCs. These results suggested that H-MnO2 in the porous β-TCP scaffold has promising potential to stimulate osteogenesis. More studies would be performed to demonstrate the other functions of urchin-like H-MnO2 nanoparticles in the porous β-TCP. Full article
Show Figures

Figure 1

19 pages, 15026 KiB  
Article
Proteomics-Based Exploration of the Hepatoprotective Mechanism of α-Lipoic Acid in Rats with Iron Overload-Induced Liver Injury
by Shuxia Jiang, Yujia Shu, Shihui Guo, Yingdong Ni, Ruqian Zhao, Hongli Shan and Wenqiang Ma
Int. J. Mol. Sci. 2025, 26(10), 4774; https://doi.org/10.3390/ijms26104774 - 16 May 2025
Viewed by 595
Abstract
Excessive iron accumulation poses a significant threat to liver health, primarily through oxidative stress and autophagy dysregulation. α-Lipoic acid (ALA), a natural antioxidant with hepatoprotective properties, may alleviate iron-induced liver damage, but its underlying mechanisms are not fully understood. This study utilized male [...] Read more.
Excessive iron accumulation poses a significant threat to liver health, primarily through oxidative stress and autophagy dysregulation. α-Lipoic acid (ALA), a natural antioxidant with hepatoprotective properties, may alleviate iron-induced liver damage, but its underlying mechanisms are not fully understood. This study utilized male Sprague Dawley rats and BRL-3A cells to explore the protective effects of ALA against iron overload in vivo and in vitro, respectively. ALA treatment significantly reduced hepatic iron accumulation, improved liver morphology, and alleviated iron-induced ultrastructural damage in rats. ALA also improved liver function markers in plasma, including alkaline phosphatase (ALP), gamma-glutamyltransferase (GGT), total bilirubin (TBIL), and the AST/ALT ratio. Furthermore, ALA mitigated iron-induced oxidative stress by lowering hepatic reactive oxygen species (ROS) and malondialdehyde (MDA), while increasing the antioxidant enzyme activities of glutathione peroxidase (GSH-Px) and catalase (CAT). In BRL-3A cells, ALA improved cell viability, decreased intracellular ROS, and reduced iron levels. Proteomics analysis indicates that NAD(P)H: quinone oxidoreductase 1 (NQO1) may play a critical role in the protective effects of ALA against iron overload-induced hepatic damage in rats. Mechanistically, ALA upregulated NQO1 expression while downregulating autophagy-related proteins, including light chain 3B (LC3B), lysosomal-associated membrane protein 1 (LAMP1), and cathepsin D (CTSD). Inhibition or knockdown of NQO1 abolished ALA’s protective effects, confirming its role in reducing oxidative stress and excessive autophagy. These findings highlight the potential of ALA as a therapeutic agent for managing hepatic iron toxicity through iron chelation and activation of NQO1. Full article
(This article belongs to the Special Issue New Advances in Proteomics in Disease)
Show Figures

Figure 1

19 pages, 2190 KiB  
Article
Evaluation of the Expression of IDO and PTEN in Human Kidney Cancer
by Gábor Kónya, Zsuzsanna Szabó, Nikoletta Dobos, József Király, Krisztián Szegedi, Anna Vass, Ákos Steli, Csaba Szász, Balázs Dezső, Barbara Zsebik and Gábor Halmos
Curr. Issues Mol. Biol. 2025, 47(5), 359; https://doi.org/10.3390/cimb47050359 - 13 May 2025
Viewed by 764
Abstract
Immunotherapy has become one of the primary forms of cancer treatment. The inhibition of immune checkpoint molecules, including indoleamine 2,3-dioxygenase (IDO), is a promising approach for immunotherapy. Phosphatase and tensin homolog (PTEN) is well known as a tumor suppressor that antagonizes oncogenic signaling [...] Read more.
Immunotherapy has become one of the primary forms of cancer treatment. The inhibition of immune checkpoint molecules, including indoleamine 2,3-dioxygenase (IDO), is a promising approach for immunotherapy. Phosphatase and tensin homolog (PTEN) is well known as a tumor suppressor that antagonizes oncogenic signaling molecules/pathways and plays a key role in the prognosis and (immuno)therapy of the disease. In this study, twenty healthy and tumorous renal tissue pairs were investigated, and the mRNA (RT-qPCR) and protein (Western blot) expression of IDO and PTEN were analyzed. In two cancer cell lines (CAKI-2; A-498), the protein of IDO and PTEN was measured followed by IDO induction with interferon alpha-2 (IFN-α2). According to our results, a significantly higher mRNA expression of IDO and PTEN was found in tumorous tissues compared to the adjacent healthy kidney specimens. The mRNA expression of IDO and PTEN showed a positive correlation in 80% of the sample pairs. Western blot results confirmed the protein expression of both IDO and PTEN. In the cell lines, immunocytochemistry showed that IDO is inducible with IFN-α2. In summary, our results suggest that IDO expression may play a role in the development of renal cancer, and IDO as well as PTEN might be potential biomarkers for patients with RCC. Full article
Show Figures

Figure 1

12 pages, 1743 KiB  
Article
Cell-Penetrating Peptide Based on Myosin Phosphatase Target Subunit Sequence Mediates Myosin Phosphatase Activity
by Andrea Kiss, Mohamad Mahfood, Zsófia Bodogán, Zoltán Kónya, Bálint Bécsi and Ferenc Erdődi
Biomolecules 2025, 15(5), 705; https://doi.org/10.3390/biom15050705 - 12 May 2025
Cited by 1 | Viewed by 492
Abstract
Myosin phosphatase (MP) holoenzyme consists of protein phosphatase-1 (PP1) catalytic subunit (PP1c) associated with myosin phosphatase target subunit-1 (MYPT1) and it plays an important role in mediating the phosphorylation of the 20 kDa light chain (MLC20) of myosin, thereby regulating cell contractility. The [...] Read more.
Myosin phosphatase (MP) holoenzyme consists of protein phosphatase-1 (PP1) catalytic subunit (PP1c) associated with myosin phosphatase target subunit-1 (MYPT1) and it plays an important role in mediating the phosphorylation of the 20 kDa light chain (MLC20) of myosin, thereby regulating cell contractility. The association of MYPT1 with PP1c increases the phosphatase activity toward myosin; therefore, disrupting/dissociating this interaction may result in inhibition of the dephosphorylation of myosin. In this study, we probed how MYPT132–58 peptide including major PP1c interactive regions coupled with biotin and cell-penetrating TAT sequence (biotin-TAT-MYPT1) may influence MP activity. Biotin-TAT-MYPT1 inhibited the activity of MP holoenzyme and affinity chromatography as well as surface plasmon resonance (SPR) binding studies established its stable association with PP1c. Biotin-TAT-MYPT1 competed for binding to PP1c with immobilized GST-MYPT1 in SPR assays and it partially relieved PP1c inhibition by thiophosphorylated (on Thr696 and Thr853) MYPT1. Moreover, biotin-TAT-MYPT1 dissociated PP1c from immunoprecipitated PP1c-MYPT1 complex implying its holoenzyme disrupting ability. Biotin-TAT-MYPT1 penetrated into A7r5 smooth muscle cells localized in the cytoplasm and nucleus and exerted inhibition on MP with a parallel increase in MLC20 phosphorylation. Our results imply that the biotin-TAT-MYPT1 peptide may serve as a specific MP regulatory cell-penetrating peptide as well as possibly being applicable to further development for pharmacological interventions. Full article
(This article belongs to the Section Enzymology)
Show Figures

Figure 1

19 pages, 2458 KiB  
Article
Pan-Cancer Analysis Identifies a Ras-Related GTPase as a Potential Modulator of Cancer
by Hsiang-Yin Hsueh, Kristyn Gumpper-Fedus, Jelmer W. Poelstra, Kenneth L. Pitter and Zobeida Cruz-Monserrate
Int. J. Mol. Sci. 2025, 26(9), 4419; https://doi.org/10.3390/ijms26094419 - 6 May 2025
Viewed by 760
Abstract
Ras signaling regulates many cellular processes in cancer development. While well-known Ras-related oncogenes, such as KRAS, have been extensively explored, the role of other Ras-related genes in cancer remains poorly studied. Dexamethasone-induced Ras-related protein 1 (RASD1), a member of the Ras superfamily, is [...] Read more.
Ras signaling regulates many cellular processes in cancer development. While well-known Ras-related oncogenes, such as KRAS, have been extensively explored, the role of other Ras-related genes in cancer remains poorly studied. Dexamethasone-induced Ras-related protein 1 (RASD1), a member of the Ras superfamily, is widely expressed across various tissues and is involved in inhibiting cell growth and inducing apoptosis, suggesting a potential role as a tumor suppressor. Here, we investigated RASD1 expression across multiple tissues and cancers, utilizing data from The Cancer Genome Atlas (TCGA), Human Protein Atlas, and Genotype-Tissue Expression (GTEx) databases. Our analysis revealed a significant downregulation of RASD1 mRNA expression in several cancer types compared to normal tissues, correlating with low levels of promoter methylation. Interestingly, high RASD1 expression correlated with a favorable prognosis in multiple cancers. Immune cell infiltration analysis indicated that elevated RASD1 expression is associated with an increased infiltration of CD4+ T cells and myeloid-derived dendritic cells in cancer. Furthermore, the expression of genes exhibiting similar expression patterns as RASD1 suggest that RASD1 may play a role in interleukin-4-mediated apoptosis and could regulate the transcription of the phosphatase and tensin homolog (PTEN) gene. Overall, these findings suggest that RASD1 may modulate immune signaling and tumor suppressive pathways. Full article
(This article belongs to the Section Molecular Oncology)
Show Figures

Figure 1

17 pages, 3066 KiB  
Article
Regulation of Pleiotrophin and PTPRZ1 Expression by Hypoxia to Restrict Hypoxia-Induced Cell Migration
by Evangelia Poimenidi, Eirini Droggiti, Katerina Karavasili, Dimitra Kotsirilou, Eleni Mourkogianni, Pieter Koolwijk and Evangelia Papadimitriou
Cancers 2025, 17(9), 1516; https://doi.org/10.3390/cancers17091516 - 30 Apr 2025
Viewed by 851
Abstract
Background/Objectives: In the tumor microenvironment, hypoxia regulates genes that support tumor cell invasion and angiogenesis under the control of the hypoxia-inducible transcription factors (HIFs). Pleiotrophin (PTN) is a secreted protein that activates cell migration in endothelial and cancer cells that express αν [...] Read more.
Background/Objectives: In the tumor microenvironment, hypoxia regulates genes that support tumor cell invasion and angiogenesis under the control of the hypoxia-inducible transcription factors (HIFs). Pleiotrophin (PTN) is a secreted protein that activates cell migration in endothelial and cancer cells that express ανβ3 integrin but has inhibitory effects in cells that do not express ανβ3 integrin. In both cases, the protein tyrosine phosphatase receptor zeta 1 (PTPRZ1) seems to mediate the effects of PTN. In the present work, we studied the effect of hypoxia on PTN and PTPRZ1 expression and the functional consequences of this effect. Methods: Western blot, quantitative real-time PCR, and luciferase assays were used to study the impact of hypoxia at the protein, mRNA, and transcriptional levels, respectively. Decoy oligonucleotides (ODNs), siRNA technology, and plasmid overexpression were used to study the involvement of the transcription factors studied. Functional assays were used to study the effect of hypoxia on cell proliferation and migration. Results: Hypoxia increases PTN expression through the transcriptional activation of the corresponding gene in ανβ3 integrin-expressing cells. The transcription factors HIF-1α, HIF-2α, and AP-1 mediate the up-regulation of PTN by hypoxia. Functional assays in endothelial cells from PTN knockout mice or endothelial and cancer cells following the downregulation of PTN expression showed that PTN negatively affects chemical hypoxia-induced cell proliferation and migration. In cancer cells that do not express ανβ3 integrin, hypoxia or chemical hypoxia inhibits PTN expression in a HIF-1α-, HIF-2α-, and AP-1-independent manner. The expression of PTPRZ1 is up-regulated by chemical hypoxia, is HIF-1α- and HIF-2α-dependent, and seems to limit the activation of HIF-1α, at least in endothelial cells. Conclusions: Hypoxia or chemical hypoxia regulates PTN and PTPRZ1 expressions to restrict the stimulatory effects of hypoxia on endothelial and cancer cell migration. Full article
(This article belongs to the Section Molecular Cancer Biology)
Show Figures

Figure 1

18 pages, 2980 KiB  
Article
The Wheat Intrinsically Disordered Protein TdRL1 Negatively Regulates the Type One Protein Phosphatase TdPP1
by Fatma Amor, Mariem Bradai, Ikram Zaidi, Vitor Amorim-Silva, Nabil Miled, Moez Hanin and Chantal Ebel
Biomolecules 2025, 15(5), 631; https://doi.org/10.3390/biom15050631 - 28 Apr 2025
Viewed by 478
Abstract
Type 1 protein phosphatases (PP1s) are crucial in various plant cellular processes. Their function is controlled by regulators known as PP1-interacting proteins (PIPs), often intrinsically disordered, such as Inhibitor 2 (I2), conserved across kingdoms. The durum wheat TdRL1 acts as a positive [...] Read more.
Type 1 protein phosphatases (PP1s) are crucial in various plant cellular processes. Their function is controlled by regulators known as PP1-interacting proteins (PIPs), often intrinsically disordered, such as Inhibitor 2 (I2), conserved across kingdoms. The durum wheat TdRL1 acts as a positive regulator of plant stress tolerance, presumably by inhibiting PP1 activity. In this work, co-immunoprecipitation and bimolecular fluorescence complementation (BiFC) assays demonstrate that the durum wheat TdPP1 interacts with both TdRL1 and At-I2 in vivo. YFP fluorescence restored after TdRL1-TdPP1 interaction decorated specifically the microtubular network of the tobacco co-infiltrated cells. In vitro phosphatase assays revealed that TdRL1 inhibited the activity of wild-type TdPP1 and two mutant forms (T243M and H135A) in a concentration-dependent manner, showing a novel and potent inhibition mechanism. Structural modeling of the TdPP1-inhibitor complexes suggested that both At-I2 and TdRL1 bind to TdPP1 by wrapping their flexible C-terminal tails around it, blocking access to the active site. Remarkably, the model showed that TdRL1 differs from At-I2 in its interaction with TdPP1 by trapping the phosphatase with its N-terminal tail. These findings provide important insights into the regulatory mechanisms governing the activity of PP1s in plants and highlight the potential for targeted inhibition to modulate plant stress responses. Full article
(This article belongs to the Section Molecular Biology)
Show Figures

Figure 1

Back to TopTop