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

Article Types

Countries / Regions

Search Results (290)

Search Parameters:
Keywords = ubiquitin dependent degradation

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
20 pages, 3707 KiB  
Article
Genome-Wide CRISPR-Cas9 Knockout Screening Identifies NUDCD2 Depletion as Sensitizer for Bortezomib, Carfilzomib and Ixazomib in Multiple Myeloma
by Sophie Vlayen, Tim Dierckx, Marino Caruso, Swell Sieben, Kim De Keersmaecker, Dirk Daelemans and Michel Delforge
Hemato 2025, 6(3), 21; https://doi.org/10.3390/hemato6030021 - 16 Jul 2025
Viewed by 378
Abstract
Background/Objectives: The treatment of multiple myeloma (MM) remains a challenge, as almost all patients will eventually relapse. Proteasome inhibitors are a cornerstone in the management of MM. Unfortunately, validated biomarkers predicting drug response are largely missing. Therefore, we aimed to identify genes associated [...] Read more.
Background/Objectives: The treatment of multiple myeloma (MM) remains a challenge, as almost all patients will eventually relapse. Proteasome inhibitors are a cornerstone in the management of MM. Unfortunately, validated biomarkers predicting drug response are largely missing. Therefore, we aimed to identify genes associated with drug resistance or sensitization to proteasome inhibitors. Methods: We performed genome-wide CRISPR-Cas9 knockout (KO) screens in human KMS-28-BM myeloma cells to identify genetic determinants associated with resistance or sensitization to proteasome inhibitors. Results: We show that KO of KLF13 and PSMC4 induces drug resistance, while NUDCD2, OSER1 and HERC1 KO cause drug sensitization. Subsequently, we focused on top sensitization hit, NUDCD2, which acts as a co-chaperone of Hsp90 to regulate the LIS1/dynein complex. RNA sequencing showed downregulation of genes involved in the ERAD pathway and in ER-associated ubiquitin-dependent protein catabolic processes in both untreated and carfilzomib-treated NUDCD2 KO cells, suggesting that NUDCD2 depletion alters protein degradation. Furthermore, bortezomib-treated NUDCD2 KO cells showed a decreased expression of genes that have a function in oxidative phosphorylation and the mitochondrial membrane, such as Carnitine Palmitoyltransferase 1A (CPT1A). CPT1A catalyzes the uptake of long chain fatty acids into mitochondria. Mitochondrial lipid metabolism has recently been reported as a possible therapeutic target for MM drug sensitivity. Conclusions: These results contribute to the search for therapeutic targets that can sensitize MM patients to proteasome inhibitors. Full article
(This article belongs to the Section Plasma Cell Disorders)
Show Figures

Figure 1

14 pages, 2737 KiB  
Article
Strengthening the Role of PSMC5 as a Potential Gene Associated with Neurodevelopmental Disorders
by Mirella Vinci, Antonino Musumeci, Carla Papa, Alda Ragalmuto, Salvatore Saccone, Concetta Federico, Donatella Greco, Vittoria Greco, Francesco Calì and Simone Treccarichi
Int. J. Mol. Sci. 2025, 26(13), 6386; https://doi.org/10.3390/ijms26136386 - 2 Jul 2025
Viewed by 253
Abstract
The 26S proteasome is a large, ATP-dependent proteolytic complex responsible for degrading ubiquitinated proteins in eukaryotic cells. It plays a crucial role in maintaining cellular protein homeostasis by selectively eliminating misfolded, damaged, or regulatory proteins marked for degradation. In this study, whole-exome sequencing [...] Read more.
The 26S proteasome is a large, ATP-dependent proteolytic complex responsible for degrading ubiquitinated proteins in eukaryotic cells. It plays a crucial role in maintaining cellular protein homeostasis by selectively eliminating misfolded, damaged, or regulatory proteins marked for degradation. In this study, whole-exome sequencing (WES) was performed on an individual presenting with developmental delay and mild intellectual disability, as well as on both of his unaffected parents. This analysis identified a de novo variant, c.959C>G (p.Pro320Arg), in the PSMC5 gene. As predicted, this gene shows a very likely autosomal dominant inheritance pattern. Notably, PSMC5 has not previously been associated with any phenotype in the OMIM database. This variant was recently submitted to the ClinVar database as a variant of uncertain significance (VUS) and remains absent in both gnomAD and dbSNP. Notably, it has been identified in six unrelated individuals presenting with clinical features comparable to those observed in the patient described in this study. Multiple in silico prediction tools classified the variant as pathogenic, and a PhyloP conservation score supports strong evolutionary conservation of the mutated nucleotide. Protein structure predictions using the AlphaFold3 algorithm revealed notable structural differences between the mutant and wild-type PSMC5 proteins. We hypothesize that the p.Pro320Arg substitution alters the structure and function of PSMC5 as a regulatory subunit of the 26S proteasome, potentially impairing the stability and activity of the entire complex. Although functional studies are imperative, this study contributes to a deeper understanding of PSMC5, expands the spectrum of associated neurodevelopmental phenotypes, and highlights its potential as a therapeutic target. Furthermore, this study resulted in the submission of the identified variant to the ClinVar database (SCV006083352), where it was classified as pathogenic. Full article
Show Figures

Figure 1

18 pages, 2646 KiB  
Article
COP1 Deficiency in BRAFV600E Melanomas Confers Resistance to Inhibitors of the MAPK Pathway
by Ada Ndoja, Christopher M. Rose, Eva Lin, Rohit Reja, Jelena Petrovic, Sarah Kummerfeld, Andrew Blair, Helen Rizos, Zora Modrusan, Scott Martin, Donald S. Kirkpatrick, Amy Heidersbach, Tao Sun, Benjamin Haley, Ozge Karayel, Kim Newton and Vishva M. Dixit
Cells 2025, 14(13), 975; https://doi.org/10.3390/cells14130975 - 25 Jun 2025
Viewed by 699
Abstract
Aberrant activation of the mitogen-activated protein kinase (MAPK) cascade promotes oncogenic transcriptomes. Despite efforts to inhibit oncogenic kinases, such as BRAFV600E, tumor responses in patients can be heterogeneous and limited by drug resistance mechanisms. Here, we describe patient tumors that acquired COP1 or [...] Read more.
Aberrant activation of the mitogen-activated protein kinase (MAPK) cascade promotes oncogenic transcriptomes. Despite efforts to inhibit oncogenic kinases, such as BRAFV600E, tumor responses in patients can be heterogeneous and limited by drug resistance mechanisms. Here, we describe patient tumors that acquired COP1 or DET1 mutations after treatment with the BRAFV600E inhibitor vemurafenib. COP1 and DET1 constitute the substrate adaptor of the E3 ubiquitin ligase CRL4COP1/DET1, which targets transcription factors, including ETV1, ETV4, and ETV5, for proteasomal degradation. MAPK-MEK-ERK signaling prevents CRL4COP1/DET1 from ubiquitinating ETV1, ETV4, and ETV5, but the mechanistic details are still being elucidated. We found that patient mutations in COP1 or DET1 inactivated CRL4COP1/DET1 in melanoma cells, stabilized ETV1, ETV4, and ETV5, and conferred resistance to inhibitors of the MAPK pathway. ETV5, in particular, enhanced cell survival and was found to promote the expression of the pro-survival gene BCL2A1. Indeed, the deletion of pro-survival BCL2A1 re-sensitized COP1 mutant cells to vemurafenib treatment. These observations indicate that the post-translational regulation of ETV5 by CRL4COP1/DET1 modulates transcriptional outputs in ERK-dependent cancers, and its inactivation contributes to therapeutic resistance. Full article
(This article belongs to the Special Issue Targeting Hallmarks of Cancer)
Show Figures

Graphical abstract

12 pages, 2647 KiB  
Article
Decursin Suppresses Esophageal Squamous Cell Carcinoma Progression via Orchestrated Cell Cycle Deceleration, Apoptotic Activation, and Oncoprotein Degradation
by Chen Fang, Lin Wu, Xiangzhe Yang, Kai Xie, Peng Zhang, Yu Feng, Haitao Ma and Xing Tong
Int. J. Mol. Sci. 2025, 26(11), 5391; https://doi.org/10.3390/ijms26115391 - 4 Jun 2025
Viewed by 552
Abstract
Esophageal squamous cell carcinoma (ESCC) remains a lethal malignancy with limited therapeutic options. This study investigated the antitumor efficacy and mechanisms of decursin, a natural pyranocoumarin derivative, against ESCC. In vitro analyses demonstrated that decursin selectively inhibited ESCC cell viability (IC50: 14.62 ± [...] Read more.
Esophageal squamous cell carcinoma (ESCC) remains a lethal malignancy with limited therapeutic options. This study investigated the antitumor efficacy and mechanisms of decursin, a natural pyranocoumarin derivative, against ESCC. In vitro analyses demonstrated that decursin selectively inhibited ESCC cell viability (IC50: 14.62 ± 0.61–26.20 ± 2.11 μM across TE-1, KYSE-30, and KYSE-150 cell lines) without affecting normal esophageal epithelial cells (Het-1A). Decursin (10 μM) suppressed colony formation, impaired wound healing (p < 0.001 at 48 h), and reduced Transwell migration/invasion in KYSE-150 cells. Subcutaneous xenograft models revealed significant tumor growth inhibition (p < 0.01) with decursin treatment (10 mg/kg, intraperitoneal), accompanied by no systemic toxicity. Mechanistically, decursin induced G0/G1 cell cycle deceleration (p < 0.01) and apoptosis through ubiquitin–proteasome-mediated degradation of oncoproteins TP63 and SOX2. Time- and dose-dependent protein suppression was reversed by proteasome inhibitor MG-132, but unaffected by lysosomal inhibition. These findings establish decursin as a promising therapeutic agent for ESCC, functioning via proteasomal degradation of key oncogenic drivers, and provide a rationale for decursin’s further development as a targeted monotherapy or chemosensitizer in multimodal regimens. Full article
Show Figures

Figure 1

19 pages, 7673 KiB  
Article
Neuronal Deletion of Tumor Susceptibility Gene 101 (Tsg101) Causes Rapid Apoptotic Loss of Hippocampal CA3 Neurons
by Will P. Walker, Megan Lea Ratz-Mitchem, Kay-Uwe Wagner and Teresa M. Gunn
Biomolecules 2025, 15(6), 786; https://doi.org/10.3390/biom15060786 - 28 May 2025
Viewed by 1201
Abstract
Endosomal dysfunction is one of the earliest cellular signs in Alzheimer’s disease. Tumor susceptibility gene 101 protein (TSG101) is a component of the endosomal sorting complex required for transport (ESCRT)-I, which plays a key role in sorting ubiquitinated cell surface proteins and lipids [...] Read more.
Endosomal dysfunction is one of the earliest cellular signs in Alzheimer’s disease. Tumor susceptibility gene 101 protein (TSG101) is a component of the endosomal sorting complex required for transport (ESCRT)-I, which plays a key role in sorting ubiquitinated cell surface proteins and lipids onto intraluminal vesicles of multivesicular bodies for trafficking to lysosomes or autophagosomes for degradation, or to the plasma membrane for exosomal secretion. TSG101-dependent trafficking has been implicated in the propagation and spread of misfolded proteins associated with neurodegenerative diseases. We used transgenesis mice to study the in vivo consequences of disrupting TSG101-dependent trafficking in adult neurons. Mice lacking Tsg101 in forebrain neurons (Tsg101ck2-null) showed rapid loss of hippocampal neurons and progressive forebrain atrophy. Astrogliosis was apparent in the dentate gyrus within 1 week of deleting Tsg101, followed by apoptosis of hippocampal CA3 neurons and accumulation of the autophagy adapter P62/SQSTM1 and ubiquitinated proteins. Failure to detect lipidated LC3 indicated autophagy was impaired rather than upregulated. Endosomal markers (RAB5 and RAB7) and amyloid protein also accumulated in hippocampal neurons of Tsg101ck2-null mice. Our data establish a critical role for TSG101 in neuronal survival and demonstrate the importance of the in vivo assessment of gene and protein functions. Full article
(This article belongs to the Special Issue Molecular and Genetic Basis of Neurodegenerative Diseases)
Show Figures

Figure 1

21 pages, 4696 KiB  
Article
Mechanism of Curcumol Targeting the OTUB1/TGFBI Ubiquitination Pathway in the Inhibition of Angiogenesis in Colon Cancer
by Yimiao Zhu, Wenya Wu, Dahai Hou, Yu Zhao, Jinshu Ye, Lizong Shen, Tong Zhao and Xiaoyu Wu
Int. J. Mol. Sci. 2025, 26(10), 4899; https://doi.org/10.3390/ijms26104899 - 21 May 2025
Cited by 1 | Viewed by 661
Abstract
Tumor angiogenesis and metastasis are critical processes in the progression of colon carcinoma. Curcumol, a bioactive sesquiterpenoid derived from curcuma, exhibits anti-angiogenic properties, though its underlying mechanisms remain unclear. In this study, an HT-29 xenograft mouse model demonstrated that curcumol combined with oxaliplatin [...] Read more.
Tumor angiogenesis and metastasis are critical processes in the progression of colon carcinoma. Curcumol, a bioactive sesquiterpenoid derived from curcuma, exhibits anti-angiogenic properties, though its underlying mechanisms remain unclear. In this study, an HT-29 xenograft mouse model demonstrated that curcumol combined with oxaliplatin significantly suppressed tumor growth (Ki67↓) and microvessel density (CD31↓). In vitro assays revealed that curcumol dose dependently inhibited proliferation (MTT), migration (Transwell), and tube formation (CAM assay) in Caco-2/HT-29 and HUVEC cells. Mechanistically, curcumol downregulated OTUB1 expression, promoting TGFB1 degradation via the ubiquitin–proteasome pathway. OTUB1 overexpression activated the TGFB1/VEGF axis, enhancing cell invasiveness and angiogenesis—effects reversed by high-dose curcumol. These findings identify the OTUB1-TGFB1/VEGF axis as a key target of curcumol in inhibiting colon cancer angiogenesis, elucidating its anti-tumor mechanism and offering a novel therapeutic strategy for targeted treatment. Full article
(This article belongs to the Section Bioactives and Nutraceuticals)
Show Figures

Figure 1

14 pages, 10417 KiB  
Article
Mechanistic Insights into the Anti-Hepatocellular Carcinoma Effects of ACY-1215: p53 Acetylation and Ubiquitination Regulation
by Yi Yin, Yutong Du, Yiting Xu, Zhuan Zhu, Yu Hu, Lingling Xu, Kunming Yang, Tian Chen, Yuyang Shi, Chengcheng Wang and Yali Zhang
Curr. Issues Mol. Biol. 2025, 47(5), 338; https://doi.org/10.3390/cimb47050338 - 8 May 2025
Viewed by 626
Abstract
As a major global health challenge, hepatocellular carcinoma (HCC) still faces substantial limitations in its treatment options. This study investigates the anti-HCC potential of ACY-1215, a selective Histone deacetylase 6 (HDAC6) inhibitor, and its mechanism targeting p53 regulation. In vitro studies conducted with [...] Read more.
As a major global health challenge, hepatocellular carcinoma (HCC) still faces substantial limitations in its treatment options. This study investigates the anti-HCC potential of ACY-1215, a selective Histone deacetylase 6 (HDAC6) inhibitor, and its mechanism targeting p53 regulation. In vitro studies conducted with HepG2 and SMMC-7721 cells revealed that ACY-1215 markedly inhibited HCC cell proliferation, migratory capacity, and invasive potential, as evidenced by CCK-8, colony formation, and Transwell assays. Furthermore, ACY-1215 induced caspase-dependent apoptosis. Mechanistically, ACY-1215 enhanced p53 acetylation by disrupting HDAC6-p53 interaction, thereby stabilizing p53 protein levels. Concurrently, it inhibited Murine Double Minute 2 (MDM2)-mediated ubiquitination, blocking proteasomal degradation and prolonging p53 half-life. This dual modulation restored p53 transcriptional activity, leading to the upregulation of downstream effector molecules associated with cell cycle regulation and apoptosis. Collectively, our findings reveal that ACY-1215 exerts potent anti-HCC effects through coordinated regulation of p53 acetylation and ubiquitination, offering a novel dual-targeting strategy for HCC therapy. Full article
(This article belongs to the Section Molecular Medicine)
Show Figures

Figure 1

31 pages, 3815 KiB  
Review
PROTAC Delivery Strategies for Overcoming Physicochemical Properties and Physiological Barriers in Targeted Protein Degradation
by Endry Wahyu Syahputra, Hyunji Lee, Hyukjun Cho, Hyun Jin Park, Kwang-Su Park and Duhyeong Hwang
Pharmaceutics 2025, 17(4), 501; https://doi.org/10.3390/pharmaceutics17040501 - 9 Apr 2025
Cited by 1 | Viewed by 3005
Abstract
Proteolysis targeting chimeras (PROTACs), heterobifunctional molecules that hijack the ubiquitin–proteasome system (UPS) to degrade specific proteins, hold great promise in treating diseases driven by traditionally “undruggable” targets. However, their large molecular weight, high hydrophobicity, and other physicochemical hurdles contribute to their limited bioavailability, [...] Read more.
Proteolysis targeting chimeras (PROTACs), heterobifunctional molecules that hijack the ubiquitin–proteasome system (UPS) to degrade specific proteins, hold great promise in treating diseases driven by traditionally “undruggable” targets. However, their large molecular weight, high hydrophobicity, and other physicochemical hurdles contribute to their limited bioavailability, suboptimal pharmacokinetics, and attenuated therapeutic efficacy. Consequently, diverse formulation innovations have been investigated to optimize PROTAC delivery. This review examines current challenges and advances in specialized drug delivery approaches designed to bolster PROTAC pharmacological performance. We first outline the fundamental limitations of PROTACs—their low aqueous solubility, poor cell permeability, rapid clearance, and concentration-dependent “hook effect”. We then discuss how various enabling formulations address these issues, including polymeric micelles, emulsions, amorphous solid dispersions, lipid-based nanoparticles, liposomes, and exosomes. Collectively, these delivery technologies substantially improve the therapeutic outcomes of PROTACs in preclinical cancer models. Future applications may extend beyond oncology to address other complex diseases using newly emerging heterobifunctional molecules. By integrating advanced formulation science with innovative degrader design, the field stands poised to unlock the clinical potential of PROTACs for protein degradation therapies. Full article
(This article belongs to the Special Issue Drug Nanocarriers for Pharmaceutical Applications)
Show Figures

Figure 1

20 pages, 3306 KiB  
Article
Mdm2-Mediated Ubiquitination Plays a Pivotal Role in Differentiating the Endocytic Roles of GRK2 and Arrestin3
by Shujie Wang, Dooti Kundu, Xiaohan Zhang, Xinru Tian, Lulu Peng and Kyeong-Man Kim
Int. J. Mol. Sci. 2025, 26(7), 3238; https://doi.org/10.3390/ijms26073238 - 31 Mar 2025
Viewed by 496
Abstract
Upon activation of certain G protein-coupled receptors, Mdm2 promotes the ubiquitination of both GRK2 and arrestin3. Similar to arrestin3, GRK2 ubiquitination was associated with its endocytic activity and proteasomal degradation. Ubiquitination of GRK2 was essential for arrestin3 ubiquitination, and vice versa. Cellular components [...] Read more.
Upon activation of certain G protein-coupled receptors, Mdm2 promotes the ubiquitination of both GRK2 and arrestin3. Similar to arrestin3, GRK2 ubiquitination was associated with its endocytic activity and proteasomal degradation. Ubiquitination of GRK2 was essential for arrestin3 ubiquitination, and vice versa. Cellular components involved in arrestin3 ubiquitination, including Gβγ, clathrin, and 14-3-3η, were also necessary for GRK2 ubiquitination. Additionally, the arrestin-biased signaling pathway contributed to the ubiquitination of both GRK2 and arrestin3. By employing Mdm2-knockdown cells alongside GRK2 and arrestin3 mutants deficient in ubiquitination sites, as well as receptors lacking phosphorylation sites, we established that the ubiquitinated forms of GRK2 and arrestin3 facilitate clathrin-dependent endocytosis, whereas non-ubiquitinated GRK2 and arrestin3 are responsible for caveolar and a distinct third endocytic pathway, respectively. In the context of clathrin-mediated endocytosis, arrestin3’s interaction with clathrin and GRK2’s interaction with the β2-adaptin subunit of adaptor protein complex 2 were critical. These findings suggest that GRK2 and arrestin3 ubiquitination are mutually dependent, with their ubiquitination states determining their roles in distinct endocytic pathways. Full article
(This article belongs to the Section Molecular Biology)
Show Figures

Figure 1

27 pages, 11801 KiB  
Article
The E3 Ubiquitin Ligase PRAJA1: A Key Regulator of Synaptic Dynamics and Memory Processes with Implications for Alzheimer’s Disease
by Chuhan Li, Yan Yan, Oliver Stork, Ruling Shen and Thomas Behnisch
Int. J. Mol. Sci. 2025, 26(7), 2909; https://doi.org/10.3390/ijms26072909 - 23 Mar 2025
Viewed by 725
Abstract
The precise regulation of synaptic function by targeted protein degradation is fundamental to learning and memory, yet the roles of many brain-enriched E3 ubiquitin ligases in this process remain elusive. Here, we uncover a critical and previously unappreciated role for the E3 ubiquitin [...] Read more.
The precise regulation of synaptic function by targeted protein degradation is fundamental to learning and memory, yet the roles of many brain-enriched E3 ubiquitin ligases in this process remain elusive. Here, we uncover a critical and previously unappreciated role for the E3 ubiquitin ligase PRAJA1 in orchestrating synaptic plasticity and hippocampus-dependent memory. Utilizing C57BL/6 and 5xFAD male mice and employing a multi-faceted approach including protein biochemistry, molecular biology, in vitro electrophysiology, and behavioral assays, we demonstrate that long-term potentiation (LTP) induction triggers a rapid, proteasome-dependent downregulation of PRAJA1 within the CA1 region of the hippocampus. Critically, selective knockdown of PRAJA1 in vivo profoundly enhanced both object recognition and spatial memory, while disrupting normal exploratory behavior. Mechanistically, we reveal that PRAJA1 acts as a key regulator of synaptic architecture and transmission: its downregulation leads to a reduction in key synaptic proteins and spine density, influencing the excitatory/inhibitory balance and facilitating synaptic plasticity. Conversely, increased PRAJA1 expression potentiates GABAergic transmission. Furthermore, we identify spinophilin as a novel substrate of PRAJA1, suggesting a direct molecular link between PRAJA1 and synaptic remodeling. Strikingly, our findings implicate dysregulation of PRAJA1 in the pathogenesis of Alzheimer’s disease, positioning PRAJA1 as a potential therapeutic target for cognitive enhancement in neurodegenerative conditions. These results unveil PRAJA1 as a critical molecular brake on synaptic plasticity and memory formation, offering a promising new avenue for understanding and potentially treating memory impairment. Full article
(This article belongs to the Section Molecular Neurobiology)
Show Figures

Figure 1

16 pages, 2388 KiB  
Review
Targeting the PARylation-Dependent Ubiquitination Signaling Pathway for Cancer Therapies
by Daoyuan Huang, Jingchao Wang, Li Chen, Weiwei Jiang, Hiroyuki Inuzuka, David K. Simon and Wenyi Wei
Biomolecules 2025, 15(2), 237; https://doi.org/10.3390/biom15020237 - 7 Feb 2025
Cited by 2 | Viewed by 1478
Abstract
Poly(ADP-ribosyl)ation (PARylation) is a dynamic protein post-translational modification (PTM) mediated by ADP-ribosyltransferases (ARTs), which regulates a plethora of essential biological processes, such as DNA repair, gene expression, and signal transduction. Among these, PAR-dependent ubiquitination (PARdU) plays a pivotal role in tagging PARylated substrates [...] Read more.
Poly(ADP-ribosyl)ation (PARylation) is a dynamic protein post-translational modification (PTM) mediated by ADP-ribosyltransferases (ARTs), which regulates a plethora of essential biological processes, such as DNA repair, gene expression, and signal transduction. Among these, PAR-dependent ubiquitination (PARdU) plays a pivotal role in tagging PARylated substrates for subsequent ubiquitination and degradation events through the coordinated action of enzymes, including the E3 ligase RNF146 and the ADP-ribosyltransferase tankyrase. Notably, this pathway has emerged as a key regulator of tumorigenesis, immune modulation, and cell death. This review elucidates the molecular mechanisms of the PARdU pathway, including the RNF146–tankyrase interaction, substrate specificity, and upstream regulatory pathways. It also highlights the biological functions of PARdU in DNA damage repair, signaling pathways, and metabolic regulation, with a focus on its therapeutic potential in cancer treatment. Strategies targeting PARdU, such as tankyrase and RNF146 inhibitors, synthetic lethality approaches, and immune checkpoint regulation, offer promising avenues for precision oncology. These developments underscore the potential of PARdU as a transformative therapeutic target in combating various types of human cancer. Full article
Show Figures

Figure 1

20 pages, 3167 KiB  
Review
Phosphodegrons in Health and Disease: From Cellular Homeostasis to Therapeutic Potential
by Tadashi Nakagawa and Makiko Nakagawa
Kinases Phosphatases 2025, 3(1), 3; https://doi.org/10.3390/kinasesphosphatases3010003 - 6 Feb 2025
Cited by 1 | Viewed by 1528
Abstract
Phosphodegrons are critical motifs that play a pivotal role in the regulation of protein stability and function via phosphorylation-dependent signaling pathways. These motifs serve as recognition elements for ubiquitin ligases, facilitating the targeted degradation of proteins. By modulating key cellular processes such as [...] Read more.
Phosphodegrons are critical motifs that play a pivotal role in the regulation of protein stability and function via phosphorylation-dependent signaling pathways. These motifs serve as recognition elements for ubiquitin ligases, facilitating the targeted degradation of proteins. By modulating key cellular processes such as cell cycle progression, DNA repair, and apoptosis, phosphodegrons are essential for maintaining cellular homeostasis. Dysregulation of phosphodegrons has been implicated in a wide range of diseases, including cancer and neurodegenerative disorders, highlighting their potential as therapeutic targets. This review provides an overview of phosphodegron functions along with their biological significance in health and disease. Additionally, we discuss current methodologies for studying phosphodegrons and explore emerging trends in their identification and therapeutic targeting. By synthesizing recent advances in the field, this article aims to offer insights into the future directions and challenges in phosphodegron research, ultimately underscoring their importance in cellular regulation and disease pathology. Full article
Show Figures

Figure 1

18 pages, 3056 KiB  
Article
TRIM38 Inhibits Zika Virus by Upregulating RIG-I/MDA5 Pathway and Promoting Ubiquitin-Mediated Degradation of Viral NS3 Protein
by Jing He, Yulian Kuang, Kui Xu, Rong Huang, Xiaoyao Yang, Liyao Deng, Xiaojuan Feng, Yang Ren, Jian Yang and Lei Yuan
Viruses 2025, 17(2), 199; https://doi.org/10.3390/v17020199 - 30 Jan 2025
Viewed by 1231
Abstract
Members of the tripartite motif (TRIM)-containing protein family play crucial roles in regulating immune system responses. The TRIM38 protein regulates host innate immunity and directly degrades some viral proteins through its E3 ubiquitin ligase activity. This study demonstrated that Zika virus (ZIKV) infection [...] Read more.
Members of the tripartite motif (TRIM)-containing protein family play crucial roles in regulating immune system responses. The TRIM38 protein regulates host innate immunity and directly degrades some viral proteins through its E3 ubiquitin ligase activity. This study demonstrated that Zika virus (ZIKV) infection can promote the expression of TRIM38 in human glioma cells (U251). TRIM38 overexpression restricted ZIKV replication in U251 cells, while TRIM38 knockout enhanced ZIKV replication. TRIM38 overexpression upregulated the RIG-I/MDA5 pathway and promoted the level of IFN-β early during viral infection, while TRIM38 knockout had the opposite effect. In addition, TRIM38 interacts with ZIKV non-structural protein 3 (NS3) and degrades the NS3 protein through a lysosome-dependent manner via the E3 ligase activity of TRIM38. Deletion of the RING domain of TRIM38 abrogates its interaction with NS3 and impairs the antiviral activity of TRIM38. Our results indicate that TRIM38 is a novel antiviral protein against ZIKV, and it exerts antiviral activity by upregulating the RIG-I/MDA5 pathway, increasing IFN-β levels, and degrading the viral NS3 protein. Full article
(This article belongs to the Section Viral Immunology, Vaccines, and Antivirals)
Show Figures

Figure 1

20 pages, 4501 KiB  
Article
Hypomyelinating Leukodystrophy 14 (HLD14)-Related UFC1 p.Arg23Gln Decreases Cell Morphogenesis: A Phenotype Reversable with Hesperetin
by Yuri Ichihara, Maho Okawa, Minori Minegishi, Hiroaki Oizumi, Masahiro Yamamoto, Katsuya Ohbuchi, Yuki Miyamoto and Junji Yamauchi
Medicines 2025, 12(1), 2; https://doi.org/10.3390/medicines12010002 - 16 Jan 2025
Viewed by 1419
Abstract
Introduction: In the central nervous system (CNS), proper interaction between neuronal and glial cells is crucial for the development of mature nervous tissue. Hypomyelinating leukodystrophies (HLDs) are a group of genetic CNS disorders characterized by hypomyelination and/or demyelination. In these conditions, genetic mutations [...] Read more.
Introduction: In the central nervous system (CNS), proper interaction between neuronal and glial cells is crucial for the development of mature nervous tissue. Hypomyelinating leukodystrophies (HLDs) are a group of genetic CNS disorders characterized by hypomyelination and/or demyelination. In these conditions, genetic mutations disrupt the biological functions of oligodendroglial cells, which are responsible for wrapping neuronal axons with myelin sheaths. Among these, an amino acid mutation of the ubiquitin-fold modifier conjugating enzyme 1 (UFC1) is associated with HLD14-related disease, characterized by hypomyelination and delayed myelination in the brain. UFC1 is a critical component of the UFMylation system, functioning similarly to E2-conjugating enzymes in the ubiquitin-dependent protein degradation system. Methodology: We describe how a missense mutation in UFC1 (p.Arg23Gln) leads to the aggregation of UFC1 primarily in lysosomes in FBD-102b cells, which are undergoing oligodendroglial cell differentiation. Results: Cells with mutated UFC1 exhibit reduced Akt kinase phosphorylation and reduced expression of differentiation and myelination marker proteins. Consistently, these cells exhibit impaired morphological differentiation with a reduced ability to extend widespread membranes. Interestingly, hesperetin, a citrus flavonoid with known neuroprotective properties, was found to restore differentiation abilities in cells with the UFC1 mutation. Conclusions: These findings indicate that the HLD14-related mutation in UFC1 causes its lysosomal aggregation, impairing its morphological differentiation. Furthermore, the study highlights potential therapeutic insights into the pathological molecular and cellular mechanisms underlying HLD14 and suggests hesperetin as a promising candidate for treatment. Full article
Show Figures

Figure 1

18 pages, 2480 KiB  
Article
Differential Activity and Expression of Proteasome in Seminiferous Epithelium During Mouse Spermatogenesis
by Héctor Zapata-Carmona, Emilce Silvina Díaz, Patricio Morales and Marco Jara
Int. J. Mol. Sci. 2025, 26(2), 494; https://doi.org/10.3390/ijms26020494 - 9 Jan 2025
Viewed by 861
Abstract
Proteasome-mediated protein degradation is essential for maintaining cellular homeostasis, particularly during spermatogenesis, where extensive cellular transformations, such as spermatid differentiation, require precise protein turnover. A key player in this process is the ubiquitin–proteasome system (UPS). This study aimed to investigate proteasome enzymatic activity [...] Read more.
Proteasome-mediated protein degradation is essential for maintaining cellular homeostasis, particularly during spermatogenesis, where extensive cellular transformations, such as spermatid differentiation, require precise protein turnover. A key player in this process is the ubiquitin–proteasome system (UPS). This study aimed to investigate proteasome enzymatic activity at different stages of the spermatogenic cycle within the seminiferous tubules of mice and explore the regulatory mechanisms that influence its proteolytic function. Specifically, we assessed the trypsin-like, chymotrypsin-like, and peptidyl-glutamyl-peptide-hydrolyzing (PGPH) activities of the proteasome. Additionally, we examined the expression of catalytic and structural subunits of the 20S core, the assembly of the 20S core with regulatory complexes, and the phosphorylation status of proteasome subunits in various segments of the seminiferous tubules. Our findings demonstrated distinct patterns of proteasomal enzymatic activity in the analyzed segments. While the expression levels of structural and catalytic subunits of the 20S core remained consistent, significant differences were detected in the assembly of the 20S core, the expression of regulatory complexes, and the phosphorylation of proteasome subunits mediated by protein kinase A. These results indicate that proteasomal activity is finely regulated through multiple mechanisms depending on the specific stage of the seminiferous epithelial cycle, highlighting the complexity of proteostasis during spermatogenesis. Full article
Show Figures

Figure 1

Back to TopTop