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

Journals

Article Types

Countries / Regions

Search Results (111)

Search Parameters:
Keywords = damaged DNA binding protein 1

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
25 pages, 1490 KB  
Review
Linking Cell Architecture to Mitochondrial Signaling in Neurodegeneration: The Role of Intermediate Filaments
by Emanuele Marzetti, Rosa Di Lorenzo, Riccardo Calvani, Hélio José Coelho-Júnior, Francesco Landi, Vito Pesce and Anna Picca
Int. J. Mol. Sci. 2025, 26(24), 11852; https://doi.org/10.3390/ijms262411852 - 8 Dec 2025
Viewed by 264
Abstract
Mitochondrial dysfunction is a pivotal contributor to neurodegeneration. Neurons heavily rely on mitochondrial oxidative metabolism and therefore need highly efficient quality control mechanisms, including proteostasis, mitochondrial biogenesis, fusion–fission dynamics, and mitophagy, to sustain bioenergetics and synaptic function. With aging, deterioration of mitochondrial quality [...] Read more.
Mitochondrial dysfunction is a pivotal contributor to neurodegeneration. Neurons heavily rely on mitochondrial oxidative metabolism and therefore need highly efficient quality control mechanisms, including proteostasis, mitochondrial biogenesis, fusion–fission dynamics, and mitophagy, to sustain bioenergetics and synaptic function. With aging, deterioration of mitochondrial quality control pathways leads to impaired oxidative phosphorylation, excessive reactive oxygen species generation, calcium imbalance, and defective clearance of damaged organelles, ultimately compromising neuronal viability. Pathological protein aggregates, such as α-synuclein in Parkinson’s disease, β-amyloid and tau in Alzheimer’s disease, and misfolded superoxide dismutase 1 and transactive response DNA-binding protein 43 in amyotrophic lateral sclerosis, further aggravate mitochondrial stress, establishing self-perpetuating cycles of neurotoxicity. Such mitochondrial defects underscore mitochondria as a convergent pathogenic hub and a promising therapeutic target for neuroprotection. Intermediate filaments (IFs), traditionally viewed as passive structural elements, have recently gained attention for their roles in cytoplasmic organization, mitochondrial positioning, and energy regulation. Emerging evidence indicates that IF–mitochondria interactions critically influence organelle morphology and function in neurons. This review highlights the multifaceted involvement of mitochondrial dysfunction and IF dynamics in neurodegeneration, emphasizing their potential as targets for novel therapeutic strategies. Full article
(This article belongs to the Section Molecular Neurobiology)
Show Figures

Figure 1

16 pages, 2922 KB  
Article
Proteomic Analysis of Liver Injury Induced by Deoxynivalenol in Piglets
by Xiaoshu Xue, Ping Wu, Shuhao Fan, Zongjun Yin and Xiaodong Zhang
Biology 2025, 14(12), 1721; https://doi.org/10.3390/biology14121721 - 1 Dec 2025
Viewed by 249
Abstract
Deoxynivalenol (DON, commonly known as vomitoxin) is one of the most prevalent mycotoxins contaminating feed in China, posing a serious threat to the health of piglets. Beyond intestinal damage, the liver is a key target organ for the systemic toxicity of DON, but [...] Read more.
Deoxynivalenol (DON, commonly known as vomitoxin) is one of the most prevalent mycotoxins contaminating feed in China, posing a serious threat to the health of piglets. Beyond intestinal damage, the liver is a key target organ for the systemic toxicity of DON, but its hepatotoxic molecular mechanisms, particularly the changes at the proteome level, remain unclear. To investigate the protein regulatory network of DON-induced liver injury in piglets, this study systematically analyzed differential expression in the liver proteome using quantitative proteomic techniques. Proteomic analysis identified 5851 proteins in total, among which 88 were differentially expressed proteins (DEPs), including 39 upregulated and 49 downregulated proteins. Bioinformatics analysis revealed that these DEPs were significantly enriched in pathways such as DNA damage repair, RNA metabolism, ribosome biogenesis, and cysteine metabolism. Suppressed expression of key proteins like Replication Factor C Subunit 4 (RFC4) and Exosome Component 9 (EXOSC9) indicated that DON exposure severely disrupted the maintenance of genomic stability and RNA processing capacity in hepatocytes. Conversely, the activation of Nucleic Acid Binding Protein 1 (NABP1) might represent a compensatory DNA protection response. Furthermore, the upregulation of Lactate Dehydrogenase B (LDHB) suggested that DON might influence epigenetic modifications by regulating lactate metabolism. This study reveals, for the first time from a proteomic perspective, a novel mechanism by which DON induces hepatotoxicity in piglets by disrupting DNA repair and RNA metabolic homeostasis, providing an important theoretical basis and data support for elucidating the toxicological effects of DON and improving feed biosafety control strategies. Full article
(This article belongs to the Special Issue Feature Papers on Toxicology)
Show Figures

Figure 1

25 pages, 581 KB  
Review
The Emerging Role of the Cancerous Inhibitor of Protein Phosphatase 2A in Pulmonary Diseases
by Hamza Hamza, Dinesh Nirmal, Stephanie Pappas, Ugochukwu Ebubechukwu, Sunydip Gill, Adam Al-Ajam, Michael Ohlmeyer and Patrick Geraghty
Medicina 2025, 61(10), 1740; https://doi.org/10.3390/medicina61101740 - 25 Sep 2025
Viewed by 1085
Abstract
Promising protein targets are observed to play a role in multiple pathways across a variety of diseases, such as the regulation of immune responses, cell cycle, senescence, and DNA repair. The oncoprotein cancerous inhibitor of protein phosphatase 2A (CIP2A) can coordinate all these [...] Read more.
Promising protein targets are observed to play a role in multiple pathways across a variety of diseases, such as the regulation of immune responses, cell cycle, senescence, and DNA repair. The oncoprotein cancerous inhibitor of protein phosphatase 2A (CIP2A) can coordinate all these cell characteristics predominately by inhibiting the activity of the serine threonine protein phosphatase 2A (PP2A). CIP2A directly interacts with PP2A and other proteins, such as the DNA damage protein topoisomerase II-binding protein 1, to regulate signal transduction. CIP2A is overexpressed in many human cancers, including small and non-small cell lung cancers. High CIP2A expression in lung cancer correlates with poor prognosis, increased tumor proliferation, and resistance to targeted therapies or chemotherapy. Interestingly, CIP2A expression or signaling is also observed in several non-cancerous pulmonary diseases, such as chronic obstructive pulmonary disease. CIP2A can determine whether DNA-damaged cells enter mitosis and can mediate whether DNA repair occurs. CIP2A is also a regulator of inflammation and possibly fibrotic responses. Its functions are linked to altered NFκB activation and TNFα, IL-1β, IL-4, IL-6, IL-10, IL-13, and TGFβ signaling. This review outlines the possible impact of CIP2A-mediated signaling in pulmonary diseases, the processes that regulate CIP2A responses, CIP2A-dependent pathways, and potential therapeutic strategies targeting CIP2A. Substantial medicinal chemistry efforts are underway to develop therapeutics aimed at modulating CIP2A activity. The development of specific inhibitors of CIP2A that selectively target its expression or protein stability could improve our understanding of CIP2A’s function in pulmonary diseases. Full article
(This article belongs to the Section Pulmonology)
Show Figures

Figure 1

17 pages, 299 KB  
Review
Indications and Mechanisms of Action of the Main Treatment Modalities for Non-Melanoma Skin Cancer
by Marcio F. Chedid, Aline C. Tregnago, Floriano Riva, Lucas Prediger, Anisha Agarwal and Jane Mattei
Life 2025, 15(9), 1447; https://doi.org/10.3390/life15091447 - 16 Sep 2025
Viewed by 1362
Abstract
Skin cancer is the most common cancer worldwide. The incidence of skin cancer has been increasing worldwide. Nearly 75% of all skin cancers are basal cell carcinomas (BCC), cutaneous squamous cell carcinoma (cSCC) represents approximately 20%, and those remaining are melanomas (4%) or [...] Read more.
Skin cancer is the most common cancer worldwide. The incidence of skin cancer has been increasing worldwide. Nearly 75% of all skin cancers are basal cell carcinomas (BCC), cutaneous squamous cell carcinoma (cSCC) represents approximately 20%, and those remaining are melanomas (4%) or other rare tumors (1%). Given the high cure rates and the ability to histologically confirm tumor clearance, surgical therapy is the gold standard for the treatment of skin cancer. Conventional surgery is the most employed technique for the removal of non-melanoma skin cancer (NMSCs). Mohs Micrographic Surgery (MMS) is the most precise surgical method for the treatment of non-melanoma skin cancer, allowing for 100% margin evaluation, being the gold-standard method for surgical treatment of non-melanoma skin cancer. Whenever it is possible to obtain wide margins (4 to 6 mm), cure rates vary from 70% to 99%. Imiquimod, a synthetic imidazoquinolinone amine, is a topical immune response modifier approved by the U.S. Food and Drug Administration (FDA) for the treatment of external anogenital warts, actinic keratosis (AK), and superficial basal cell carcinoma (sBCC). The efficacy of imiquimod is primarily attributed to its ability to modulate both innate and adaptive immune responses, as well as its direct effects on cancer cells. Imiquimod exerts its immunomodulatory effects by activating Toll-like receptors 7 and 8 (TLR7/8) on various immune cells, including dendritic cells, macrophages, and natural killer (NK) cells. Upon binding to these receptors, imiquimod triggers the MyD88-dependent signaling pathway, leading to the activation of nuclear factor kappa B (NF-κB) and interferon regulatory factors (IRFs). This cascade leads to the production of pro-inflammatory cytokines, including interferon-alpha (IFN-α), tumor necrosis factor-alpha (TNF-α), interleukin-12 (IL-12), and interleukin-6 (IL-6). These cytokines enhance local inflammation, recruit additional immune cells to the tumor site, and stimulate antigen presentation, thereby promoting an anti-tumor immune response. Radiation therapy (RTh) may be employed as a primary treatment to BCC. It may also be employed as an adjuvant treatment to surgery for SCC and aggressive subtypes of BCC. RTh triggers both direct and indirect DNA damage on cancer cells and generates reactive oxygen species (ROS) within cells. ROS trigger oxidative damage to DNA, proteins, and lipids, exacerbating the cellular stress and contributing to tumor cell death. Recently, immunotherapy emerged as a revolutionary treatment for all stages of SCC. Cemiplimab is a human programmed cell death 1 (PD-1)-blocking antibody that triggers a response to over 50% of patients with locally advanced and metastatic SCC. A randomized clinical trial (RCT) published in 2022 revealed that cemiplimab was highly effective in the neoadjuvant treatment of large SCCs. The drug promoted a significant tumor size decrease, enabling organ-sparing operations and a much better cosmetic effect. A few months ago, a RCT of cemiplimab on adjuvant therapy for locally aggressive SCC was published. Interestingly, cemiplimab was administered to patients with local or regional cutaneous squamous cell carcinoma after surgical resection and postoperative radiotherapy, at high risk for recurrence owing to nodal features, revealed that cemiplimab led to much lower risks both of locoregional recurrence and distant recurrence. Full article
18 pages, 2164 KB  
Article
The Fanconi Anemia Pathway Inhibits mTOR Signaling and Prevents Accelerated Translation in Head and Neck Cancer Cells
by Bianca Ruffolo, Sara Vicente-Muñoz, Khyati Y. Mehta, Cosette M. Rivera-Cruz, Xueheng Zhao, Lindsey Romick, Kenneth D. R. Setchell, Adam Lane and Susanne I. Wells
Cancers 2025, 17(15), 2583; https://doi.org/10.3390/cancers17152583 - 6 Aug 2025
Viewed by 1335
Abstract
Background/Objectives: The Fanconi anemia (FA) pathway is essential for the repair of DNA interstrand crosslinks and maintenance of genomic stability. Germline loss of FA pathway function in the inherited Fanconi anemia syndrome leads to increased DNA damage and a range of clinical phenotypes, [...] Read more.
Background/Objectives: The Fanconi anemia (FA) pathway is essential for the repair of DNA interstrand crosslinks and maintenance of genomic stability. Germline loss of FA pathway function in the inherited Fanconi anemia syndrome leads to increased DNA damage and a range of clinical phenotypes, including a heightened risk of head and neck squamous cell carcinoma (HNSCC). Non-synonymous FA gene mutations are also observed in up to 20% of sporadic HNSCCs. The mechanistic target of rapamycin (mTOR) is known to stimulate cell growth, anabolic metabolism including protein synthesis, and survival following genotoxic stress. Methods/Results: Here, we demonstrate that FA− deficient (FA−) HNSCC cells exhibit elevated intracellular amino acid levels, increased total protein content, and an increase in protein synthesis indicative of enhanced translation. These changes are accompanied by hyperactivation of the mTOR effectors translation initiation factor 4E Binding Protein 1 (4E-BP1) and ribosomal protein S6. Treatment with the mTOR inhibitor rapamycin reduced the phosphorylation of these targets and blocked translation specifically in FA− cells but not in their isogenic FA− proficient (FA+) counterparts. Rapamycin-mediated mTOR inhibition sensitized FA− but not FA+ cells to rapamycin under nutrient stress, supporting a therapeutic metabolism-based vulnerability in FA− cancer cells. Conclusions: These findings uncover a novel role for the FA pathway in suppressing mTOR signaling and identify mTOR inhibition as a potential strategy for targeting FA− HNSCCs. Full article
(This article belongs to the Special Issue Targeted Therapy in Head and Neck Cancer)
Show Figures

Figure 1

11 pages, 775 KB  
Review
Cooperation Between Aflatoxin-Induced p53 Aberrations and Hepatitis B Virus in Hepatocellular Carcinoma
by Carolina Moreno-León and Francisco Aguayo
J. Xenobiot. 2025, 15(4), 96; https://doi.org/10.3390/jox15040096 - 20 Jun 2025
Cited by 3 | Viewed by 3770
Abstract
Hepatocellular carcinoma (HCC) imposes a significant burden on global public health. Exposure to aflatoxins, potent mycotoxins produced by Aspergillus fungi contaminating staple foods, and chronic hepatitis B virus (HBV) infection are major etiological factors, especially where they co-exist. This review examines the critical [...] Read more.
Hepatocellular carcinoma (HCC) imposes a significant burden on global public health. Exposure to aflatoxins, potent mycotoxins produced by Aspergillus fungi contaminating staple foods, and chronic hepatitis B virus (HBV) infection are major etiological factors, especially where they co-exist. This review examines the critical role of the p53 tumor suppressor pathway as a primary target and convergence point for the carcinogenic actions of aflatoxins and HBV. Aflatoxin B1 (AFB1), a Group 1 carcinogen, exerts significant genotoxicity, characteristically inducing a specific hotspot mutation (R249S) in the TP53 gene via DNA adduct formation, thereby compromising p53’s critical tumor suppressor functions. This R249S mutation is considered a molecular fingerprint of aflatoxin exposure. Concurrently, the HBV X protein (HBx) functionally inactivates wild-type p53 through direct binding and by promoting its degradation. The synergistic disruption of the p53 pathway, driven by AFB1-induced mutation and amplified by HBV-mediated functional inhibition, significantly enhances the risk of HCC development. This review addresses how aflatoxin exposure alters key aspects of p53 and how this damage interacts with HBV-mediated p53 suppression, providing crucial insights into hepatocarcinogenesis. The knowledge synthesized here underscores the importance of mitigating aflatoxin exposure alongside HBV control for effective HCC prevention and treatment strategies. Full article
Show Figures

Figure 1

18 pages, 5795 KB  
Article
C1QBP Modulates DNA Damage Response and Radiosensitivity in Hepatocellular Carcinoma by Regulating NF-κB Activity
by Haitao Zhou, Yanjin Wu, Jiahui Meng, Xiaotong Zhao, Yujia Hou, Qin Wang and Yang Liu
Int. J. Mol. Sci. 2025, 26(10), 4513; https://doi.org/10.3390/ijms26104513 - 9 May 2025
Viewed by 1163
Abstract
C1QBP (Complement Component 1 Q Subcomponent-Binding Protein) plays a critical role in maintaining cellular metabolism, but its function in radiation-induced damage remains unclear. In this study, we generated C1QBP-deficient Huh-7 hepatocellular carcinoma (HCC) cells using CRISPR/Cas9 technology and observed that C1QBP deficiency significantly [...] Read more.
C1QBP (Complement Component 1 Q Subcomponent-Binding Protein) plays a critical role in maintaining cellular metabolism, but its function in radiation-induced damage remains unclear. In this study, we generated C1QBP-deficient Huh-7 hepatocellular carcinoma (HCC) cells using CRISPR/Cas9 technology and observed that C1QBP deficiency significantly enhanced radiation-induced damage, as indicated by reduced cell proliferation, impaired colony formation, and increased γ-H2AX foci, a marker of DNA double-strand breaks. Additionally, C1QBP deficiency resulted in elevated phosphorylation of key DNA damage response (DDR) molecules, ATM and CHK2, and caused pronounced S phase cell cycle arrest. Mechanistic investigations revealed that C1QBP modulates NF-κB nuclear activity via the AMPK signaling pathway. The loss of C1QBP reduced NF-κB nuclear translocation, further exacerbating radiation-induced damage. Reintroducing C1QBP alleviated DNA damage, enhanced cell proliferation, and improved survival following radiation exposure. These findings highlight the critical role of C1QBP in modulating HCC cells radiosensitivity and underscore its potential as a therapeutic target to enhance radiotherapy outcomes. Full article
(This article belongs to the Special Issue Radiation-Induced DNA Damage and Toxicity)
Show Figures

Figure 1

35 pages, 5069 KB  
Review
Small-Molecule Mitotic Inhibitors as Anticancer Agents: Discovery, Classification, Mechanisms of Action, and Clinical Trials
by Yazmin Salinas, Subhash C. Chauhan and Debasish Bandyopadhyay
Int. J. Mol. Sci. 2025, 26(7), 3279; https://doi.org/10.3390/ijms26073279 - 1 Apr 2025
Cited by 5 | Viewed by 4972
Abstract
Despite decades of research, cancer continues to be a disease of great concern to millions of people around the world. It has been responsible for a total of 609,820 deaths in the U.S. alone in 2023. Over the years, many drugs have been [...] Read more.
Despite decades of research, cancer continues to be a disease of great concern to millions of people around the world. It has been responsible for a total of 609,820 deaths in the U.S. alone in 2023. Over the years, many drugs have been developed to remove or reduce the disease’s impact, all with varying mechanisms of action and side effects. One class of these drugs is small-molecule mitotic inhibitors. These drugs inhibit cancer cell mitosis or self-replication, impeding cell proliferation and eventually leading to cell death. In this paper, small-molecule mitotic inhibitors are discussed and classified through their discovery, underlying chemistry, and mechanism(s) of action. The binding/inhibition of microtubule-related proteins, DNA damage through the inhibition of Checkpoint Kinase 1 protein, and the inhibition of mitotic kinase proteins are discussed in terms of their anticancer activity to provide an overview of a variety of mitotic inhibitors currently commercially available or under investigation, including those in ongoing clinical trial. Clinical trials for anti-mitotic agents are discussed to track research progress, gauge current understanding, and identify possible future prospects. Additionally, antibody–drug conjugates that use mitotic inhibitors as cytotoxic payloads are discussed as possible ways of administering effective anticancer treatments with minimal toxicity. Full article
(This article belongs to the Collection Feature Papers in Molecular Oncology)
Show Figures

Figure 1

13 pages, 2428 KB  
Article
Identification of Quantitative Trait Loci and Analysis of Novel Candidate Genes for Resistance to False Smut of Rice Based on SSR Molecular Markers
by Rongtao Fu, Liyu Zhao, Cheng Chen, Jian Wang, Yu Chen and Daihua Lu
Biomolecules 2025, 15(2), 186; https://doi.org/10.3390/biom15020186 - 28 Jan 2025
Cited by 4 | Viewed by 1316
Abstract
Rice false smut (RFS), an emerging disease caused by the fungus Ustilaginoidea virens (Cooke), reduces rice grain yield and quality in rice-planting regions worldwide. The identification of the genes or quantitative trait loci (QTLs) associated with RFS resistance is vital to resistance breeding [...] Read more.
Rice false smut (RFS), an emerging disease caused by the fungus Ustilaginoidea virens (Cooke), reduces rice grain yield and quality in rice-planting regions worldwide. The identification of the genes or quantitative trait loci (QTLs) associated with RFS resistance is vital to resistance breeding and the mitigation of RFS damage. In this study, RFS resistance QTLs were located in the resistant variety IR77298-14-1-2::IRGC117374-1. A total of 4 RFS resistance QTLs were detected on rice chromosomes 1, 3, 5, and 12 in the F2 and F4 mapping populations using 119 polymorphic simple sequence repeat (SSR) molecular markers. Of these QTLs, qRFS3.01 and qRFS12.01-1 were repeatedly detected in both populations. Interestingly, QTL qRFS3.01 on chromosome 3 is a novel resistance locus that exhibited the largest phenotypic effect. These results suggest that SSR markers linked to qRFS3.01 are valuable for marker-assisted breeding for RFS resistance in rice. The prediction of putative candidate genes within qRFS3.01 revealed three resistance-related proteins containing an F-box domain, Myb-like DNA-binding domain, and kinase protein. In summary, our findings provide new QTLs/genes for resistance to RFS and will promote rice disease resistance through molecular-marker-assisted breeding. Full article
(This article belongs to the Collection Feature Papers in Molecular Genetics)
Show Figures

Figure 1

17 pages, 1695 KB  
Article
Interaction of DDB1 with NBS1 in a DNA Damage Checkpoint Pathway
by Hoe Eun Lim, Hee Jung Lim and Hae Yong Yoo
Int. J. Mol. Sci. 2024, 25(23), 13097; https://doi.org/10.3390/ijms252313097 - 5 Dec 2024
Cited by 1 | Viewed by 1816
Abstract
Various DNA damage checkpoint control mechanisms in eukaryotic cells help maintain genomic integrity. Among these, NBS1, a key component of the MRE11-RAD50-NBS1 (MRN) complex, is an essential protein involved in the DNA damage response (DDR). In this study, we discovered that DNA damage-binding [...] Read more.
Various DNA damage checkpoint control mechanisms in eukaryotic cells help maintain genomic integrity. Among these, NBS1, a key component of the MRE11-RAD50-NBS1 (MRN) complex, is an essential protein involved in the DNA damage response (DDR). In this study, we discovered that DNA damage-binding protein 1 (DDB1) interacts with NBS1. DDB1 is a DDR sensor protein found in UV-induced DNA replication blocks. Through pull-down and immunoprecipitation assays conducted in Xenopus egg extracts and human cell lines, we demonstrated a specific interaction between NBS1 and DDB1. DDB1 was also found to associate with several proteins that interact with NBS1, including DNA topoisomerase 2-binding protein 1 (TopBP1) and Mediator of DNA damage checkpoint protein 1 (MDC1). Notably, the interaction between DDB1 and NBS1 is disrupted in MDC1-depleted egg extracts, indicating that MDC1 is necessary for this interaction. Furthermore, the depletion of DDB1 leads to increased Chk1 activation upon DNA damage. These novel findings regarding the interaction between NBS1 and DDB1 provide new insights into how DDB1 regulates DNA damage pathways. Full article
(This article belongs to the Section Molecular Biology)
Show Figures

Figure 1

23 pages, 4579 KB  
Review
Mechanistic Insights into Influenza A Virus-Induced Cell Death and Emerging Treatment Strategies
by Yuling Sun and Kaituo Liu
Vet. Sci. 2024, 11(11), 555; https://doi.org/10.3390/vetsci11110555 - 10 Nov 2024
Cited by 6 | Viewed by 5827
Abstract
Influenza A virus (IAV) infection initiates a complex interplay of cell death modalities, including apoptosis, necroptosis, pyroptosis, and their integration, known as PANoptosis, which significantly impacts host immune responses and tissue integrity. These pathways are intricately regulated by viral proteins and host factors, [...] Read more.
Influenza A virus (IAV) infection initiates a complex interplay of cell death modalities, including apoptosis, necroptosis, pyroptosis, and their integration, known as PANoptosis, which significantly impacts host immune responses and tissue integrity. These pathways are intricately regulated by viral proteins and host factors, contributing to both viral clearance and pathogenesis-related tissue damage. This review comprehensively explores the molecular mechanisms underlying these cell death processes in influenza infection. We highlight the roles of key regulatory proteins, such as ZBP1 (Z-DNA binding protein 1) and RIPK3 (receptor-interacting protein kinase 3), in orchestrating these responses, emphasizing the dual roles of cell death in both antiviral defense and tissue injury. Furthermore, we discuss emerging therapeutic strategies targeting these pathways, aiming to enhance antiviral efficacy while minimizing collateral tissue damage. Future research should focus on targeted approaches to modulate cell death mechanisms, aiming to reduce tissue damage and improve clinical outcomes for patients with severe influenza. Full article
(This article belongs to the Special Issue Advances in Veterinary Clinical Microbiology)
Show Figures

Figure 1

14 pages, 6226 KB  
Article
Significance of P53-Binding Protein 1 as a Novel Molecular Histological Marker for Hypopharyngeal Squamous Neoplasms
by Hiroko Kawasaki-Inomata, Maiko Tabuchi, Kiyuu Norimatsu, Tetsuro Honda, Katsuya Matsuda, Keiichi Hashiguchi, Naoyuki Yamaguchi, Hideaki Nishi, Yoshihiko Kumai, Masahiro Nakashima, Hisamitsu Miyaaki, Kazuhiko Nakao and Yuko Akazawa
Cancers 2024, 16(17), 2987; https://doi.org/10.3390/cancers16172987 - 28 Aug 2024
Viewed by 1469
Abstract
The DNA damage response protein p53-binding protein 1 (53BP1) accumulates and forms foci at double-strand DNA breaks, indicating the extent of DNA instability. However, the potential role of 53BP1 as a molecular biomarker for hypopharyngeal squamous cell carcinoma (HPSCC) diagnosis remains unknown. Here, [...] Read more.
The DNA damage response protein p53-binding protein 1 (53BP1) accumulates and forms foci at double-strand DNA breaks, indicating the extent of DNA instability. However, the potential role of 53BP1 as a molecular biomarker for hypopharyngeal squamous cell carcinoma (HPSCC) diagnosis remains unknown. Here, we evaluated the potential of immunofluorescence-based analysis of 53BP1 expression to differentiate the histology of hypopharyngeal neoplasms. A total of 125 lesions from 39 surgically or endoscopically resected specimens from patients with HPSCC was histologically evaluated. 53BP1 expression in the nucleus was examined using immunofluorescence. The number of 53BP1 nuclear foci increased with the progression from non-tumorous to low-grade dysplasia, high-grade dysplasia, and squamous cell carcinoma. Unstable 53BP1 expression served as an independent factor for distinguishing lesions that required intervention. Colocalization of 53BP1 foci in proliferating cells, as assessed by Ki67, was increased in tumors ≥ 1000 µm in depth compared to those <1000 µm in depth at the tumor surface. Hence, the expression patterns of nuclear 53BP1 foci were associated with the progression of hypopharyngeal neoplasms. These findings suggest that 53BP1 could serve as an ancillary marker to support histological diagnosis and predict the factors that influence prognosis in patients with HPSCC. Full article
(This article belongs to the Section Cancer Biomarkers)
Show Figures

Figure 1

19 pages, 4269 KB  
Article
Visual Evidence for the Recruitment of Four Enzymes with RNase Activity to the Bacillus subtilis Replication Forks
by Rebecca Hinrichs and Peter L. Graumann
Cells 2024, 13(16), 1381; https://doi.org/10.3390/cells13161381 - 20 Aug 2024
Cited by 2 | Viewed by 1774
Abstract
Removal of RNA/DNA hybrids for the maturation of Okazaki fragments on the lagging strand, or due to misincorporation of ribonucleotides by DNA polymerases, is essential for all types of cells. In prokaryotic cells such as Escherichia coli, DNA polymerase 1 and RNase [...] Read more.
Removal of RNA/DNA hybrids for the maturation of Okazaki fragments on the lagging strand, or due to misincorporation of ribonucleotides by DNA polymerases, is essential for all types of cells. In prokaryotic cells such as Escherichia coli, DNA polymerase 1 and RNase HI are supposed to remove RNA from Okazaki fragments, but many bacteria lack HI-type RNases, such as Bacillus subtilis. Previous work has demonstrated in vitro that four proteins are able to remove RNA from RNA/DNA hybrids, but their actual contribution to DNA replication is unclear. We have studied the dynamics of DNA polymerase A (similar to Pol 1), 5′->3′ exonuclease ExoR, and the two endoribonucleases RNase HII and HIII in B. subtilis using single-molecule tracking. We found that all four enzymes show a localization pattern similar to that of replicative DNA helicase. By scoring the distance of tracks to replication forks, we found that all four enzymes are enriched at DNA replication centers. After inducing UV damage, RNase HIII was even more strongly recruited to the replication forks, and PolA showed a more static behavior, indicative of longer binding events, whereas RNase HII and ExoR showed no response. Inhibition of replication by 6(p hydroxyphenylazo)-uracil (HPUra) demonstrated that both RNase HII and RNase HIII are directly involved in the replication. We found that the absence of ExoR increases the likelihood of RNase HIII at the forks, indicating that substrate availability rather than direct protein interactions may be a major driver for the recruitment of RNases to the lagging strands. Thus, B. subtilis replication forks appear to be an intermediate between E. coli type and eukaryotic replication forks and employ a multitude of RNases, rather than any dedicated enzyme for RNA/DNA hybrid removal. Full article
Show Figures

Graphical abstract

20 pages, 6027 KB  
Article
Promising Effects of Novel Supplement Formulas in Preventing Skin Aging in 3D Human Keratinocytes
by Angela Punzo, Matteo Perillo, Alessia Silla, Marco Malaguti, Silvana Hrelia, Diogo Barardo, Cristiana Caliceti and Antonello Lorenzini
Nutrients 2024, 16(16), 2770; https://doi.org/10.3390/nu16162770 - 20 Aug 2024
Cited by 3 | Viewed by 8346
Abstract
Dietary intervention is considered a safe preventive strategy to slow down aging. This study aimed to evaluate the protective effects of a commercially available supplement and six simpler formulations against DNA damage in 3D human keratinocytes. The ingredients used are well known and [...] Read more.
Dietary intervention is considered a safe preventive strategy to slow down aging. This study aimed to evaluate the protective effects of a commercially available supplement and six simpler formulations against DNA damage in 3D human keratinocytes. The ingredients used are well known and were combined into various formulations to test their potential anti-aging properties. Firstly, we determined the formulations’ safe concentration by evaluating cytotoxicity and cell viability through spectrophotometric assays. We then examined the presence of tumor p53 binding protein 1 and phosphorylated histone H2AX foci, which are markers of genotoxicity. The foci count revealed that a 24-h treatment with the supplement did not induce DNA damage, and significantly reduced DNA damage in cells exposed to neocarzinostatin for 2 h. Three of the simpler formulations showed similar results. Moreover, the antioxidant activity was tested using a recently developed whole cell-based chemiluminescent bioassay; results showed that a 24-h treatment with the supplement and three simpler formulations significantly reduced intracellular H2O2 after pro-oxidant injury, thus suggesting their possible antiaging effect. This study’s originality lies in the use of a 3D human keratinocyte cell model and a combination of natural ingredients targeting DNA damage and oxidative stress, providing a robust evaluation of their anti-aging potential. Full article
(This article belongs to the Section Nutrition and Public Health)
Show Figures

Figure 1

13 pages, 13343 KB  
Article
Nanoscale Interaction of Endonuclease APE1 with DNA
by Sridhar Vemulapalli, Mohtadin Hashemi, Yingling Chen, Suravi Pramanik, Kishor K. Bhakat and Yuri L. Lyubchenko
Int. J. Mol. Sci. 2024, 25(10), 5145; https://doi.org/10.3390/ijms25105145 - 9 May 2024
Viewed by 1884
Abstract
Apurinic/apyrimidinic endonuclease 1 (APE1) is involved in DNA repair and transcriptional regulation mechanisms. This multifunctional activity of APE1 should be supported by specific structural properties of APE1 that have not yet been elucidated. Herein, we applied atomic force microscopy (AFM) to characterize the [...] Read more.
Apurinic/apyrimidinic endonuclease 1 (APE1) is involved in DNA repair and transcriptional regulation mechanisms. This multifunctional activity of APE1 should be supported by specific structural properties of APE1 that have not yet been elucidated. Herein, we applied atomic force microscopy (AFM) to characterize the interactions of APE1 with DNA containing two well-separated G-rich segments. Complexes of APE1 with DNA containing G-rich segments were visualized, and analysis of the complexes revealed the affinity of APE1 to G-rich DNA sequences, and their yield was as high as 53%. Furthermore, APE1 is capable of binding two DNA segments leading to the formation of loops in the DNA–APE1 complexes. The analysis of looped APE1-DNA complexes revealed that APE1 can bridge G-rich segments of DNA. The yield of loops bridging two G-rich DNA segments was 41%. Analysis of protein size in various complexes was performed, and these data showed that loops are formed by APE1 monomer, suggesting that APE1 has two DNA binding sites. The data led us to a model for the interaction of APE1 with DNA and the search for the specific sites. The implication of these new APE1 properties in organizing DNA, by bringing two distant sites together, for facilitating the scanning for damage and coordinating repair and transcription is discussed. Full article
(This article belongs to the Collection Feature Papers in Molecular Nanoscience)
Show Figures

Figure 1

Back to TopTop