1
|
Duan Y, Yang F, Zhang Y, Zhang M, Shi Y, Lang Y, Sun H, Wang X, Jin H, Kang X. Role of mitophagy in spinal cord ischemia-reperfusion injury. Neural Regen Res 2026; 21:598-611. [PMID: 39665804 DOI: 10.4103/nrr.nrr-d-24-00668] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Accepted: 10/29/2024] [Indexed: 12/13/2024] Open
Abstract
Spinal cord ischemia-reperfusion injury, a severe form of spinal cord damage, can lead to sensory and motor dysfunction. This injury often occurs after traumatic events, spinal cord surgeries, or thoracoabdominal aortic surgeries. The unpredictable nature of this condition, combined with limited treatment options, poses a significant burden on patients, their families, and society. Spinal cord ischemia-reperfusion injury leads to reduced neuronal regenerative capacity and complex pathological processes. In contrast, mitophagy is crucial for degrading damaged mitochondria, thereby supporting neuronal metabolism and energy supply. However, while moderate mitophagy can be beneficial in the context of spinal cord ischemia-reperfusion injury, excessive mitophagy may be detrimental. Therefore, this review aims to investigate the potential mechanisms and regulators of mitophagy involved in the pathological processes of spinal cord ischemia-reperfusion injury. The goal is to provide a comprehensive understanding of recent advancements in mitophagy related to spinal cord ischemia-reperfusion injury and clarify its potential clinical applications.
Collapse
Affiliation(s)
- Yanni Duan
- Department of Orthopedics, The Second Hospital of Lanzhou University, Lanzhou, Gansu Province, China
- The Second Clinical Medical School, Lanzhou University, Lanzhou, Gansu Province, China
- Orthopaedics Key Laboratory of Gansu Province, The Second Hospital of Lanzhou University, Lanzhou, Gansu Province, China
| | - Fengguang Yang
- Department of Orthopedics, The Second Hospital of Lanzhou University, Lanzhou, Gansu Province, China
- The Second Clinical Medical School, Lanzhou University, Lanzhou, Gansu Province, China
- Orthopaedics Key Laboratory of Gansu Province, The Second Hospital of Lanzhou University, Lanzhou, Gansu Province, China
| | - Yibao Zhang
- Department of Orthopedics, The Second Hospital of Lanzhou University, Lanzhou, Gansu Province, China
- The Second Clinical Medical School, Lanzhou University, Lanzhou, Gansu Province, China
- Orthopaedics Key Laboratory of Gansu Province, The Second Hospital of Lanzhou University, Lanzhou, Gansu Province, China
| | - Mingtao Zhang
- Department of Orthopedics, The Second Hospital of Lanzhou University, Lanzhou, Gansu Province, China
- The Second Clinical Medical School, Lanzhou University, Lanzhou, Gansu Province, China
- Orthopaedics Key Laboratory of Gansu Province, The Second Hospital of Lanzhou University, Lanzhou, Gansu Province, China
| | - Yujun Shi
- Department of Orthopedics, The Second Hospital of Lanzhou University, Lanzhou, Gansu Province, China
- The Second Clinical Medical School, Lanzhou University, Lanzhou, Gansu Province, China
- Orthopaedics Key Laboratory of Gansu Province, The Second Hospital of Lanzhou University, Lanzhou, Gansu Province, China
| | - Yun Lang
- Department of Orthopedics, The Second Hospital of Lanzhou University, Lanzhou, Gansu Province, China
- The Second Clinical Medical School, Lanzhou University, Lanzhou, Gansu Province, China
- Orthopaedics Key Laboratory of Gansu Province, The Second Hospital of Lanzhou University, Lanzhou, Gansu Province, China
| | - Hongli Sun
- Department of Orthopedics, The Second Hospital of Lanzhou University, Lanzhou, Gansu Province, China
- The Second Clinical Medical School, Lanzhou University, Lanzhou, Gansu Province, China
- Orthopaedics Key Laboratory of Gansu Province, The Second Hospital of Lanzhou University, Lanzhou, Gansu Province, China
| | - Xin Wang
- Department of Orthopedics, The Second Hospital of Lanzhou University, Lanzhou, Gansu Province, China
- The Second Clinical Medical School, Lanzhou University, Lanzhou, Gansu Province, China
- Orthopaedics Key Laboratory of Gansu Province, The Second Hospital of Lanzhou University, Lanzhou, Gansu Province, China
| | - Hongyun Jin
- Department of Orthopedics, The Second Hospital of Lanzhou University, Lanzhou, Gansu Province, China
- The Second Clinical Medical School, Lanzhou University, Lanzhou, Gansu Province, China
- Orthopaedics Key Laboratory of Gansu Province, The Second Hospital of Lanzhou University, Lanzhou, Gansu Province, China
| | - Xuewen Kang
- Department of Orthopedics, The Second Hospital of Lanzhou University, Lanzhou, Gansu Province, China
- The Second Clinical Medical School, Lanzhou University, Lanzhou, Gansu Province, China
- Orthopaedics Key Laboratory of Gansu Province, The Second Hospital of Lanzhou University, Lanzhou, Gansu Province, China
| |
Collapse
|
2
|
Desai M, Gulati K, Agrawal M, Ghumra S, Sahoo PK. Stress granules: Guardians of cellular health and triggers of disease. Neural Regen Res 2026; 21:588-597. [PMID: 39995077 DOI: 10.4103/nrr.nrr-d-24-01196] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2024] [Accepted: 01/15/2025] [Indexed: 02/26/2025] Open
Abstract
Stress granules are membraneless organelles that serve as a protective cellular response to external stressors by sequestering non-translating messenger RNAs (mRNAs) and regulating protein synthesis. Stress granules formation mechanism is conserved across species, from yeast to mammals, and they play a critical role in minimizing cellular damage during stress. Composed of heterogeneous ribonucleoprotein complexes, stress granules are enriched not only in mRNAs but also in noncoding RNAs and various proteins, including translation initiation factors and RNA-binding proteins. Genetic mutations affecting stress granule assembly and disassembly can lead to abnormal stress granule accumulation, contributing to the progression of several diseases. Recent research indicates that stress granule dynamics are pivotal in determining their physiological and pathological functions, with acute stress granule formation offering protection and chronic stress granule accumulation being detrimental. This review focuses on the multifaceted roles of stress granules under diverse physiological conditions, such as regulation of mRNA transport, mRNA translation, apoptosis, germ cell development, phase separation processes that govern stress granule formation, and their emerging implications in pathophysiological scenarios, such as viral infections, cancer, neurodevelopmental disorders, neurodegeneration, and neuronal trauma.
Collapse
Affiliation(s)
- Meghal Desai
- Department of Biological Sciences, Rutgers University - Newark, Newark, NJ, USA
| | - Keya Gulati
- College of Science and Liberal Arts, New Jersey Institute of Technology, Newark, NJ, USA
| | - Manasi Agrawal
- Department of Biological Sciences, Rutgers University - Newark, Newark, NJ, USA
| | - Shruti Ghumra
- Department of Biological Sciences, Rutgers University - Newark, Newark, NJ, USA
| | - Pabitra K Sahoo
- Department of Biological Sciences, Rutgers University - Newark, Newark, NJ, USA
| |
Collapse
|
3
|
Chen X, Lai J, Wu Z, Chen J, Yang B, Chen C, Ding C. Fat mass and obesity-mediated N 6 -methyladenosine modification modulates neuroinflammatory responses after traumatic brain injury. Neural Regen Res 2026; 21:730-741. [PMID: 39248160 DOI: 10.4103/nrr.nrr-d-23-01854] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2023] [Accepted: 05/24/2024] [Indexed: 09/10/2024] Open
Abstract
JOURNAL/nrgr/04.03/01300535-202602000-00042/figure1/v/2025-05-05T160104Z/r/image-tiff The neuroinflammatory response mediated by microglial activation plays an important role in the secondary nerve injury of traumatic brain injury. The post-transcriptional modification of N 6 -methyladenosine is ubiquitous in the immune response of the central nervous system. The fat mass and obesity-related protein catalyzes the demethylation of N 6 -methyladenosine modifications on mRNA and is widely expressed in various tissues, participating in the regulation of multiple diseases' biological processes. However, the role of fat mass and obesity in microglial activation and the subsequent neuroinflammatory response after traumatic brain injury is unclear. In this study, we found that the expression of fat mass and obesity was significantly down-regulated in both lipopolysaccharide-treated BV2 cells and a traumatic brain injury mouse model. After fat mass and obesity interference, BV2 cells exhibited a pro-inflammatory phenotype as shown by the increased proportion of CD11b + /CD86 + cells and the secretion of pro-inflammatory cytokines. Fat mass and obesity-mediated N 6 -methyladenosine demethylation accelerated the degradation of ADAM17 mRNA, while silencing of fat mass and obesity enhanced the stability of ADAM17 mRNA. Therefore, down-regulation of fat mass and obesity expression leads to the abnormally high expression of ADAM17 in microglia. These results indicate that the activation of microglia and neuroinflammatory response regulated by fat mass and obesity-related N 6 -methyladenosine modification plays an important role in the pro-inflammatory process of secondary injury following traumatic brain injury.
Collapse
Affiliation(s)
- Xiangrong Chen
- Department of Neurosurgery, Second Clinical Medical College, Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian Province, China
| | - Jinqing Lai
- Department of Neurosurgery, Second Clinical Medical College, Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian Province, China
| | - Zhe Wu
- Department of Neurosurgery, Second Clinical Medical College, Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian Province, China
| | - Jianlong Chen
- Department of Neurosurgery, Second Clinical Medical College, Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian Province, China
| | - Baoya Yang
- Department of Neurosurgery, Second Clinical Medical College, Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian Province, China
| | - Chunnuan Chen
- Department of Neurology, Second Clinical Medical College, Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian Province, China
| | - Chenyu Ding
- Department of Neurosurgery, Neurosurgery Research Institute, National Regional Medical Center, Binhai Campus, First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian Province, China
| |
Collapse
|
4
|
Shahin R, Jaafreh S, Azzam Y. Tracking protein kinase targeting advances: integrating QSAR into machine learning for kinase-targeted drug discovery. Future Sci OA 2025; 11:2483631. [PMID: 40181786 PMCID: PMC11980485 DOI: 10.1080/20565623.2025.2483631] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Accepted: 03/06/2025] [Indexed: 04/05/2025] Open
Abstract
Protein kinases are vital drug targets, yet designing selective inhibitors is challenging, compounded by resistance and kinome complexity. This review explores Quantitative Structure-Activity Relationship (QSAR) modeling for kinase drug discovery, focusing on integrating traditional QSAR with machine learning (ML)-CNNs, RNNs-and structural data. Methods include structural databases, docking, and deep learning QSAR. Key findings show ML-integrated QSAR significantly improves selective inhibitor design for CDKs, JAKs, PIM kinases. The IDG-DREAM challenge exemplifies ML's potential for accurate kinase-inhibitor interaction prediction, outperforming traditional methods and enabling inhibitors with enhanced selectivity, efficacy, and resistance mitigation. QSAR combined with advanced computation and experimental data accelerates kinase drug discovery, offering transformative precision medicine potential. This review highlights deep learning-enhanced QSAR's novelty in automating feature extraction and capturing complex relationships, surpassing traditional QSAR, while emphasizing interpretability and experimental validation for clinical translation.
Collapse
Affiliation(s)
- Rand Shahin
- Drug Design Unit, Department of Pharmaceutical Chemistry, Hashemite University, Zarqa, Jordan
| | - Sawsan Jaafreh
- Department of Chemistry, The Hashemite University, Zarqa, Jordan
| | - Yusra Azzam
- Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY, USA
| |
Collapse
|
5
|
Wang Y, Yu X, Sun F, Fu Y, Hu T, Shi Q, Man Q. METTL14 Mediates Glut3 m6A methylation to improve osteogenesis under oxidative stress condition. Redox Rep 2025; 30:2435241. [PMID: 39737912 DOI: 10.1080/13510002.2024.2435241] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2025] Open
Abstract
OBJECTIVES Bone remodeling imbalance contributes to osteoporosis. Though current medications enhance osteoblast involvement in bone formation, the underlying pathways remain unclear. This study was aimed to explore the pathways involved in bone formation by osteoblasts, we investigate the protective role of glycolysis and N6-methyladenosine methylation (m6A) against oxidative stress-induced impairment of osteogenesis in MC3T3-E1 cells. METHODS We utilized a concentration of 200 μM hydrogen peroxide (H2O2) to establish an oxidative damage model of MC3T3-E1 cells. Subsequently, we examined the alterations in the m6A methyltransferases (METTL3, METTL14), glucose transporter proteins (GLUT1, GLUT3) and validated m6A methyltransferase overexpression in vitro and in an osteoporosis model. The osteoblast differentiation and osteogenesis-related molecules and serum bone resorption markers were measured by biochemical analysis, Alizarin Red S staining, Western blot and ELISA. RESULTS H2O2 treatment inhibited glycolysis and osteoblast differentiation in MC3T3-E1 cells. However, when METTL14 was overexpressed, these changes induced by H2O2 could be mitigated. Our findings indicate that METTL14 promotes GLUT3 expression via YTHDF1, leading to the modulation of various parameters in the H2O2-induced model. Similar positive effects of METTL14 on osteogenesis were observed in an ovariectomized mouse osteoporosis model. DISCUSSION METTL14 could serve as a potential therapeutic approach for enhancing osteoporosis treatment.
Collapse
Affiliation(s)
- Ying Wang
- Department of Clinical Laboratory, Shanghai Fourth People's Hospital, School of Medicine, Tongji University, Shanghai, People's Republic of China
| | - Xueying Yu
- Department of Clinical Laboratory, Shanghai Fourth People's Hospital, School of Medicine, Tongji University, Shanghai, People's Republic of China
| | - Fenyong Sun
- Department of Clinical Laboratory, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, People's Republic of China
| | - Yan Fu
- Department of Clinical Laboratory, Shanghai Fourth People's Hospital, School of Medicine, Tongji University, Shanghai, People's Republic of China
| | - Tingting Hu
- Department of Clinical Laboratory, Shanghai Fourth People's Hospital, School of Medicine, Tongji University, Shanghai, People's Republic of China
| | - Qiqing Shi
- Department of Anesthesiology, Minhang Hospital, Fudan University, Shanghai, People's Republic of China
| | - Qiuhong Man
- Department of Clinical Laboratory, Shanghai Fourth People's Hospital, School of Medicine, Tongji University, Shanghai, People's Republic of China
| |
Collapse
|
6
|
Lin C, Hsu JL, Hsu YT, Fan KC, Wu SS, Lin MH, Guh JH, Yu CW. Design and synthesis of novel HDAC6 inhibitor dimer as HDAC6 degrader for cancer treatment by palladium catalysed dimerisation. J Enzyme Inhib Med Chem 2025; 40:2468355. [PMID: 40013582 PMCID: PMC11869342 DOI: 10.1080/14756366.2025.2468355] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2024] [Revised: 02/07/2025] [Accepted: 02/11/2025] [Indexed: 02/28/2025] Open
Abstract
The enigmatic histone deacetylase 6 (HDAC6) is one of a kind among its family. Recent reports revealed that HDAC6 CD1 exhibits E3 ligase activity. Inspired by these researches, we attempted to develop drugs targeting HDAC6 via novel mechanism. Herein, we report a palladium catalysed transformation and purification method for hydroxamic acid dimers, and series of HDAC6 inhibitor-based dimer showing outstanding biological activities and capability of inducing auto-degradation. Our proof-of-concept was highlighted with 2-amino benzamide-based HDAC6 inhibitor dimers that exhibit great HDAC6 inhibition activity (3.9-15.4 nM), good HDAC1/6 selectivity (95-577), and excellent cytotoxicity against human hormone-resistant prostate cancer (HRPC) PC-3 and non-small cell lung cancer (NSCLC) A549 cell lines (5.9-11.3 and 6.6-17.9 μM, respectively) while simultaneously inducing HDAC6 degradation. These dimers not only induce apoptosis and autophagy but also interfere with kinetochore attachment by the detection of BUBR1 phosphorylation at S670.
Collapse
Affiliation(s)
- Ching Lin
- School of Pharmacy, National Taiwan University, Taipei, ROC
| | - Jui-Ling Hsu
- School of Pharmacy, National Taiwan University, Taipei, ROC
- Department of Nursing, Chang Gung University of Science and Technology, Taoyuan City, ROC
- Division of Hematology-Oncology, Department of Internal Medicine, Chang Gung Memorial Hospital, Linkou Medical Center, Taoyuan City, ROC
| | - Yu-Tung Hsu
- School of Pharmacy, National Taiwan University, Taipei, ROC
| | - Kuo-Chen Fan
- School of Pharmacy, National Taiwan University, Taipei, ROC
| | - Sian-Siou Wu
- School of Pharmacy, National Taiwan University, Taipei, ROC
| | - Miao-Hsia Lin
- Department and Graduate Institute of Medical Microbiology, College of Medicine, National Taiwan University, Taipei, ROC
| | - Jih-Hwa Guh
- School of Pharmacy, National Taiwan University, Taipei, ROC
| | - Chao-Wu Yu
- School of Pharmacy, National Taiwan University, Taipei, ROC
| |
Collapse
|
7
|
Wu S, Ma X, Zhang X, Du K, Shi C, Almaamari AA, Han B, Su S, Liu Y. Knockdown of NDUFAF6 inhibits breast cancer progression via promoting mitophagy and apoptosis. Cancer Biol Ther 2025; 26:2445220. [PMID: 39706687 DOI: 10.1080/15384047.2024.2445220] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2024] [Revised: 12/04/2024] [Accepted: 12/17/2024] [Indexed: 12/23/2024] Open
Abstract
BACKGROUND While NDUFAF6 is implicated in breast cancer, its specific role remains unclear. METHODS The expression levels and prognostic significance of NDUFAF6 in breast cancer were assessed using The Cancer Genome Atlas, Gene Expression Omnibus, Kaplan-Meier plotter and cBio-Portal databases. We knocked down NDUFAF6 in breast cancer cells using small interfering RNA and investigated its effects on cell proliferation and migration ability. We performed gene expression analysis and validated key findings using protein analysis. We also assessed mitochondrial activity and cellular metabolism. RESULTS NDUFAF6 was highly expressed in breast cancer, which was associated with a poorer prognosis. Knockdown of NDUFAF6 reduced the proliferation and migration ability of breast cancer cells. Transcriptome analysis revealed 2,101 differentially expressed genes enriched in apoptosis and mitochondrial signaling pathways. Western blot results showed NDUFAF6 knockdown enhanced apoptosis. In addition, differential gene enrichment analysis was related to mitochondrial signaling pathways, and western blot results verified that mitophagy was enhanced in NDUFAF6 knockdown breast cancer cells. JC-1 assay also showed that mitochondrial dysfunction and reactive oxygen species content were increased after knocking down NDUFAF6. In addition, basal and maximal mitochondrial oxygen consumption decreased, and intracellular glycogen content increased. CONCLUSIONS Knockdown of NDUFAF6 resulted in apoptosis and mitophagy in breast cancer cells and NDUFAF6 may be a potential molecular target for breast cancer therapy.
Collapse
Affiliation(s)
- Shang Wu
- Department of Breast Center, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
- Hebei Provincial Key Laboratory of Tumor Microenvironment and Drug Resistance, Hebei Medical University, Shijiazhuang, China
| | - Xindi Ma
- Department of Breast Center, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
- Hebei Provincial Key Laboratory of Tumor Microenvironment and Drug Resistance, Hebei Medical University, Shijiazhuang, China
| | - Xiangmei Zhang
- Hebei Provincial Key Laboratory of Tumor Microenvironment and Drug Resistance, Hebei Medical University, Shijiazhuang, China
- Department of Research Center, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Kaiye Du
- Hebei Provincial Key Laboratory of Tumor Microenvironment and Drug Resistance, Hebei Medical University, Shijiazhuang, China
- Radiotherapy Department, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Chao Shi
- Department of Breast Center, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
- Hebei Provincial Key Laboratory of Tumor Microenvironment and Drug Resistance, Hebei Medical University, Shijiazhuang, China
| | - Ahmed Ali Almaamari
- The Key Laboratory of Neural and Vascular Biology, The Key Laboratory of New Drug Pharmacology and Toxicology, Department of Pharmacology, Ministry of Education, Hebei Medical University, Shijiazhuang, China
| | - Boye Han
- The Key Laboratory of Neural and Vascular Biology, The Key Laboratory of New Drug Pharmacology and Toxicology, Department of Pharmacology, Ministry of Education, Hebei Medical University, Shijiazhuang, China
| | - Suwen Su
- The Key Laboratory of Neural and Vascular Biology, The Key Laboratory of New Drug Pharmacology and Toxicology, Department of Pharmacology, Ministry of Education, Hebei Medical University, Shijiazhuang, China
| | - Yunjiang Liu
- Hebei Provincial Key Laboratory of Tumor Microenvironment and Drug Resistance, Hebei Medical University, Shijiazhuang, China
- The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| |
Collapse
|
8
|
Hiruthyaswamy SP, Bose A, Upadhyay A, Raha T, Bhattacharjee S, Singha I, Ray S, Nicky Macarius NM, Viswanathan P, Deepankumar K. Molecular signaling pathways in osteoarthritis and biomaterials for cartilage regeneration: a review. Bioengineered 2025; 16:2501880. [PMID: 40336219 PMCID: PMC12064066 DOI: 10.1080/21655979.2025.2501880] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2024] [Revised: 03/07/2025] [Accepted: 04/04/2025] [Indexed: 05/09/2025] Open
Abstract
Osteoarthritis is a prevalent degenerative joint disease characterized by cartilage degradation, synovial inflammation, and subchondral bone alterations, leading to chronic pain and joint dysfunction. Conventional treatments provide symptomatic relief but fail to halt disease progression. Recent advancements in biomaterials, molecular signaling modulation, and gene-editing technologies offer promising therapeutic strategies. This review explores key molecular pathways implicated in osteoarthritis, including fibroblast growth factor, phosphoinositide 3-kinase/Akt, and bone morphogenetic protein signaling, highlighting their roles in chondrocyte survival, extracellular matrix remodeling, and inflammation. Biomaterial-based interventions such as hydrogels, nanoparticles, and chitosan-based scaffolds have demonstrated potential in enhancing cartilage regeneration and targeted drug delivery. Furthermore, CRISPR/Cas9 gene editing holds promise in modifying osteoarthritis-related genes to restore cartilage integrity. The integration of regenerative biomaterials with precision medicine and molecular therapies represents a novel approach for mitigating osteoarthritis progression. Future research should focus on optimizing biomaterial properties, refining gene-editing efficiency, and developing personalized therapeutic strategies. The convergence of bioengineering and molecular science offers new hope for improving joint function and patient quality of life in osteoarthritis management.
Collapse
Affiliation(s)
- Samson Prince Hiruthyaswamy
- Department of Biotechnology, School of Biosciences and Technology, Vellore Institute of Technology, Vellore, India
| | - Arohi Bose
- Department of Biotechnology, School of Biosciences and Technology, Vellore Institute of Technology, Vellore, India
| | - Ayushi Upadhyay
- Department of Biotechnology, School of Biosciences and Technology, Vellore Institute of Technology, Vellore, India
| | - Tiasa Raha
- Department of Biotechnology, School of Biosciences and Technology, Vellore Institute of Technology, Vellore, India
| | - Shangomitra Bhattacharjee
- Department of Biotechnology, School of Biosciences and Technology, Vellore Institute of Technology, Vellore, India
| | - Isheeta Singha
- Department of Biotechnology, School of Biosciences and Technology, Vellore Institute of Technology, Vellore, India
| | - Swati Ray
- Department of Biotechnology, School of Biosciences and Technology, Vellore Institute of Technology, Vellore, India
| | | | - Pragasam Viswanathan
- Department of Biotechnology, School of Biosciences and Technology, Vellore Institute of Technology, Vellore, India
| | - Kanagavel Deepankumar
- Department of Biotechnology, School of Biosciences and Technology, Vellore Institute of Technology, Vellore, India
| |
Collapse
|
9
|
Yu J, Tang H, Chen Y, Wang Z, Huang W, Zhou T, Wen B, Wang C, Gu S, Ni J, Tao J, Wang D, Lu J, Xie Q, Yao YF. Salmonella utilizes L-arabinose to silence virulence gene expression for accelerated pathogen growth within the host. Gut Microbes 2025; 17:2467187. [PMID: 39954030 PMCID: PMC11834461 DOI: 10.1080/19490976.2025.2467187] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/07/2024] [Revised: 01/11/2025] [Accepted: 02/05/2025] [Indexed: 02/17/2025] Open
Abstract
Carbon source is an important nutrient for bacteria to sustain growth and often acts as a signal that modulates virulence expression. L-arabinose is produced by plants and plays an important role in regulating the global gene expression of bacteria. Previously, we have shown that L-arabinose induces a more severe systemic infection in Salmonella-infected mice with normal microbiota, but does not affect the disease progression in mice with microbiota depleted by antibiotic treatment. The underlying mechanism remains elusive. In this study, we demonstrate that L-arabinose represses the expression of Salmonella type III secretion system 1 (T3SS-1) genes by negatively regulating the activity of the cyclic 3' 5'-AMP (cAMP)-cAMP receptor protein (CRP) complex. The cAMP-CRP complex can activate ribosome-associated inhibitor A, encoded by yfiA, to maintain the stability of HilD, a key transcriptional regulator of T3SS-1. L-arabinose supplementation promotes Salmonella initial bloom in the antibiotic-pretreated mouse gut and ultimately compensates for reduced virulence within the host. These results decipher the molecular mechanism by which cAMP-CRP directs regulatory changes of virulence in response to L-arabinose in Salmonella. It further implies that Salmonella exploits L-arabinose both as a nutrient and a regulatory signal to maintain a balance between growth and virulence within the host.
Collapse
Affiliation(s)
- Jingchen Yu
- Department of Infectious Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Huang Tang
- Laboratory of Bacterial Pathogenesis, Shanghai Institute of Immunology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yana Chen
- Department of Pediatrics, Anhui Provincial Hospital, The First Affiliated Hospital of USTC, Hefei, Anhui, China
| | - Zuoqiang Wang
- Laboratory of Bacterial Pathogenesis, Shanghai Institute of Immunology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Wanqiu Huang
- Laboratory of Bacterial Pathogenesis, Shanghai Institute of Immunology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Tao Zhou
- Laboratory of Bacterial Pathogenesis, Shanghai Institute of Immunology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Bingjie Wen
- Laboratory of Bacterial Pathogenesis, Shanghai Institute of Immunology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Chengyue Wang
- Laboratory of Bacterial Pathogenesis, Shanghai Institute of Immunology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Shuang Gu
- Laboratory of Bacterial Pathogenesis, Shanghai Institute of Immunology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jinjing Ni
- Laboratory of Bacterial Pathogenesis, Shanghai Institute of Immunology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jing Tao
- Laboratory of Bacterial Pathogenesis, Shanghai Institute of Immunology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Danni Wang
- Laboratory of Bacterial Pathogenesis, Shanghai Institute of Immunology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jie Lu
- Department of Infectious Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Qing Xie
- Department of Infectious Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yu-Feng Yao
- Laboratory of Bacterial Pathogenesis, Shanghai Institute of Immunology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- State Key Laboratory of Microbial Metabolism, and School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, China
- Diagnosis and Treatment of Respiratory Infectious Diseases (20dz2261100), Shanghai Key Laboratory of Emergency Prevention, Shanghai, China
| |
Collapse
|
10
|
Wang S, Li X, Ma J, Duan X, Wang H, Wang L, Hu D, Jiang W, Li X, Qian P. Structural and functional analysis reveals the catalytic mechanism and substrate binding mode of the broad-spectrum endolysin Ply2741. Virulence 2025; 16:2449025. [PMID: 39810299 PMCID: PMC11740692 DOI: 10.1080/21505594.2024.2449025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Revised: 12/09/2024] [Accepted: 12/29/2024] [Indexed: 01/16/2025] Open
Abstract
The emergence of antibiotic-resistant bacteria has attracted interest in the field of endolysins. Here, we analyzed the diversity of Streptococcus endolysins and identified a new endolysin, Ply2741, that exhibited broad-spectrum bactericidal activity. Our results demonstrated that Ply2741 could effectively eradicate multidrug-resistant gram-positive pathogens in vitro and in vivo. Structural analysis revealed that the bactericidal activity of Ply2741 depends on the classic "Cys-His-Asn" catalytic triad. Site-directed mutagenesis results further identified that the conserved residue Gln29, located near the catalytic triad, also contributes to the lytic activity of Ply2741. Furthermore, the key residues (R189 and W250) in the Ply2741 cell wall binding domain (CBD) responsible for binding to peptidoglycan were revealed by molecular docking and fluorescence-activated cell sorting (FACS) analysis. Ply2741 demonstrates a broad lytic spectrum, with significant bactericidal activity against Enterococcus, Staphylococcus, and Streptococcus and species. To the best of our knowledge, we found that residue Gln29 participated in the lytic activity of endolysin for the first time. Additionally, we systematically elucidate the binding mode and key residues of the Ply2741CBD. This study proposes Ply2741 as a potential antibiotic substitute and provides a structural basis for the modification and design of endolysins.
Collapse
Affiliation(s)
- Shuang Wang
- National Key Laboratory of Agricultural Microbiology, Hubei Hongshan Laboratory, Huazhong Agricultural University, Wuhan, China
- The Cooperative Innovation Centre for Sustainable Pig Production, Huazhong Agricultural University, Wuhan, China
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Xinxin Li
- National Key Laboratory of Agricultural Microbiology, Hubei Hongshan Laboratory, Huazhong Agricultural University, Wuhan, China
- The Cooperative Innovation Centre for Sustainable Pig Production, Huazhong Agricultural University, Wuhan, China
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Jiahui Ma
- National Key Laboratory of Agricultural Microbiology, Hubei Hongshan Laboratory, Huazhong Agricultural University, Wuhan, China
- The Cooperative Innovation Centre for Sustainable Pig Production, Huazhong Agricultural University, Wuhan, China
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Xiaochao Duan
- National Key Laboratory of Agricultural Microbiology, Hubei Hongshan Laboratory, Huazhong Agricultural University, Wuhan, China
- The Cooperative Innovation Centre for Sustainable Pig Production, Huazhong Agricultural University, Wuhan, China
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Haiyan Wang
- National Key Laboratory of Agricultural Microbiology, Hubei Hongshan Laboratory, Huazhong Agricultural University, Wuhan, China
- The Cooperative Innovation Centre for Sustainable Pig Production, Huazhong Agricultural University, Wuhan, China
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Linkang Wang
- National Key Laboratory of Agricultural Microbiology, Hubei Hongshan Laboratory, Huazhong Agricultural University, Wuhan, China
- The Cooperative Innovation Centre for Sustainable Pig Production, Huazhong Agricultural University, Wuhan, China
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Dayue Hu
- National Key Laboratory of Agricultural Microbiology, Hubei Hongshan Laboratory, Huazhong Agricultural University, Wuhan, China
- The Cooperative Innovation Centre for Sustainable Pig Production, Huazhong Agricultural University, Wuhan, China
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Wenwu Jiang
- National Key Laboratory of Agricultural Microbiology, Hubei Hongshan Laboratory, Huazhong Agricultural University, Wuhan, China
- The Cooperative Innovation Centre for Sustainable Pig Production, Huazhong Agricultural University, Wuhan, China
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Xiangmin Li
- National Key Laboratory of Agricultural Microbiology, Hubei Hongshan Laboratory, Huazhong Agricultural University, Wuhan, China
- The Cooperative Innovation Centre for Sustainable Pig Production, Huazhong Agricultural University, Wuhan, China
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Ping Qian
- National Key Laboratory of Agricultural Microbiology, Hubei Hongshan Laboratory, Huazhong Agricultural University, Wuhan, China
- The Cooperative Innovation Centre for Sustainable Pig Production, Huazhong Agricultural University, Wuhan, China
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| |
Collapse
|
11
|
Lee H, Park H, Kwak K, Lee CE, Yun J, Lee D, Lee JH, Lee SH, Kang LW. Structural comparison of substrate-binding pockets of serine β-lactamases in classes A, C, and D. J Enzyme Inhib Med Chem 2025; 40:2435365. [PMID: 39714271 DOI: 10.1080/14756366.2024.2435365] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Revised: 10/08/2024] [Accepted: 11/22/2024] [Indexed: 12/24/2024] Open
Abstract
β-lactams have been the most successful antibiotics, but the rise of multi-drug resistant (MDR) bacteria threatens their effectiveness. Serine β-lactamases (SBLs), among the most common causes of resistance, are classified as A, C, and D, with numerous variants complicating structural and substrate spectrum comparisons. This study compares representative SBLs of these classes, focusing on the substrate-binding pocket (SBP). SBP is kidney bean-shaped on the indented surface, formed mainly by loops L1, L2, and L3, and an additional loop Lc in class C. β-lactams bind in a conserved orientation, with the β-lactam ring towards L2 and additional rings towards the space between L1 and L3. Structural comparison shows each class has distinct SBP structures, but subclasses share a conserved scaffold. The SBP structure, accommodating complimentary β-lactams, determines the substrate spectrum of SBLs. The systematic comparison of SBLs, including structural compatibility between β-lactams and SBPs, will help understand their substrate spectrum.
Collapse
Affiliation(s)
- Hyeonmin Lee
- Department of Biological Sciences, Konkuk University, Seoul, Republic of Korea
| | - Hyunjae Park
- Department of Biological Sciences, Konkuk University, Seoul, Republic of Korea
| | - Kiwoong Kwak
- Department of Biological Sciences, Konkuk University, Seoul, Republic of Korea
| | - Chae-Eun Lee
- Department of Biological Sciences, Konkuk University, Seoul, Republic of Korea
| | - Jiwon Yun
- Department of Biological Sciences, Konkuk University, Seoul, Republic of Korea
| | - Donghyun Lee
- Department of Biological Sciences, Konkuk University, Seoul, Republic of Korea
| | - Jung Hun Lee
- National Leading Research Laboratory of Drug Resistance Proteomics, Department of Biological Sciences, Myongji University, Yongin, Republic of Korea
| | - Sang Hee Lee
- National Leading Research Laboratory of Drug Resistance Proteomics, Department of Biological Sciences, Myongji University, Yongin, Republic of Korea
| | - Lin-Woo Kang
- Department of Biological Sciences, Konkuk University, Seoul, Republic of Korea
| |
Collapse
|
12
|
Masaadeh AH, Eletrebi M, Parajuli B, De Jager N, Bosch DE. Human colitis-associated colorectal carcinoma progression is accompanied by dysbiosis with enriched pathobionts. Gut Microbes 2025; 17:2479774. [PMID: 40094201 PMCID: PMC11917176 DOI: 10.1080/19490976.2025.2479774] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/06/2025] [Revised: 02/14/2025] [Accepted: 03/10/2025] [Indexed: 03/19/2025] Open
Abstract
Dysbiosis and pathobionts contribute to inflammation and the risk of colitis-associated carcinoma (CAC) in animal models, but their roles in humans with this uncommon disease are unknown. We identified microbiome differences in human CAC compared with longstanding inflammatory bowel disease (IBD) and sporadic colorectal carcinoma (CRC). Twenty-four CAC resections were matched with CRC and IBD controls. Methods included histopathology, 16S rDNA metagenomics, and pathobiont-specific qPCR. Beta diversity differed by diagnosis (PERMANOVA p = 0.007). The distinguishing taxa included Akkermansia enriched in CRC, and Bacteroides spp. enriched in IBD. The non-neoplastic mucosae presented distinct beta diversity (p = 0.005), but the CAC/CRC tumor microbiomes were similar (p = 0.7). Within metastases and margins, Enterobacteriaceae were enriched in CAC, and Bacteroidales in CRC. Pathobiont-specific qPCR confirmed a greater frequency of pks+ E. coli and enterotoxigenic Bacteroides fragilis in CAC than IBD. High alpha diversity was associated with active inflammation, advanced cancer stage, and shorter overall survival (log-rank p = 0.008). Mucosal microbiomes distinguish CAC from longstanding IBD, implicating pathobionts as markers for disease progression. Integrating our findings with prior animal model research, pathobionts promote carcinogenesis in IBD patients through genotoxicity and host cell signaling.
Collapse
Affiliation(s)
- Amr H. Masaadeh
- Department of Pathology, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | - Mohamed Eletrebi
- Department of Pathology, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | - Bishal Parajuli
- Department of Pathology, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | - Nicola De Jager
- Department of Pathology, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | - Dustin E. Bosch
- Department of Pathology, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, IA, USA
- Holden Comprehensive Cancer Center, Iowa City, IA, USA
| |
Collapse
|
13
|
Yi J, Jiang C, Xia L. Mediated roles of oxidative stress and kidney function to leukocyte telomere length and prognosis in chronic kidney disease. Ren Fail 2025; 47:2464828. [PMID: 40011224 PMCID: PMC11866651 DOI: 10.1080/0886022x.2025.2464828] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2024] [Revised: 02/03/2025] [Accepted: 02/04/2025] [Indexed: 02/28/2025] Open
Abstract
BACKGROUND Few studies have focused on the correlation between leukocyte telomere length (LTL) and cancer-related mortality or identified potential factors that mediate the relationship between LTL and mortality among chronic kidney disease (CKD) patients. Our study aimed to explore the associations between LTL and all-cause and cause-specific mortality and to identify the underlying mediators. METHODS CKD patients were obtained from the National Health and Nutrition Examination Survey (NHANES) 1999-2002. Cox regression analysis and restricted cubic spline analysis were used to explore the associations between LTL and all-cause or specific-cause mortality and their nonlinear connections. Stratified analyses were executed to assess the relationships among the different subgroups. The latent mediated factors were confirmed using mediation analysis. Sensitivity analyses were used to evaluate the robustness of our findings. RESULTS Longer LTL associated with the lower risk of all-cause mortality, cardiovascular disease (CVD) and cancer-related mortality, and U-shaped relationships were detected. Patients younger than 65 years with greater LTL or who had hypertension had better prognoses. Age and history of hypertension were associated with LTL and overall mortality. In addition, estimated glomerular filtration rate (eGFR), albumin, and total bilirubin mediated the association, and the proportions of indirect effects were 7.81%, 3.77%, and 2.50%, respectively. Six sensitivity analyses confirmed the robustness of our findings. CONCLUSIONS This study revealed that LTL was a protective factor for survival among patients with CKD and emphasized the mediating roles of oxidative stress and kidney function.
Collapse
Affiliation(s)
- Jiahong Yi
- Department of VIP Region, Collaborative Innovation Center for Cancer Medicine, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, PR China
| | - Chang Jiang
- Department of VIP Region, Collaborative Innovation Center for Cancer Medicine, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, PR China
| | - Liangping Xia
- Department of VIP Region, Collaborative Innovation Center for Cancer Medicine, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, PR China
| |
Collapse
|
14
|
Miao M, Chen Y, Wang X, Li S, Hu R. The critical role of ferroptosis in virus-associated hematologic malignancies and its potential value in antiviral-antitumor therapy. Virulence 2025; 16:2497908. [PMID: 40302035 PMCID: PMC12045570 DOI: 10.1080/21505594.2025.2497908] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2024] [Revised: 03/06/2025] [Accepted: 04/21/2025] [Indexed: 05/01/2025] Open
Abstract
Epstein-Barr Virus (EBV), Kaposi's sarcoma-associated herpesvirus (KSHV), and human T-cell leukemia virus type 1 (HTLV-1) are key infectious agents linked to the development of various hematological malignancies, including Hodgkin's lymphoma, non-Hodgkin's lymphoma, and adult T-cell leukemia/lymphoma. This review highlights the critical knowledge gaps in understanding the role of ferroptosis, a novel form of cell death, in virus-related tumors. We focus on how ferroptosis influences the host cell response to these viral infections, revealing groundbreaking mechanisms by which the three viruses differentially regulate core pathways of ferroptosis, such as iron homeostasis, lipid peroxidation, and antioxidant systems, thereby promoting malignant transformation of host cells. Additionally, we explore the potential of antiviral drugs and ferroptosis modulators in the treatment of virus-associated hematological malignancies.
Collapse
Affiliation(s)
- Miao Miao
- Department of Hematology, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
| | - Yuelei Chen
- College of Animal Science and Veterinary Medicine, Shenyang Agricultural University, Shenyang, Liaoning, China
| | - Xuehan Wang
- Shenyang Shenhua Institute Test Technology, Shenyang, Liaoning, China
| | - Shengyang Li
- Publishing Department, Shenyang Agricultural University, Shenyang, Liaoning, China
| | - Rong Hu
- Department of Hematology, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
| |
Collapse
|
15
|
Pierre F, Baillez A, Dewitte A, Rolandelli A, Sebbane F. Proteins of the SubB family provide multiple mechanisms of serum resistance in Yersinia pestis. Emerg Microbes Infect 2025; 14:2493926. [PMID: 40237516 PMCID: PMC12064104 DOI: 10.1080/22221751.2025.2493926] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2024] [Revised: 03/21/2025] [Accepted: 04/11/2025] [Indexed: 04/18/2025]
Abstract
The serum complement system is a cornerstone element of the innate immune response. Bacterial resistance to this system is a multifaceted process involving various proteins and molecular mechanisms. Here, we report several genes required for the growth of Yersinia pestis in serum. Among them, we found that ypo0337 encodes an outer-membrane-associated lectin that recruits factor H, C4BP and hemopexin, conferring resistance to the serum complement system. YPO0337 displays high sequence similarity with the SubB subunit of the AB5 toxin from Escherichia coli, as well as other SubB-like proteins, and subB from E. coli restores the ability of Y. pestis Δypo0337 mutant to resist to serum complement. Altogether, the data suggest that at least two members of the SubB protein family function as virulence factors, conferring resistance to serum complement through a unique mode of action.
Collapse
Affiliation(s)
- François Pierre
- Univ. Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, U1019 – UMR 9017 – CIIL – Center for Infection and Immunity of Lille, Lille, France
| | - Alexandre Baillez
- Univ. Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, U1019 – UMR 9017 – CIIL – Center for Infection and Immunity of Lille, Lille, France
| | - Amélie Dewitte
- Univ. Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, U1019 – UMR 9017 – CIIL – Center for Infection and Immunity of Lille, Lille, France
| | - Agustin Rolandelli
- Univ. Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, U1019 – UMR 9017 – CIIL – Center for Infection and Immunity of Lille, Lille, France
| | - Florent Sebbane
- Univ. Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, U1019 – UMR 9017 – CIIL – Center for Infection and Immunity of Lille, Lille, France
| |
Collapse
|
16
|
Zhang C, Zhou T, Li C, Wang D, Tao J, Zhu X, Lu J, Ni J, Yao YF. Deciphering novel enzymatic and non-enzymatic lysine lactylation in Salmonella. Emerg Microbes Infect 2025; 14:2475838. [PMID: 40035788 PMCID: PMC11924271 DOI: 10.1080/22221751.2025.2475838] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2024] [Revised: 02/01/2025] [Accepted: 02/28/2025] [Indexed: 03/06/2025]
Abstract
Lysine lactylation, a novel post-translational modification, is involved in multiple cellular processes. The role of lactylation remains largely unknown, especially in bacteria. Here, we identified 1090 lactylation sites on 469 proteins by mass spectrometry in Salmonella Typhimurium. Many proteins involved in metabolic processes, protein translation, and other biological functions are lactylated, with lactylation levels varying according to the growth phase or lactate supplementation. Lactylation is regulated by glycolysis, and inhibition of L-lactate utilization can enhance lactylation levels. In addition to the known lactylase in E. coli, the acetyltransferase YfiQ can also catalyse lactylation. More importantly, L-lactyl coenzyme A (L-La-CoA) and S,D-lactoylglutathione (LGSH) can directly donate lactyl groups to target proteins for chemical lactylation. Lactylation is involved in Salmonella invasion of eukaryotic cells, suggesting that lactylation is crucial for bacterial virulence. Collectively, we have comprehensively investigated protein lactylome and the regulatory mechanisms of lactylation in Salmonella, providing valuable insights into studying lactylation function across diverse bacterial species.
Collapse
Affiliation(s)
- Chuanzhen Zhang
- Laboratory of Bacterial Pathogenesis, Shanghai Institute of Immunology, Shanghai Jiao Tong University School of Medicine, Shanghai, People’s Republic of China
- Shanghai Public Health Clinical Center, Fudan University, Shanghai, People’s Republic of China
| | - Tao Zhou
- Laboratory of Bacterial Pathogenesis, Shanghai Institute of Immunology, Shanghai Jiao Tong University School of Medicine, Shanghai, People’s Republic of China
| | - Chengxi Li
- Anhui Key Laboratory of Infection and Immunity, Department of Microbiology, Bengbu Medical College, Bengbu, People’s Republic of China
| | - Danni Wang
- Laboratory of Bacterial Pathogenesis, Shanghai Institute of Immunology, Shanghai Jiao Tong University School of Medicine, Shanghai, People’s Republic of China
| | - Jing Tao
- Laboratory of Bacterial Pathogenesis, Shanghai Institute of Immunology, Shanghai Jiao Tong University School of Medicine, Shanghai, People’s Republic of China
| | - Xiaocen Zhu
- Core Facility of Basic Medical Sciences, Shanghai Jiao Tong University School of Medicine, Shanghai, People’s Republic of China
| | - Jie Lu
- Department of Infectious Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People’s Republic of China
| | - Jinjing Ni
- Laboratory of Bacterial Pathogenesis, Shanghai Institute of Immunology, Shanghai Jiao Tong University School of Medicine, Shanghai, People’s Republic of China
| | - Yu-Feng Yao
- Laboratory of Bacterial Pathogenesis, Shanghai Institute of Immunology, Shanghai Jiao Tong University School of Medicine, Shanghai, People’s Republic of China
- Department of Infectious Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People’s Republic of China
- State Key Laboratory of Microbial Metabolism, and School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, People’s Republic of China
- Shanghai Key Laboratory of Emergency Prevention, Diagnosis and Treatment of Respiratory Infectious Diseases, Shanghai, People’s Republic of China
| |
Collapse
|
17
|
Chen X, Qin Y, Gan J, Wei T, Wei X, Xiong Y, Zhang Z, Wei B. Uncovering global research frontiers in deubiquitinating enzymes and immunotherapy: A bibliometric study. Hum Vaccin Immunother 2025; 21:2483558. [PMID: 40130728 PMCID: PMC11938311 DOI: 10.1080/21645515.2025.2483558] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2024] [Revised: 02/28/2025] [Accepted: 03/15/2025] [Indexed: 03/26/2025] Open
Abstract
Recently, immunotherapy has been a key therapeutic strategy for cancer. Deubiquitinating enzymes (DUBs), which are protein-modifying enzymes, have a crucial role in the pathogenesis of cancer, autoimmune diseases, and inflammation. DUBs influence the tumor immune microenvironment by regulating immune cell functions and key signaling pathways. Thus, the potential applications of DUBs in immunotherapy have piqued the interest of the scientific community. This study performed bibliometric analysis to comprehensively examine the research hotspots and trends in this field, providing theoretical foundations and guidance for future research. Studies associated with DUBs and immunotherapy conducted over a decade (2014 to 2024) were searched and extracted from Web of Science Collection database. The analysis was performed using CiteSpace, VOSviewer, and the Bibliometrix package in R software. Visualizations were generated for countries, institutions, authors, journals, references, and keyword co-occurrences. In total, 321 articles related to DUBs and immunotherapy were retrieved. The number of publications increased markedly since 2020. China had the highest number of publications, while the United States exerted the most influence in this field. Zhang Jinfang was the most influential author in this field. Zhejiang University was the institution with the highest number of publications. Nature was the most cited journal (807 total citations). Keyword analysis revealed that the primary research hotspots were expression, immunotherapy, ubiquitination, degradation, and cancer. This bibliometric analysis revealed the research trends and emerging frontiers in DUBs and immunotherapy, offering novel strategies for the application of DUBs in immunotherapy.
Collapse
Affiliation(s)
- Xia Chen
- Department of Geriatrics, Affiliated Hospital of Guilin Medical University, Guilin, China
| | - Yang Qin
- Department of Rheumatology and immunology, Affiliated Hospital of Guilin Medical University, Guilin, China
| | - Jinfeng Gan
- Guangxi Key Laboratory of Tumor Immunology and Microenvironmental Regulation, Guilin Medical University, Guilin, China
- Guangxi Health Commission Key Laboratory of Tumor Immunology and Receptor‑Targeted Drug Basic Research, Guilin Medical University, Guilin, China
| | - Tangwen Wei
- School of Public Health, Guilin Medical University, Guilin, China
| | - Xinyi Wei
- School of Public Health, Guangxi Medical University, Nanning, China
| | - Yaling Xiong
- Department of Gastrointestinal Surgery, Affiliated Hospital of Guilin Medical University, Guilin, China
| | - Zhichang Zhang
- Department of Computer, School of Intelligent Medicine China Medical University, Shenyang, Liaoning Province, China
| | - Bing Wei
- Department of Geriatrics, Affiliated Hospital of Guilin Medical University, Guilin, China
- Guangxi Key Laboratory of Diabetic Systems Medicine, Guilin Medical University, Guilin, China
| |
Collapse
|
18
|
Zhang M, Lu Z. tRNA modifications: greasing the wheels of translation and beyond. RNA Biol 2025; 22:1-25. [PMID: 39723662 DOI: 10.1080/15476286.2024.2442856] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Revised: 11/29/2024] [Accepted: 12/11/2024] [Indexed: 12/28/2024] Open
Abstract
Transfer RNA (tRNA) is one of the most abundant RNA types in cells, acting as an adaptor to bridge the genetic information in mRNAs with the amino acid sequence in proteins. Both tRNAs and small fragments processed from them play many nonconventional roles in addition to translation. tRNA molecules undergo various types of chemical modifications to ensure the accuracy and efficiency of translation and regulate their diverse functions beyond translation. In this review, we discuss the biogenesis and molecular mechanisms of tRNA modifications, including major tRNA modifications, writer enzymes, and their dynamic regulation. We also summarize the state-of-the-art technologies for measuring tRNA modification, with a particular focus on 2'-O-methylation (Nm), and discuss their limitations and remaining challenges. Finally, we highlight recent discoveries linking dysregulation of tRNA modifications with genetic diseases.
Collapse
Affiliation(s)
- Minjie Zhang
- Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Tianjin, China
- Tianjin Key Laboratory of Medical Epigenetics, Department of Bioinformatics, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Zhipeng Lu
- Department of Pharmacology and Pharmaceutical Sciences, University of Southern California, Los Angeles, CA, USA
- Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, CA, USA
- Eli and Edythe Broad CIRM Center for Regenerative Medicine and Stem Cell Research, University of Southern California, Los Angeles, CA, USA
| |
Collapse
|
19
|
Pinz MP, Medeiros I, Carvalho LADC, Meotti FC. Is uric acid a true antioxidant? Identification of uric acid oxidation products and their biological effects. Redox Rep 2025; 30:2498105. [PMID: 40415203 DOI: 10.1080/13510002.2025.2498105] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/27/2025] Open
Abstract
Uric acid (UA), the final product of purine metabolism in humans, exhibits a dual role as an anti or pro-oxidant, depending on the microenvironment. The two-electron oxidation of UA by biological oxidants can neutralize such harmful molecules. Additionally, UA chelates metals and can activate adaptive response against oxidation. However, some products of the reaction between UA and oxidants are not inert and, therefore, do not confer the anticipated antioxidant protection. A direct pro-oxidant effect is favoured in the one-electron oxidation of UA by heme-peroxidases yielding free radical intermediates that can initiate or propagate a radical-chain reaction. Additionally, an indirect pro-oxidant effect has been proposed by eliciting the expression or activation of enzymes that catalyse oxidant production, e.g. NADPH oxidase (NOX). This review brings together fundamental concepts and the molecular mechanisms of the redox reactions involving UA. The signature metabolites from these reactions are discussed to give valuable insights on whether these intermediates are being formed and what role they may play in disease pathogenesis. It proposes that, through identifying specific products, it may be possible to elucidate whether a harmful or protective action is linked to downstream bioactivities.
Collapse
Affiliation(s)
- Mikaela Peglow Pinz
- Department of Biochemistry, Institute of Chemistry, University of São Paulo, São Paulo, Brazil
| | - Isadora Medeiros
- Department of Biochemistry, Institute of Chemistry, University of São Paulo, São Paulo, Brazil
| | - Larissa Anastácio da Costa Carvalho
- Department of Biochemistry, Institute of Chemistry, University of São Paulo, São Paulo, Brazil
- Department of Clinical and Toxicological Analysis, School of Pharmaceutical Sciences, University of São Paulo, São Paulo, Brazil
| | - Flavia Carla Meotti
- Department of Biochemistry, Institute of Chemistry, University of São Paulo, São Paulo, Brazil
| |
Collapse
|
20
|
Bachmann MF, van Damme P, Lienert F, Schwarz TF. Virus-like particles: a versatile and effective vaccine platform. Expert Rev Vaccines 2025; 24:444-456. [PMID: 40387310 DOI: 10.1080/14760584.2025.2508517] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2025] [Revised: 04/25/2025] [Accepted: 05/15/2025] [Indexed: 05/20/2025]
Abstract
INTRODUCTION Traditional live-attenuated or inactivated vaccines have limitations, including risks associated with uncontrolled replication, reduced immunogenicity, or production complexities. To address these issues, alternative platforms such as virus-like particles (VLPs) have been developed. AREAS COVERED VLPs are self-assembling structures composed of viral proteins that mimic native viruses but are noninfectious. This review provides an overview of their structure, design and manufacture that make them an attractive platform for vaccine development. We then discuss the clinical development of some recently approved VLP vaccines and those widely used in immunization programs, summarizing the clinical trial data that underpins their efficacy and safety profiles. Additionally, we explore VLP vaccines in late-stage clinical development for respiratory syncytial virus and human metapneumovirus. EXPERT OPINION VLPs are a versatile and promising platform for vaccine development. Their ability to mimic native viruses while eliminating the risks associated with live vaccines positions them as an attractive platform for vaccine design. Currently approved VLP vaccines demonstrate that they can provide effective protection against a wide range of diseases. Advances in VLP design and production are likely to lead to highly effective vaccines, significantly contributing to global immunization efforts.
Collapse
Affiliation(s)
- Martin F Bachmann
- Department of Immunology, University Hospital of Bern, Bern, Switzerland
- University of Oxford, Oxford, UK
| | - Pierre van Damme
- Faculty of Medicine and Health Sciences, University of Antwerp, Antwerp, Belgium
| | - Florian Lienert
- Medical Affairs, Bavarian Nordic Berna GmbH, Thörishaus, Switzerland
| | - Tino F Schwarz
- Institute of Laboratory Medicine and Vaccination Centre, Klinikum Würzburg, Würzburg, Germany
| |
Collapse
|
21
|
Zheng XQ, Wang DB, Jiang YR, Song CL. Gut microbiota and microbial metabolites for osteoporosis. Gut Microbes 2025; 17:2437247. [PMID: 39690861 DOI: 10.1080/19490976.2024.2437247] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/11/2024] [Revised: 11/13/2024] [Accepted: 11/26/2024] [Indexed: 12/19/2024] Open
Abstract
Osteoporosis is an age-related bone metabolic disease. As an essential endocrine organ, the skeletal system is intricately connected with extraosseous organs. The crosstalk between bones and other organs supports this view. In recent years, the link between the gut microecology and bone metabolism has become an important research topic, both in preclinical studies and in clinical trials. Many studies have shown that skeletal changes are accompanied by changes in the composition and structure of the gut microbiota (GM). At the same time, natural or artificial interventions targeting the GM can subsequently affect bone metabolism. Moreover, microbiome-related metabolites may have important effects on bone metabolism. We aim to review the relationships among the GM, microbial metabolites, and bone metabolism and to summarize the potential mechanisms involved and the theory of the gut‒bone axis. We also describe existing bottlenecks in laboratory studies, as well as existing challenges in clinical settings, and propose possible future research directions.
Collapse
Affiliation(s)
- Xuan-Qi Zheng
- Department of Orthopaedics, Peking University Third Hospital, Beijing, China
| | - Ding-Ben Wang
- Department of Orthopaedics, Peking University Third Hospital, Beijing, China
| | - Yi-Rong Jiang
- Department of Orthopaedics, Peking University Third Hospital, Beijing, China
| | - Chun-Li Song
- Department of Orthopaedics, Peking University Third Hospital, Beijing, China
- Beijing Key Laboratory of Spinal Disease Research, Beijing, China
- Engineering Research Center of Bone and Joint Precision Medicine, Beijing, China
| |
Collapse
|
22
|
Chen L, Wu Y, Lv T, Tuo R, Xiao Y. Mesenchymal stem cells enchanced by salidroside to inhibit ferroptosis and improve premature ovarian insufficiency via Keap1/Nrf2/GPX4 signaling. Redox Rep 2025; 30:2455914. [PMID: 39874130 PMCID: PMC11776066 DOI: 10.1080/13510002.2025.2455914] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2025] Open
Abstract
BACKGROUND Regenerative medicine researches have shown that mesenchymal stem cells (MSCs) may be an effective treatment method for premature ovarian insufficiency (POI). However, the efficacy of MSCs is still limited. PURPOSE This study aims to explain whether salidroside and MSCs combination is a therapeutic strategy to POI and to explore salidroside-enhanced MSCs inhibiting ferroptosis via Keap1/Nrf2/GPX4 signaling. METHODS The effect of salidroside and MSCs on ovarian granular cells (GCs) was analyzed. After treatment, hormone levels and -fertility of rats were measured. Lipid peroxidation levels, iron deposition and mitochondrial morphology were detected. The genes and proteins of Keap1/Nrf2/GPX4 signaling were examined. RESULTS Salidroside and MSCs were found to inhibit cell death of GCs by reducing peroxidation and intracellular ferrous. Salidroside promotes the proliferation of MSCs and supports cell survival in ovary. Salidroside combined with MSCs therapy restored ovarian function, which was better than MSCs monotherapy. Salidroside-enhanced MSCs to inhibit ferroptosis. The results showed activation of the Keap1/Nrf2/GPX4 signaling and an increase in anti-ferroptosis molecule. CONCLUSIONS Salidroside-enhanced MSCs as a ferroptosis inhibitor and provide new therapeutic strategies for POI. The possible mechanisms of MSCs were related to maintaining redox homeostasis via a Keap1/Nrf2/GPX4 signaling.
Collapse
Affiliation(s)
- Lixuan Chen
- Department of Hematology, Shenzhen Qianhai Shekou Pilot Free Trade Zone Hospital, Shenzhen, People’s Republic of China
| | - Yingnan Wu
- Translational Medicine Center, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, People’s Republic of China
| | - Tiying Lv
- Guangzhou University of Chinese Medicine, Guangzhou, People’s Republic of China
| | - Rui Tuo
- Guangzhou University of Chinese Medicine, Guangzhou, People’s Republic of China
| | - Yang Xiao
- Department of Hematology, Shenzhen Qianhai Shekou Pilot Free Trade Zone Hospital, Shenzhen, People’s Republic of China
- Guangzhou University of Chinese Medicine, Guangzhou, People’s Republic of China
| |
Collapse
|
23
|
Chen R, Chai X, Zhang Y, Zhou T, Xia Y, Jiang X, Lv B, Zhang J, Zhou L, Tian X, Wang R, Mao L, Zhao F, Zhang H, Hu J, Qiu J, Zou Z, Chen C. Novel role of FTO in regulation of gut-brain communication via Desulfovibrio fairfieldensis-produced hydrogen sulfide under arsenic exposure. Gut Microbes 2025; 17:2438471. [PMID: 39852343 PMCID: PMC11776478 DOI: 10.1080/19490976.2024.2438471] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Revised: 10/26/2024] [Accepted: 11/26/2024] [Indexed: 01/26/2025] Open
Abstract
Fat mass and obesity-associated protein (FTO) is the key demethylase that reverses the abnormally altered N6-methyladenosine (m6A) modification in eukaryotic cells under environmental pollutants exposure. Arsenic is an environmental metalloid and can cause severe symptoms in human mainly through drinking water. However, there is no specific treatment for its toxic effects due to the uncovered mechanisms. We previously revealed that exposure to arsenic increased the level of m6A via down-regulation of FTO, which might serve as a potential target for intervention against arsenic-related disorders. In this study, our results demonstrated that chronic exposure to arsenic significantly disrupted the intestinal barrier and microenvironment. Also, this administration resulted in the enhancement of m6A modification and the reduction of FTO expression in the intestine. By using both CRISPR/Cas9-based FTO knock-in strategy and adeno-associated virus (AAV)-mediated overexpression of FTO in the intestine, we established for the first time that up-regulation of FTO remarkably ameliorated arsenic-induced disruption of intestinal barriers and altered microenvironment of mice. We also firstly identified a dominant gut microbial species, Desulfovibrio fairfieldensis, which was sharply reduced in arsenic-exposed mice, was able to proceed arsenic-induced neurobehavioral impairments by declining the levels of its major metabolite hydrogen sulfide. Administration of Desulfovibrio fairfieldensis could significantly alleviate the neurotoxicity of arsenic. Intriguingly, the beneficial effects of FTO against arsenic neurotoxicity possibly occurred through a novel gut-brain communication via Desulfovibrio fairfieldensis and its produced hydrogen sulfide. Collectively, these findings will provide new ideas for understanding the mechanisms of arsenic-induced toxic effects from a gut-brain communication perspective, and will assist the development of explicit intervention strategy via regulation of a new potential target FTO for prevention and treatment against arsenic-related both intestinal and neurological disorders.
Collapse
Affiliation(s)
- Ruonan Chen
- Department of Occupational and Environmental Health, School of Public Health, Chongqing Medical University, Chongqing, People’s Republic of China
| | - Xiaoqin Chai
- Department of Occupational and Environmental Health, School of Public Health, Chongqing Medical University, Chongqing, People’s Republic of China
| | - Yunxiao Zhang
- Department of Occupational and Environmental Health, School of Public Health, Chongqing Medical University, Chongqing, People’s Republic of China
| | - Tianxiu Zhou
- Department of Occupational and Environmental Health, School of Public Health, Chongqing Medical University, Chongqing, People’s Republic of China
| | - Yinyin Xia
- Department of Occupational and Environmental Health, School of Public Health, Chongqing Medical University, Chongqing, People’s Republic of China
| | - Xuejun Jiang
- Center of Experimental Teaching for Public Health, Experimental Teaching and Management Center, Chongqing Medical University, Chongqing, People’s Republic of China
| | - Bo Lv
- Department of Occupational and Environmental Health, School of Public Health, Chongqing Medical University, Chongqing, People’s Republic of China
| | - Jun Zhang
- Molecular Biology Laboratory of Respiratory Disease, Key Laboratory of Clinical Laboratory Diagnostics (Ministry of Education), College of Laboratory Medicine, Chongqing Medical University, Chongqing, People’s Republic of China
- Research Center for Environment and Human Health, School of Public Health, Chongqing Medical University, Chongqing, People’s Republic of China
| | - Lixiao Zhou
- Department of Occupational and Environmental Health, School of Public Health, Chongqing Medical University, Chongqing, People’s Republic of China
| | - Xin Tian
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Ruonan Wang
- Department of Health Laboratory Technology, School of Public Health, Chongqing Medical University, Chongqing, People’s Republic of China
| | - Lejiao Mao
- Molecular Biology Laboratory of Respiratory Disease, Key Laboratory of Clinical Laboratory Diagnostics (Ministry of Education), College of Laboratory Medicine, Chongqing Medical University, Chongqing, People’s Republic of China
| | - Feng Zhao
- Department of Health Laboratory Technology, School of Public Health, Chongqing Medical University, Chongqing, People’s Republic of China
| | - Hongyang Zhang
- Research Center for Environment and Human Health, School of Public Health, Chongqing Medical University, Chongqing, People’s Republic of China
- Department of Health Laboratory Technology, School of Public Health, Chongqing Medical University, Chongqing, People’s Republic of China
| | - Jun Hu
- Department of Neurology, Southwest Hospital, Third Military Medical University, Chongqing, People’s Republic of China
| | - Jingfu Qiu
- Research Center for Environment and Human Health, School of Public Health, Chongqing Medical University, Chongqing, People’s Republic of China
- Department of Health Laboratory Technology, School of Public Health, Chongqing Medical University, Chongqing, People’s Republic of China
| | - Zhen Zou
- Molecular Biology Laboratory of Respiratory Disease, Key Laboratory of Clinical Laboratory Diagnostics (Ministry of Education), College of Laboratory Medicine, Chongqing Medical University, Chongqing, People’s Republic of China
- Research Center for Environment and Human Health, School of Public Health, Chongqing Medical University, Chongqing, People’s Republic of China
| | - Chengzhi Chen
- Department of Occupational and Environmental Health, School of Public Health, Chongqing Medical University, Chongqing, People’s Republic of China
- Research Center for Environment and Human Health, School of Public Health, Chongqing Medical University, Chongqing, People’s Republic of China
| |
Collapse
|
24
|
Mu J, Chen SS, Li SQ, Jin Q, Geng J, Zou LW. Discovery of lignans as the effective inhibitors of CES1A alleviate lipid droplets formation. J Enzyme Inhib Med Chem 2025; 40:2472817. [PMID: 40207794 PMCID: PMC11986867 DOI: 10.1080/14756366.2025.2472817] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2024] [Revised: 02/11/2025] [Accepted: 02/22/2025] [Indexed: 04/11/2025] Open
Abstract
ER carboxylesterase 1A (CES1A) is an important metabolic enzyme involved in lipid metabolism. Targeting the CES1A is a promising approach for diseases associated with disorders of lipid metabolism therapy. In this study, screening of 26 natural lignans, three of them were found displaying potent inhibition on CES1A and high specificity over other serine hydrolases. Inhibition kinetic analyses demonstrated that Schisandrin C and Anwuligan were mixed-type inhibitors, while Magnolol acts as a competitive inhibitor. Further investigation showed that they were cell permeable and exhibited minimal cytotoxicity and mitochondrial toxicity, as well as capable of inhibiting intracellular CES1A in living cells. Further investigation found that three Schisandras decreased the number of lipid droplets (LDs) in free fatty acid (FFA)-treated HepG2 cells. Collectively, our findings suggest that Schisandrin C is a potent and highly selective inhibitor of CES1A, which can be served as a promising lead compound.
Collapse
Affiliation(s)
- Jie Mu
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Department of Ophthalmology, The First Hospital of China Medical University, Shenyang, China
| | - Si-Si Chen
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Shi-Qing Li
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Qiang Jin
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Jin Geng
- Department of Ophthalmology, The First Hospital of China Medical University, Shenyang, China
| | - Li-Wei Zou
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| |
Collapse
|
25
|
Nakajima A, Arzamasov AA, Sakanaka M, Murakami R, Kozakai T, Yoshida K, Katoh T, Ojima MN, Hirose J, Nagao S, Xiao JZ, Odamaki T, Rodionov DA, Katayama T. In vitro competition with Bifidobacterium strains impairs potentially pathogenic growth of Clostridium perfringens on 2'-fucosyllactose. Gut Microbes 2025; 17:2478306. [PMID: 40102238 PMCID: PMC11956901 DOI: 10.1080/19490976.2025.2478306] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/06/2024] [Revised: 02/23/2025] [Accepted: 03/04/2025] [Indexed: 03/20/2025] Open
Abstract
Fortifying infant formula with human milk oligosaccharides, such as 2'-fucosyllactose (2'-FL), is a global trend. Previous studies have shown the inability of pathogenic gut microbes to utilize 2'-FL. However, the present study demonstrates that the type strain (JCM 1290T) of Clostridium perfringens, a pathobiont species often more prevalent and abundant in the feces of C-section-delivered infants, exhibits potentially pathogenic growth on 2'-FL. The expression of genes for α-toxin, an activator of NLRP3 inflammasome, and ethanolamine ammonia-lyase, a factor responsible for the progression of gas gangrene, was significantly upregulated during 2'-FL assimilation compared to growth on lactose. However, colony-forming unit of C. perfringens JCM 1290T markedly decreased when co-cultivated with selected strains of Bifidobacterium, a taxon frequently detected in the breastfed infant gut. Moreover, during co-cultivation, the expression of virulence-related genes, including the gene for perfringolysin O - another activator of NLRP3 inflammasome - were significantly downregulated, while the lactate oxidation genes were upregulated. This can occur through two different mechanisms: direct competition for 2'-FL between the two organisms, or cross-feeding of lactose, released from 2'-FL by C. perfringens JCM 1290T, to Bifidobacterium. Attenuation of α-toxin production by the selected Bifidobacterium strains was observed to varying extents in 2'-FL-utilizing C. perfringens strains clinically isolated from healthy infants. Our results warrant detailed in vivo studies using animal models with dysbiotic microbiota dominated by various types of C. perfringens strains to further validate the safety of 2'-FL for clinical interventions, particularly on vulnerable preterm infants.
Collapse
Affiliation(s)
- Aruto Nakajima
- Graduate School of Biostudies, Kyoto University, Kyoto, Japan
| | - Aleksandr A. Arzamasov
- Infectious and Inflammatory Disease Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA
| | | | - Ryuta Murakami
- Innovative Research Institute, Morinaga Milk Industry Co, Ltd, Zama, Kanagawa, Japan
| | - Tomoya Kozakai
- Graduate School of Biostudies, Kyoto University, Kyoto, Japan
| | - Keisuke Yoshida
- Innovative Research Institute, Morinaga Milk Industry Co, Ltd, Zama, Kanagawa, Japan
| | - Toshihiko Katoh
- Graduate School of Biostudies, Kyoto University, Kyoto, Japan
| | - Miriam N. Ojima
- Graduate School of Biostudies, Kyoto University, Kyoto, Japan
| | - Junko Hirose
- Department of Food and Nutrition, Kyoto Women’s University, Kyoto, Japan
| | | | - Jin-Zhong Xiao
- Graduate School of Biostudies, Kyoto University, Kyoto, Japan
- Innovative Research Institute, Morinaga Milk Industry Co, Ltd, Zama, Kanagawa, Japan
| | - Toshitaka Odamaki
- Graduate School of Biostudies, Kyoto University, Kyoto, Japan
- Innovative Research Institute, Morinaga Milk Industry Co, Ltd, Zama, Kanagawa, Japan
| | - Dmitry A. Rodionov
- Infectious and Inflammatory Disease Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA
| | - Takane Katayama
- Graduate School of Biostudies, Kyoto University, Kyoto, Japan
| |
Collapse
|
26
|
Ganglberger M, Koschak A. Exploring the potential for gene therapy in Cav1.4-related retinal channelopathies. Channels (Austin) 2025; 19:2480089. [PMID: 40129245 PMCID: PMC11938310 DOI: 10.1080/19336950.2025.2480089] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2025] [Revised: 03/07/2025] [Accepted: 03/11/2025] [Indexed: 03/26/2025] Open
Abstract
The visual process begins with photon detection in photoreceptor outer segments within the retina, which processes light signals before transmission to the thalamus and visual cortex. Cav1.4 L-type calcium channels play a crucial role in this process, and dysfunction of these channels due to pathogenic variants in corresponding genes leads to specific manifestations in visual impairments. This review explores the journey from basic research on Cav1.4 L-type calcium channel complexes in retinal physiology and pathophysiology to their potential as gene therapy targets. Moreover, we provide a concise overview of key findings from studies using different animal models to investigate retinal diseases. It will critically examine the constraints these models present when attempting to elucidate retinal channelopathies. Additionally, the paper will explore potential strategies for addressing Cav1.4 channel dysfunction and discuss the current challenges facing gene therapy approaches in this area of research.
Collapse
Affiliation(s)
- Matthias Ganglberger
- Pharmacology and Toxicology, Institute of Pharmacy, University of Innsbruck, Innsbruck, Austria
| | - Alexandra Koschak
- Pharmacology and Toxicology, Institute of Pharmacy, University of Innsbruck, Innsbruck, Austria
| |
Collapse
|
27
|
Chen J, Zhou Q, Su L, Ni L. Mitochondrial dysfunction: the hidden catalyst in chronic kidney disease progression. Ren Fail 2025; 47:2506812. [PMID: 40441691 PMCID: PMC12123951 DOI: 10.1080/0886022x.2025.2506812] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2025] [Accepted: 05/10/2025] [Indexed: 06/02/2025] Open
Abstract
Chronic kidney disease (CKD) represents a global health epidemic, with approximately one-third of affected individuals ultimately necessitating renal replacement therapy or transplantation. The kidney, characterized by its exceptionally high energy demands, exhibits significant sensitivity to alterations in energy supply and mitochondrial function. In CKD, a compromised capacity for mitochondrial ATP synthesis has been documented. As research advances, the multifaceted roles of mitochondria, extending beyond their traditional functions in oxygen sensing and energy production, are increasingly acknowledged. Empirical studies have demonstrated a strong association between mitochondrial dysfunction and the pathogenesis of fibrosis and cellular apoptosis in CKD. Targeting mitochondrial dysfunction holds substantial therapeutic promise, with emerging insights into its epigenetic regulation in CKD, particularly involving non-coding RNAs and DNA methylation. This article presents a comprehensive review of contemporary research on mitochondrial dysfunction in relation to the onset and progression of CKD. It elucidates the associated molecular mechanisms across various renal cell types and proposes novel research avenues for CKD treatment.
Collapse
Affiliation(s)
- Jinhu Chen
- Department of Nephrology, Huanggang Central Hospital of Yangtze University, Huanggang, China
- Department of Nephrology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Qiuyuan Zhou
- Department of Nephrology, Zhongnan Hospital of Wuhan University, Wuhan, China
- Department of Pathology, Liang Ping People’s Hospital of Chongqing, Chongqing, People’s Republic of China
| | - Lianjiu Su
- Department of Critical Care Medicine, Zhongnan Hospital of Wuhan University, Wuhan, China
- Clinical Research Center of Hubei Critical Care Medicine, Wuhan, China
- Department of Cardiology, David Geffen School of Medicine, University of California, Los Angeles, CA, USA
| | - Lihua Ni
- Department of Nephrology, Zhongnan Hospital of Wuhan University, Wuhan, China
| |
Collapse
|
28
|
Lv X, Yang C, Li X, Liu Y, Yang Y, Jin T, Chen Z, Jia J, Wang M, Li L. Ferroptosis and hearing loss: from molecular mechanisms to therapeutic interventions. J Enzyme Inhib Med Chem 2025; 40:2468853. [PMID: 39992186 PMCID: PMC11852237 DOI: 10.1080/14756366.2025.2468853] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2024] [Revised: 02/11/2025] [Accepted: 02/13/2025] [Indexed: 02/25/2025] Open
Abstract
Hearing loss profoundly affects social engagement, mental health, cognition, and brain development, with sensorineural hearing loss (SNHL) being a major concern. Linked to ototoxic medications, ageing, and noise exposure, SNHL presents significant treatment challenges, highlighting the need for effective prevention and regeneration strategies. Ferroptosis, a distinct form of cell death featuring iron-dependent lipid peroxidation, has garnered interest due to its potential role in cancer, ageing, and neuronal degeneration, especially hearing loss. The emerging role of ferroptosis as a crucial mediator in SNHL suggests that it may offer a novel therapeutic target for otoprotection. This review aims to summarise the intricate connection between ferroptosis and SNHL, offering a fresh perspective for exploring targeted therapeutic strategies that could potentially mitigate cochlear cells damage and enhance the quality of life for individuals with hearing impairments.
Collapse
Affiliation(s)
- Xingyi Lv
- Department of Physiology, College of Medicine, Jiaxing University, Jiaxing, Zhejiang, China
| | - Chenyi Yang
- Department of Physiology, College of Medicine, Jiaxing University, Jiaxing, Zhejiang, China
| | - Xianying Li
- Department of Physiology, College of Medicine, Jiaxing University, Jiaxing, Zhejiang, China
| | - Yun Liu
- Department of Physiology, College of Medicine, Jiaxing University, Jiaxing, Zhejiang, China
| | - Yu Yang
- Department of Physiology, College of Medicine, Jiaxing University, Jiaxing, Zhejiang, China
| | - Tongyan Jin
- Department of Physiology, College of Medicine, Jiaxing University, Jiaxing, Zhejiang, China
| | - Zhijian Chen
- Department of Physiology, College of Medicine, Jiaxing University, Jiaxing, Zhejiang, China
| | - Jinjing Jia
- Department of Physiology, College of Medicine, Jiaxing University, Jiaxing, Zhejiang, China
| | - Min Wang
- Department of Physiology, College of Medicine, Jiaxing University, Jiaxing, Zhejiang, China
| | - Li Li
- Department of Physiology, College of Medicine, Jiaxing University, Jiaxing, Zhejiang, China
| |
Collapse
|
29
|
Chan P, Ye ZW, Zhao W, Ong CP, Sun XY, Cheung PHH, Jin DY. Mpox virus poxin-schlafen fusion protein suppresses innate antiviral response by sequestering STAT2. Emerg Microbes Infect 2025; 14:2477639. [PMID: 40066622 PMCID: PMC11921170 DOI: 10.1080/22221751.2025.2477639] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2024] [Revised: 03/03/2025] [Accepted: 03/05/2025] [Indexed: 03/20/2025]
Abstract
Mpox virus (MPXV) has to establish efficient interferon (IFN) antagonism for effective replication. MPXV-encoded IFN antagonists have not been fully elucidated. In this study, the IFN antagonism of poxin-schlafen (PoxS) fusion gene of MPXV was characterized. MPXV PoxS was capable of decreasing cGAS-produced 2'3'-cGAMP, like its ortholog poxin of vaccinia virus, which is the first known cytosolic nuclease that hydrolyses the 3'-5' bond of 2'3'-cyclic GMP-AMP (cGAMP). However, MPXV PoxS did not suppress cGAS-STING-mediated type I IFN production. Instead, MPXV PoxS antagonized basal and type I IFN-induced expression of IFN-stimulated genes such as OAS1, SAMD9, SAMD9L, ISG15, ISG56 and IFIT3. Consistently, MPXV PoxS inhibited both basal and type I IFN-stimulated activity of interferon-stimulated response elements, but did not affect activation of IFN-γ-activated sites. Mechanistically, MPXV PoxS interacted with STAT2 and sequestered it in the cytoplasm. Both the viral schlafen fusion and the active site of 2'3'-cGAMP nuclease were required for STAT2 sequestration and consequent suppression of IFN-stimulated gene expression. MPXV PoxS conferred resistance to the suppression of MPXV replication by type I IFN. Taken together, our findings suggested that MPXV PoxS counteracts host antiviral response by sequestering STAT2 to circumvent basal and type I IFN-induced expression of antiviral genes.
Collapse
Affiliation(s)
- Pearl Chan
- School of Biomedical Sciences, The University of Hong Kong, Pokfulam, Hong Kong
| | - Zi-Wei Ye
- School of Biomedical Sciences, The University of Hong Kong, Pokfulam, Hong Kong
| | - Wenlong Zhao
- School of Biomedical Sciences, The University of Hong Kong, Pokfulam, Hong Kong
| | - Chon-Phin Ong
- School of Biomedical Sciences, The University of Hong Kong, Pokfulam, Hong Kong
| | - Xiao-Yu Sun
- School of Biomedical Sciences, The University of Hong Kong, Pokfulam, Hong Kong
| | | | - Dong-Yan Jin
- School of Biomedical Sciences, The University of Hong Kong, Pokfulam, Hong Kong
| |
Collapse
|
30
|
Jungfleisch J, Gebauer F. RNA-binding proteins as therapeutic targets in cancer. RNA Biol 2025; 22:1-8. [PMID: 40016176 PMCID: PMC11869776 DOI: 10.1080/15476286.2025.2470511] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Revised: 02/04/2025] [Accepted: 02/17/2025] [Indexed: 03/01/2025] Open
Abstract
RNA-binding proteins (RBPs) have emerged as critical regulators of cancer progression, influencing virtually all hallmarks of cancer. Their ability to modulate gene expression patterns that promote or inhibit tumorigenesis has positioned RBPs as promising targets for novel anti-cancer therapies. This mini-review summarizes the current state of RBP-targeted cancer treatments, focusing on five examples, eIF4F, FTO, SF3B1, RBM39 and nucleolin. We highlight the diversity of current targeting approaches and discuss ongoing challenges including the complexity of RBP regulatory networks, potential off-target effects and the need for more specific targeting methods. By assessing the future potential of novel therapeutic avenues, we provide insights into the evolving landscape of cancer treatment and the critical role RBPs may play in next-generation therapeutics.
Collapse
Affiliation(s)
- Jennifer Jungfleisch
- Centre for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology, Barcelona, Spain
| | - Fátima Gebauer
- Centre for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology, Barcelona, Spain
- Universitat Pompeu Fabra (UPF), Barcelona, Spain
| |
Collapse
|
31
|
Boucard AS, Kulakauskas S, Alazzaz J, Chaouch S, Mammeri M, Millan-Oropeza A, Machado C, Henry C, Péchoux C, Richly H, Gassel M, Langella P, Polack B, Florent I, Bermúdez-Humarán LG. Isolation of derivatives from the food-grade probiotic Lactobacillus johnsonii CNCM I-4884 with enhanced anti- Giardia activity. Gut Microbes 2025; 17:2474149. [PMID: 40145272 PMCID: PMC11951713 DOI: 10.1080/19490976.2025.2474149] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/15/2024] [Revised: 01/03/2025] [Accepted: 02/25/2025] [Indexed: 03/28/2025] Open
Abstract
Giardiasis, a widespread intestinal parasitosis affecting humans and animals, is a growing concern due to the emergence of drug-resistant strains of G. intestinalis. Probiotics offer a promising alternative for preventing and treating giardiasis. Recent studies have shown that the probiotic Lactobacillus johnsonii CNCM I-4884 inhibits G. intestinalis growth both in vitro and in vivo. This protective effect is largely mediated by bile salt hydrolase (BSH) enzymes, which convert conjugated bile acids (BAs) into free forms that are toxic to the parasite. The objective of this study was to use adaptive evolution to develop stress-resistant derivatives of L. johnsonii CNCM I-4884, with the aim of improving its anti-Giardia activity. Twelve derivatives with enhanced resistance to BAs and reduced autolysis were generated. Among them, derivative M11 exhibited the highest in vitro anti-Giardia effect with enhanced BSH activity. Genomic and proteomic analyses of M11 revealed two SNPs and the upregulation of the global stress response by SigB, which likely contributed to its increased BAs resistance and BSH overproduction. Finally, the anti-Giardia efficacy of M11 was validated in a murine model of giardiasis. In conclusion, our results demonstrate that adaptive evolution is an effective strategy to generate robust food-grade bacteria with improved health benefits.
Collapse
Affiliation(s)
- Anne-Sophie Boucard
- Département Adaptation du Vivant, Université Paris-Saclay, INRAE, AgroParisTech, Micalis Institute, Jouy-en-Josas, France
| | - Saulius Kulakauskas
- Département Adaptation du Vivant, Université Paris-Saclay, INRAE, AgroParisTech, Micalis Institute, Jouy-en-Josas, France
| | - Jana Alazzaz
- UMR 7245, Muséum National d’Histoire Naturelle, Centre National de la Recherche Scientifique, Sorbonne Universités, Paris, France
| | - Soraya Chaouch
- UMR 7245, Muséum National d’Histoire Naturelle, Centre National de la Recherche Scientifique, Sorbonne Universités, Paris, France
| | - Mohamed Mammeri
- Anses, INRAE, Ecole Nationale Vétérinaire d’Alfort, UMR BIPAR, Laboratoire de Santé Animale, Maisons-Alfort, France
| | - Aaron Millan-Oropeza
- Plateforme d’Analyse Protéomique Paris Sud-Ouest (PAPPSO), INRAE, MICALIS Institute, Université Paris-Saclay, Jouy-en-Josas, France
| | - Carine Machado
- Plateforme d’Analyse Protéomique Paris Sud-Ouest (PAPPSO), INRAE, MICALIS Institute, Université Paris-Saclay, Jouy-en-Josas, France
| | - Céline Henry
- Plateforme d’Analyse Protéomique Paris Sud-Ouest (PAPPSO), INRAE, MICALIS Institute, Université Paris-Saclay, Jouy-en-Josas, France
| | - Christine Péchoux
- Université Paris-Saclay, INRAE, AgroParisTech, GABI, Jouy-en-Josas, France
| | - Holger Richly
- Boehringer Ingelheim Vetmedica GmbH, Kathrinenhof Research Center, Rohrdorf, Germany
| | - Michael Gassel
- Boehringer Ingelheim Vetmedica GmbH, Kathrinenhof Research Center, Rohrdorf, Germany
| | - Philippe Langella
- Département Adaptation du Vivant, Université Paris-Saclay, INRAE, AgroParisTech, Micalis Institute, Jouy-en-Josas, France
| | - Bruno Polack
- Anses, INRAE, Ecole Nationale Vétérinaire d’Alfort, UMR BIPAR, Laboratoire de Santé Animale, Maisons-Alfort, France
| | - Isabelle Florent
- UMR 7245, Muséum National d’Histoire Naturelle, Centre National de la Recherche Scientifique, Sorbonne Universités, Paris, France
| | - Luis G. Bermúdez-Humarán
- Département Adaptation du Vivant, Université Paris-Saclay, INRAE, AgroParisTech, Micalis Institute, Jouy-en-Josas, France
| |
Collapse
|
32
|
Wu Y, Wen F, Gou S, Ran Q, Chu Y, Ma W, Zhao K. Multifaceted quorum-sensing inhibiting activity of 3-(Benzo[d][1,3]dioxol-4-yl)oxazolidin-2-one mitigates Pseudomonas aeruginosa virulence. Virulence 2025; 16:2479103. [PMID: 40104940 DOI: 10.1080/21505594.2025.2479103] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2023] [Revised: 11/10/2024] [Accepted: 03/03/2025] [Indexed: 03/20/2025] Open
Abstract
As antibiotic resistance escalates into a global health crisis, novel therapeutic approaches against infectious diseases are in urgent need. Pseudomonas aeruginosa, an adaptable opportunistic pathogen, poses substantial challenges in treating a range of infections. The quorum-sensing (QS) system plays a pivotal role in orchestrating the production of a large set of virulence factors in a cell density-dependent manner, and the anti-virulence strategy targeting QS may show huge potential. Here, we present a comprehensive investigation into the potential of the synthesized compound 3-(benzo[d][1,3]dioxol-4-yl)oxazolidin-2-one (OZDO, C10H9NO4) as a QS inhibitor to curb the virulence of P. aeruginosa. By employing an integrated approach encompassing in silico screening, in vitro and in vivo functional identification, we elucidated the multifaceted effects of OZDO. Molecular docking predicted that OZDO interfered with three core regulatory proteins of P. aeruginosa QS system. Notably, OZDO exhibited significant inhibition on the production of pyocyanin, rhamnolipid and extracellular proteases, biofilm formation, and cell motilities of P. aeruginosa. Transcriptomic analysis and quantitative real-time PCR displayed the down-regulation of QS-controlled genes in OZDO-treated PAO1, reaffirming the QS-inhibition activity of OZDO. In vivo assessments using a Caenorhabditis elegans-infection model demonstrated OZDO mitigated P. aeruginosa pathogenicity, particularly against the hypervirulent strain PA14. Moreover, OZDO in combination with polymyxin B and aztreonam presented a promising avenue for innovative anti-infective therapy. Our study sheds light on the multifaceted potential of OZDO as an anti-virulence agent and its significance in combating P. aeruginosa-associated infections.
Collapse
Affiliation(s)
- Yi Wu
- Antibiotics Research and Re-Evaluation Key Laboratory of Sichuan Province, School of Pharmacy, Chengdu University, Chengdu, Sichuan, China
| | - Fulong Wen
- Antibiotics Research and Re-Evaluation Key Laboratory of Sichuan Province, School of Pharmacy, Chengdu University, Chengdu, Sichuan, China
| | - Shiyi Gou
- Antibiotics Research and Re-Evaluation Key Laboratory of Sichuan Province, School of Pharmacy, Chengdu University, Chengdu, Sichuan, China
| | - Qiman Ran
- Antibiotics Research and Re-Evaluation Key Laboratory of Sichuan Province, School of Pharmacy, Chengdu University, Chengdu, Sichuan, China
| | - Yiwen Chu
- Antibiotics Research and Re-Evaluation Key Laboratory of Sichuan Province, School of Pharmacy, Chengdu University, Chengdu, Sichuan, China
| | - Wenbo Ma
- Antibiotics Research and Re-Evaluation Key Laboratory of Sichuan Province, School of Pharmacy, Chengdu University, Chengdu, Sichuan, China
| | - Kelei Zhao
- Antibiotics Research and Re-Evaluation Key Laboratory of Sichuan Province, School of Pharmacy, Chengdu University, Chengdu, Sichuan, China
| |
Collapse
|
33
|
Liu J, Ma C, Cheng Y, Wang M, Zhao G, Huang L, Song R, Wang X, Li H. METTL14 and WTAP play a crucial role in the regulation of bovine preadipocyte differentiation. Anim Biotechnol 2025; 36:2476531. [PMID: 40094566 DOI: 10.1080/10495398.2025.2476531] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2024] [Accepted: 03/03/2025] [Indexed: 03/19/2025]
Abstract
m6A methylation is the most common mRNA modification in mammals and plays a significant role in regulating various biological functions. Some studies have demonstrated that the methyltransferase METTL3 can promote adipogenesis. However, the regulatory mechanisms of METTL14 and WTAP, both methyltransferases, in adipogenesis remain unclear. This study investigated their effects on bovine preadipocyte differentiation using siRNA-mediated knockdown combined with transcriptomic analysis. Silencing METTL14 and WTAP significantly impaired lipid droplet formation and revealed distinct regulatory pathways: METTL14 knockdown affected genes like JAK2 and STAT3, while WTAP suppression down-regulated PPARγ/FABP4 signalling pathway components. These findings demonstrate that WTAP specifically modulates bovine adipocyte differentiation through the PPARγ/FABP4 pathway.
Collapse
Affiliation(s)
- Jia Liu
- College of Animal Science, Shanxi Agricultural University, Taigu, Shanxi, China
| | - Chicheng Ma
- College of Animal Science, Shanxi Agricultural University, Taigu, Shanxi, China
| | - Yu Cheng
- College of Animal Science, Shanxi Agricultural University, Taigu, Shanxi, China
| | - Minzhi Wang
- College of Animal Science, Shanxi Agricultural University, Taigu, Shanxi, China
| | - Guoqing Zhao
- College of Animal Science, Shanxi Agricultural University, Taigu, Shanxi, China
| | - Liwei Huang
- College of Animal Science, Shanxi Agricultural University, Taigu, Shanxi, China
| | - Ruigao Song
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Third Hospital of Shanxi Medical University, Tongji Shanxi Hospital, Taiyuan, China
- Shanxi Academy of Advanced Research and Innovation, Taiyuan, China
| | - Xi Wang
- College of Animal Science, Shanxi Agricultural University, Taigu, Shanxi, China
| | - Hongxia Li
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Third Hospital of Shanxi Medical University, Tongji Shanxi Hospital, Taiyuan, China
| |
Collapse
|
34
|
Zhou S, Hui X, Wang W, Zhao C, Jin M, Qin Y, Chen M. SARS-CoV-2 and HCoV-OC43 regulate host m6A modification via activation of the mTORC1 signalling pathway to facilitate viral replication. Emerg Microbes Infect 2025; 14:2447620. [PMID: 39745173 PMCID: PMC11852242 DOI: 10.1080/22221751.2024.2447620] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2024] [Revised: 12/08/2024] [Accepted: 12/22/2024] [Indexed: 02/25/2025]
Abstract
N6-methyladenosine (m6A) is the most prevalent post-transcriptional modification in eukaryotic RNA and is also present in various viral RNAs, where it plays a crucial role in regulating the viral life cycle. However, the molecular mechanisms through which viruses regulate host RNA m6A methylation are not fully understood. In this study, we reveal that SARS-CoV-2 and HCoV-OC43 infection enhance host m6A modification by activating the mTORC1 signalling pathway. Specifically, the viral non-structural protein nsp14 upregulates the expression of S-adenosylmethionine synthase MAT2A in an mTORC1-dependent manner. This mTORC1-MAT2A axis subsequently stimulates the synthesis of S-adenosylmethionine (SAM). The increase of SAM then enhances the m6A methylation of host RNA and facilitates viral replication. Our findings uncover a molecular mechanism by which viruses regulate host m6A methylation and provide insights into how SARS-CoV-2 hijacks host cellular epitranscriptomic modifications to promote its replication.
Collapse
Affiliation(s)
- Shixiong Zhou
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan, People’s Republic of China
| | - Xianfeng Hui
- National key laboratory of agricultural microbiology, Huazhong Agricultural University, Wuhan, People’s Republic of China
| | - Weiwei Wang
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan, People’s Republic of China
| | - Chunbei Zhao
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan, People’s Republic of China
| | - Meilin Jin
- National key laboratory of agricultural microbiology, Huazhong Agricultural University, Wuhan, People’s Republic of China
| | - Yali Qin
- School of Life Sciences, Hubei University, Wuhan, People’s Republic of China
| | - Mingzhou Chen
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan, People’s Republic of China
- School of Life Sciences, Hubei University, Wuhan, People’s Republic of China
| |
Collapse
|
35
|
Zhu Q, Cheng J, Gao Y, Zhang Z, Pan J, Su X, Fei D, Cai L, Yu J, Chen Y, Jiao W, Wu D, Li X, Xiao P. NVP-2, in combination with Orlistat, represents a promising therapeutic strategy for acute myeloid leukemia. Cancer Biol Ther 2025; 26:2450859. [PMID: 39800696 PMCID: PMC11730633 DOI: 10.1080/15384047.2025.2450859] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2024] [Revised: 12/31/2024] [Accepted: 01/02/2025] [Indexed: 01/16/2025] Open
Abstract
Cell cycle dysregulation and the corresponding metabolic reprogramming play significant roles in tumor development and progression. CDK9, a kinase that regulates gene transcription and cell cycle, also induces oncogene transcription and abnormal cell cycle in AML cells. The function of CDK9 for gene regulation in AML cells requires further exploration. In this study, we knocked down the CDK9 to investigate its effects on the growth and survival of AML cells. Through RNA-seq analysis, we identified that in U937 cells CDK9 regulates numerous genes involved in proliferation and apoptosis, including mTOR, SREBF1, and Bcl-2. Furthermore, our results demonstrated that both CDK9 and FASN are crucial for the proliferation and survival of Kasumi-1 and U937 cells. Mechanistically, MCL1, c-Myc, and Akt/mTOR/SREBF1 may be critical factors and pathways in the combined therapy of NVP-2 and Orlistat. In summary, our study revealed that CDK9 and FASN are vital for maintaining AML cell survival and proliferation. Treatment with NVP-2 and Orlistat may be a promising clinical candidate for patients with AML.
Collapse
MESH Headings
- Humans
- Leukemia, Myeloid, Acute/drug therapy
- Leukemia, Myeloid, Acute/genetics
- Leukemia, Myeloid, Acute/pathology
- Leukemia, Myeloid, Acute/metabolism
- Orlistat/pharmacology
- Cell Proliferation/drug effects
- Cyclin-Dependent Kinase 9/genetics
- Cyclin-Dependent Kinase 9/metabolism
- Apoptosis/drug effects
- Cell Line, Tumor
- Fatty Acid Synthase, Type I/metabolism
- Fatty Acid Synthase, Type I/genetics
- Lactones/pharmacology
- Antineoplastic Combined Chemotherapy Protocols/pharmacology
- Antineoplastic Combined Chemotherapy Protocols/therapeutic use
- U937 Cells
- Cell Survival/drug effects
Collapse
Affiliation(s)
- Qing Zhu
- Children’s Hospital of Soochow University, Suzhou, China
- Department of Pediatrics, The Third People’s Hospital of Kunshan, Suzhou, China
| | - Jia Cheng
- Children’s Hospital of Soochow University, Suzhou, China
| | - Yuqing Gao
- Institute of Pediatric Research, Children’s Hospital of Soochow University, Suzhou, China
| | - Zimu Zhang
- Institute of Pediatric Research, Children’s Hospital of Soochow University, Suzhou, China
| | - Jian Pan
- Institute of Pediatric Research, Children’s Hospital of Soochow University, Suzhou, China
| | - Xin Su
- Department of Hematology, Children’s Hospital of Soochow University, Suzhou, China
| | - Danhong Fei
- Children’s Hospital of Soochow University, Suzhou, China
- Department of Pediatrics, Municipal Hospital Affiliated to Taizhou University, Taizhou, China
| | - Linbo Cai
- Children’s Hospital of Soochow University, Suzhou, China
| | - Juanjuan Yu
- Children’s Hospital of Soochow University, Suzhou, China
| | - Yanling Chen
- Children’s Hospital of Soochow University, Suzhou, China
| | - Wanyan Jiao
- Children’s Hospital of Soochow University, Suzhou, China
- Department of Pediatrics, The Third People’s Hospital of Yancheng, Yancheng, China
| | - Di Wu
- Institute of Pediatric Research, Children’s Hospital of Soochow University, Suzhou, China
| | - Xiaolu Li
- Institute of Pediatric Research, Children’s Hospital of Soochow University, Suzhou, China
| | - Peifang Xiao
- Department of Hematology, Children’s Hospital of Soochow University, Suzhou, China
| |
Collapse
|
36
|
Taba N, Fischer K, Estonian Biobank Research Team, Org E, Aasmets O. A novel framework for assessing causal effect of microbiome on health: long-term antibiotic usage as an instrument. Gut Microbes 2025; 17:2453616. [PMID: 39849320 PMCID: PMC11776458 DOI: 10.1080/19490976.2025.2453616] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Revised: 07/25/2024] [Accepted: 01/02/2025] [Indexed: 01/25/2025] Open
Abstract
Assessing causality is undoubtedly one of the key questions in microbiome studies for the upcoming years. Since randomized trials in human subjects are often unethical or difficult to pursue, analytical methods to derive causal effects from observational data deserve attention. As simple covariate adjustment is not likely to account for all potential confounders, the idea of instrumental variable (IV) analysis is worth exploiting. Here we propose a novel framework of antibiotic instrumental variable regression (AB-IVR) for estimating the causal relationships between microbiome and various diseases. We rely on the recent studies showing that antibiotic treatment has a cumulative long-term effect on the microbiome, resulting in individuals with higher antibiotic usage to have a more perturbed microbiome. We apply the AB-IVR method on the Estonian Biobank data and show that the microbiome has a causal role in numerous diseases including migraine, depression and irritable bowel syndrome. We show with a plethora of sensitivity analyses that the identified causal effects are robust and propose ways for further methodological developments.
Collapse
Affiliation(s)
- Nele Taba
- Estonian Genome Center, Institute of Genomics, University of Tartu, Tartu, Estonia
| | - Krista Fischer
- Estonian Genome Center, Institute of Genomics, University of Tartu, Tartu, Estonia
- Institute of Mathematics and Statistics, Faculty of Science and Technology, University of Tartu, Tartu, Estonia
| | | | - Elin Org
- Estonian Genome Center, Institute of Genomics, University of Tartu, Tartu, Estonia
| | - Oliver Aasmets
- Estonian Genome Center, Institute of Genomics, University of Tartu, Tartu, Estonia
| |
Collapse
|
37
|
Razzaq Meo S, Van de Wiele T, Defoirdt T. Indole signaling in Escherichia coli: a target for antivirulence therapy? Gut Microbes 2025; 17:2499573. [PMID: 40329925 PMCID: PMC12064070 DOI: 10.1080/19490976.2025.2499573] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/27/2025] [Revised: 04/14/2025] [Accepted: 04/24/2025] [Indexed: 05/08/2025] Open
Abstract
Pathogenic Escherichia coli are a major cause of infections in both humans and animals, leading to conditions such as severe diarrheal diseases, urinary tract infections, enteritis, and septicemia. To combat bacterial infections, antibiotics are widely utilized. However, the extensive and inappropriate use of antibiotics has fueled the development and spread of antibiotic resistance, posing a significant challenge to the effective treatment of E. coli. There is consequently an urgent need to explore alternative therapies to control such infections. This review provides an overview of the recent findings concerning indole signaling in E. coli. E. coli uses indole as a quorum sensing molecule, and indole signaling has been reported to decrease various virulence factors in pathogenic E. coli, including motility, biofilm formation, adherence to host cells, expression of the LEE pathogenicity island, and formation of attaching and effacing lesions. This makes indole signaling an interesting target for the development of new therapeutics in the framework of antivirulence therapy. Both natural and synthetic indole analogues have been explored as potential virulence inhibitors. This alternative approach could be advantageous, as it will exert less selective pressure for resistance development than conventional antibiotics.
Collapse
Affiliation(s)
- Sofia Razzaq Meo
- Center for Microbial Ecology and Technology (CMET), Department of Biotechnology, Ghent University, Gent, Belgium
| | - Tom Van de Wiele
- Center for Microbial Ecology and Technology (CMET), Department of Biotechnology, Ghent University, Gent, Belgium
| | - Tom Defoirdt
- Center for Microbial Ecology and Technology (CMET), Department of Biotechnology, Ghent University, Gent, Belgium
| |
Collapse
|
38
|
Xiao Y, He M, Zhang X, Yang M, Yuan Z, Yao S, Qin Y. Research progress on the mechanism of tumor cell ferroptosis regulation by epigenetics. Epigenetics 2025; 20:2500949. [PMID: 40327848 PMCID: PMC12064064 DOI: 10.1080/15592294.2025.2500949] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2025] [Revised: 04/24/2025] [Accepted: 04/28/2025] [Indexed: 05/08/2025] Open
Abstract
Cancer remains a significant barrier to human longevity and a leading cause of mortality worldwide. Despite advancements in cancer therapies, challenges such as cellular toxicity and drug resistance to chemotherapy persist. Regulated cell death (RCD), once regarded as a passive process, is now recognized as a programmed mechanism with distinct biochemical and morphological characteristics, thereby presenting new therapeutic opportunities. Ferroptosis, a novel form of RCD characterized by iron-dependent lipid peroxidation and unique mitochondrial damage, differs from apoptosis, autophagy, and necroptosis. It is driven by reactive oxygen species (ROS)-induced lipid peroxidation and is implicated in tumorigenesis, anti-tumor immunity, and resistance, particularly in tumors undergoing epithelial-mesenchymal transition. Moreover, ferroptosis is associated with ischemic organ damage, degenerative diseases, and aging, regulated by various cellular metabolic processes, including redox balance, iron metabolism, and amino acid, lipid, and glucose metabolism. This review focuses on the role of epigenetic factors in tumor ferroptosis, exploring their mechanisms and potential applications in cancer therapy. It synthesizes current knowledge to provide a comprehensive understanding of epigenetic regulation in tumor cell ferroptosis, offering insights for future research and clinical applications.
Collapse
Affiliation(s)
- Yuyang Xiao
- Department of Health Management Medical, The Third Xiangya Hospital of Central South University, Changsha, Hunan, China
- Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Mengyang He
- Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Xupeng Zhang
- Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Meng Yang
- Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Zhangchi Yuan
- Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Shanhu Yao
- Department of Radiology, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China
- Key Laboratory of Medical Information Research, Central South University, Changsha, Hunan, China
| | - Yuexiang Qin
- Department of Health Management Medical, The Third Xiangya Hospital of Central South University, Changsha, Hunan, China
- Department of Otolaryngology, Head and Neck Surgery, Xiangya Hospital, Central South University, Changsha, Hunan, China
| |
Collapse
|
39
|
Liu J, Wang L, Xie M, Zhao W, Sun J, Jin Y, Liu M, Zhao J, Cheng L, Wen C, Bi X, Huang C. Varespladib attenuates Naja atra-induced acute liver injury via reversing Nrf2 signaling-mediated ferroptosis and mitochondrial dysfunction. Redox Rep 2025; 30:2507557. [PMID: 40399141 PMCID: PMC12096701 DOI: 10.1080/13510002.2025.2507557] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/23/2025] Open
Abstract
Objective: To investigate the protective effects of varespladib against Naja atra-induced acute liver injury (ALI) and to elucidate the toxic mechanism of snake venom phospholipase A2 (SVPLA2)-induced hepatic oxidative stress, with a particular focus on the role of Nrf2 signaling and its downstream pathways.Methods: A combination of in vivo and in vitro models of N. atra envenomation was employed to assess liver injury, oxidative stress, and mitochondrial dysfunction. The interaction between SVPLA2 and Nrf2 was analyzed, and the effects of varespladib treatment on these processes were evaluated using histological analysis, biochemical assays, and molecular techniques targeting oxidative stress, ferroptosis, mitophagy, and apoptosis.Results: Varespladib significantly alleviated N. atra-induced ALI. SVPLA2 was found to directly bind to Nrf2, leading to severe oxidative stress. This oxidative stress initiated a cascade involving Nrf2-mediated ferroptosis, mitochondrial dysfunction, excessive mitophagy, and mitochondria-dependent apoptosis. Treatment with varespladib effectively reversed these pathological events by inhibiting SVPLA2 activity.Conclusion: Varespladib shows strong therapeutic potential for N. atra envenomation by targeting SVPLA2. Nrf2 was identified as a direct toxic target of SVPLA2, and Nrf2-mediated ferroptosis and mitochondrial dysfunction were key mechanisms underlying SVPLA2-induced hepatic injury.
Collapse
Affiliation(s)
- Jiahao Liu
- School of Basic Medicine Sciences, Jiangxi Medical College, Nanchang University, Nanchang, People’s Republic of China
| | - Linfeng Wang
- School of Basic Medicine Sciences, Jiangxi Medical College, Nanchang University, Nanchang, People’s Republic of China
| | - Mengxia Xie
- School of Basic Medicine Sciences, Jiangxi Medical College, Nanchang University, Nanchang, People’s Republic of China
| | - Wenjie Zhao
- School of Basic Medicine Sciences, Jiangxi Medical College, Nanchang University, Nanchang, People’s Republic of China
| | - Jiaqi Sun
- Queen Mary School, Jiangxi Medical College, Nanchang University, Nanchang, People’s Republic of China
| | - Yuji Jin
- Department of Basic Medicine, Jilin Medical University, Jilin, People’s Republic of China
| | - Meiling Liu
- School of Basic Medicine Sciences, Jiangxi Medical College, Nanchang University, Nanchang, People’s Republic of China
| | - Jianqi Zhao
- School of Basic Medicine Sciences, Jiangxi Medical College, Nanchang University, Nanchang, People’s Republic of China
| | - Lixia Cheng
- School of Basic Medicine Sciences, Jiangxi Medical College, Nanchang University, Nanchang, People’s Republic of China
| | - Cheng Wen
- School of clinical medicine, Jilin Medical University, Jilin, People’s Republic of China
| | - Xiaowen Bi
- School of Basic Medicine Sciences, Jiangxi Medical College, Nanchang University, Nanchang, People’s Republic of China
| | - Chunhong Huang
- School of Basic Medicine Sciences, Jiangxi Medical College, Nanchang University, Nanchang, People’s Republic of China
| |
Collapse
|
40
|
Farahani A, Farahani A, Kashfi K, Ghasemi A. Inhibition of hepatic gluconeogenesis in type 2 diabetes by metformin: complementary role of nitric oxide. Med Gas Res 2025; 15:507-519. [PMID: 40300886 PMCID: PMC12124709 DOI: 10.4103/mgr.medgasres-d-24-00100] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2024] [Revised: 10/15/2024] [Accepted: 01/21/2025] [Indexed: 05/01/2025] Open
Abstract
Metformin is the first-line treatment for type 2 diabetes mellitus. Type 2 diabetes mellitus is associated with decreased nitric oxide bioavailability, which has significant metabolic implications, including enhanced insulin secretion and peripheral glucose utilization. Similar to metformin, nitric oxide also inhibits hepatic glucose production, mainly by suppressing gluconeogenesis. This review explores the combined effects of metformin and nitric oxide on hepatic gluconeogenesis and proposes the potential of a hybrid metformin-nitric oxide drug for managing type 2 diabetes mellitus. Both metformin and nitric oxide inhibit gluconeogenesis through overlapping and distinct mechanisms. In hepatic gluconeogenesis, mitochondrial oxaloacetate is exported to the cytoplasm via various pathways, including the malate, direct, aspartate, and fumarate pathways. The effects of nitric oxide and metformin on the exportation of oxaloacetate are complementary; nitric oxide primarily inhibits the malate pathway, while metformin strongly inhibits the fumarate and aspartate pathways. Furthermore, metformin effectively blocks gluconeogenesis from lactate, glycerol, and glutamine, whereas nitric oxide mainly inhibits alanine-induced gluconeogenesis. Additionally, nitric oxide contributes to the adenosine monophosphate-activated protein kinase-dependent inhibition of gluconeogenesis induced by metformin. The combined use of metformin and nitric oxide offers the potential to mitigate common side effects. For example, lactic acidosis, a known side effect of metformin, is linked to nitric oxide deficiency, while the oxidative and nitrosative stress caused by nitric oxide could be counterbalanced by metformin's enhancement of glutathione. Metformin also amplifies nitric oxide -induced activation of adenosine monophosphate-activated protein kinase. In conclusion, a metformin-nitric oxide hybrid drug can benefit patients with type 2 diabetes mellitus by enhancing the inhibition of hepatic gluconeogenesis, decreasing the required dose of metformin for maintaining optimal glycemia, and lowering the incidence of metformin-associated lactic acidosis.
Collapse
Affiliation(s)
- Arman Farahani
- Endocrine Physiology Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Aryan Farahani
- Endocrine Physiology Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Khosrow Kashfi
- Department of Molecular, Cellular and Biomedical Sciences, Sophie Davis School of Biomedical Education, City University of New York School of Medicine, New York, NY, USA
| | - Asghar Ghasemi
- Endocrine Physiology Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
41
|
Sharma R, Mishra A, Bhardwaj M, Singh G, Indira Harahap LV, Vanjani S, Pan CH, Nepali K. Medicinal chemistry breakthroughs on ATM, ATR, and DNA-PK inhibitors as prospective cancer therapeutics. J Enzyme Inhib Med Chem 2025; 40:2489720. [PMID: 40256842 PMCID: PMC12013171 DOI: 10.1080/14756366.2025.2489720] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2025] [Revised: 03/28/2025] [Accepted: 04/01/2025] [Indexed: 04/22/2025] Open
Abstract
This review discusses the critical roles of Ataxia Telangiectasia Mutated Kinase (ATM), ATM and Rad3-related Kinase (ATR), and DNA-dependent protein kinase (DNA-PK) in the DNA damage response (DDR) and their implications in cancer. Emphasis is placed on the intricate interplay between these kinases, highlighting their collaborative and distinct roles in maintaining genomic integrity and promoting tumour development under dysregulated conditions. Furthermore, the review covers ongoing clinical trials, patent literature, and medicinal chemistry campaigns on ATM/ATR/DNA-PK inhibitors as antitumor agents. Notably, the medicinal chemistry campaigns employed robust drug design strategies and aimed at assembling new structural templates with amplified DDR kinase inhibitory ability, as well as outwitting the pharmacokinetic liabilities of the existing DDR kinase inhibitors. Given the success attained through such endeavours, the clinical pipeline of DNA repair kinase inhibitors is anticipated to be supplemented by a reasonable number of tractable entries (DDR kinase inhibitors) soon.
Collapse
Affiliation(s)
- Ram Sharma
- School of Pharmacy, College of Pharmacy, Taipei Medical University, Taipei, Taiwan
| | - Anshul Mishra
- School of Pharmacy, College of Pharmacy, Taipei Medical University, Taipei, Taiwan
| | - Monika Bhardwaj
- Institute of Biological Chemistry, Academia Sinica, Taipei, Taiwan
| | - Gurpreet Singh
- Department of Pharmaceutical Chemistry, ISF College of Pharmacy, Moga, India
| | | | - Sakshi Vanjani
- Molecular Medicine, University of South Florida, Tampa, FL, USA
| | - Chun Hsu Pan
- Ph.D. Program in Drug Discovery and Development Industry, College of Pharmacy, Taipei Medical University, Taipei, Taiwan
| | - Kunal Nepali
- School of Pharmacy, College of Pharmacy, Taipei Medical University, Taipei, Taiwan
- Ph.D. Program in Drug Discovery and Development Industry, College of Pharmacy, Taipei Medical University, Taipei, Taiwan
| |
Collapse
|
42
|
Kato K, Yasui H, Sato-Akaba H, Emoto MC, Fujii HG, Kmiec MM, Kuppusamy P, Nagane M, Yamashita T, Inanami O. Non-invasive electron paramagnetic resonance imaging detects tumor redox imbalance induced by ferroptosis. Redox Rep 2025; 30:2454887. [PMID: 39836064 PMCID: PMC11753017 DOI: 10.1080/13510002.2025.2454887] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2025] Open
Abstract
Targeting ferroptosis, cell death caused by the iron-dependent accumulation of lipid peroxides, and disruption of the redox balance are promising strategies in cancer therapy owing to the physiological characteristics of cancer cells. However, the detection of ferroptosis using in vivo imaging remains challenging. We previously reported that redox maps showing the reduction power per unit time of implanted tumor tissues via non-invasive redox imaging using a novel, compact, and portable electron paramagnetic resonance imaging (EPRI) device could be compared with tumor tissue sections. This study aimed to apply the EPRI technique to the in vivo detection of ferroptosis. Notably, redox maps reflecting changes in the redox status of tumors induced by the ferroptosis-inducing agent imidazole ketone erastin (IKE) were compared with the immunohistochemical images of 4-hydroxynonenal (4-HNE) in tumor tissue sections. Our comparison revealed a negative correlation between the reducing power of tumor tissue and the number of 4-HNE-positive cells. Furthermore, the control and IKE-treated groups exhibited significantly different distributions on the correlation map. Therefore, redox imaging using EPRI may contribute to the non-invasive detection of ferroptosis in vivo.
Collapse
Affiliation(s)
- Kazuhiro Kato
- Laboratory of Radiation Biology, Department of Applied Veterinary Sciences, Faculty of Veterinary Medicine, Hokkaido University, Sapporo, Japan
| | - Hironobu Yasui
- Laboratory of Radiation Biology, Department of Applied Veterinary Sciences, Faculty of Veterinary Medicine, Hokkaido University, Sapporo, Japan
- One Health Research Center, Hokkaido University, Sapporo, Japan
| | - Hideo Sato-Akaba
- Department of Electrical and Electronic Engineering, Graduate School of Integrated Science and Technology, Shizuoka University, Hamamatsu, Japan
| | - Miho C. Emoto
- Department of Clinical Laboratory Science, School of Medical Technology, Health Sciences University of Hokkaido, Sapporo, Japan
| | - Hirotada G. Fujii
- Advanced Research Promotion Center, Health Sciences University of Hokkaido, Ishikari, Japan
| | - Maciej M. Kmiec
- Departments of Radiology and Radiation Oncology, Geisel School of Medicine, Dartmouth College, Hanover, NH, USA
| | - Periannan Kuppusamy
- Departments of Radiology and Radiation Oncology, Geisel School of Medicine, Dartmouth College, Hanover, NH, USA
| | - Masaki Nagane
- Laboratory of Biochemistry, School of Veterinary Medicine, Azabu University, Sagamihara, Japan
| | - Tadashi Yamashita
- Laboratory of Biochemistry, School of Veterinary Medicine, Azabu University, Sagamihara, Japan
| | - Osamu Inanami
- Laboratory of Radiation Biology, Department of Applied Veterinary Sciences, Faculty of Veterinary Medicine, Hokkaido University, Sapporo, Japan
| |
Collapse
|
43
|
Wencker FDR, Lyon SE, Breaker RR. Evidence that ribosomal protein bS21 is a component of the OLE ribonucleoprotein complex. RNA Biol 2025; 22:1-14. [PMID: 40322971 PMCID: PMC12054373 DOI: 10.1080/15476286.2025.2491842] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Revised: 03/25/2025] [Accepted: 04/03/2025] [Indexed: 05/08/2025] Open
Abstract
OLE RNAs represent a large and highly structured noncoding RNA (ncRNA) class that is mostly found in Gram-positive extremophiles and/or anaerobes of the Bacillota phylum. These ~600-nucleotide RNAs are among the most structurally complex and well-conserved large ncRNAs whose precise biochemical functions remain to be established. In Halalkalibacterium halodurans, OLE RNA is involved in the adaptation to various unfavourable growth conditions, including exposure to cold (≤20°C), ethanol (≥3% [v/v]), excess Mg2+ (≥4 mM), and non-glucose carbon/energy sources. OLE forms a ribonucleoprotein (RNP) complex with the OLE-associated proteins A, B and C, which are known to be essential for OLE RNP complex function in this species. Bacteria lacking OLE RNA (Δole) or a functional OLE RNP complex exhibit growth defects under the stresses listed above. Here, we demonstrate that ribosomal protein bS21 is a natural component of the OLE RNP complex and we map its precise RNA binding site. The presence of bS21 results in a conformational change in OLE RNA resembling a k-turn substructure previously reported to be relevant to the function of the OLE RNP complex. Mutational disruption of the bS21 protein or its OLE RNA binding site results in growth inhibition under cold and ethanol stress to the same extent as the deletion of the gene for OLE RNA. These findings are consistent with the hypothesis that bS21 is a biologically relevant component of the OLE RNP complex under a subset of stresses managed by the OLE RNP complex.
Collapse
Affiliation(s)
- Freya D. R. Wencker
- Department of Molecular, Cellular and Developmental Biology, Yale University, New Haven, CT, USA
| | - Seth E. Lyon
- Department of Molecular Biophysics and Biochemistry, Yale University, New Haven, CT, USA
| | - Ronald R. Breaker
- Department of Molecular, Cellular and Developmental Biology, Yale University, New Haven, CT, USA
- Department of Molecular Biophysics and Biochemistry, Yale University, New Haven, CT, USA
| |
Collapse
|
44
|
Guo H, Huang G, Long H, Wu W, Lin K, Qiao B, Zhang N, Huang T, Tan Y, Zhang Q, Zhang M, Xie X, Shuai X, Xu M, Zhang C. Harnessing PD-1-overexpressing macrophage membrane for preparation of lenvatinib-loaded vesicles to boost immunotherapy against HCC recurrence after radiofrequency ablation. Biomaterials 2025; 323:123433. [PMID: 40424831 DOI: 10.1016/j.biomaterials.2025.123433] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2025] [Revised: 05/20/2025] [Accepted: 05/20/2025] [Indexed: 05/29/2025]
Abstract
Hepatocellular carcinoma (HCC) is characterized by high malignancy, high recurrence rate and poor prognosis. Radiofrequency ablation (RFA) is the first-line curative treatment for early-stage HCC. Yet, effective inhibition of local recurrent HCC is still challenging because of immunosuppressive tumor microenvironment (TME) and upregulation of multiple tyrosine kinase receptors in the post-RFA residual tumor. The combination of tyrosine kinase inhibitor lenvatinib and immune checkpoint blockade (ICB) therapy is a promising strategy to tackle HCC, but the limited bioavailability and weak targeting still restrict the therapeutic effect. Inspired by the predominant proinflammatory stress reaction and infiltration of macrophages in the TME of residual HCC after RFA, we developed a lenvatinib-loaded hybrid nanovesicles (PML@Len) consisting of lipid and engineered macrophage membrane overexpressing programmed cell death protein 1 (PD-1). The incorporation of macrophage membrane prevented PML@Len from being phagocytosed by kupffer cells. The replenished PD-1 not only facilitated tumor accumulation but also blocked programmed cell death ligand 1(PD-L1) overexpressed on the tumor. Additionally, delivery of lenvatinib by PML@Len resulted in effective anti-angiogenesis and regulation of immunosuppressive TME to boost anti-tumor immunity. Consequently, these hybrid nanovesicles based on engineered macrophage membrane demonstrated great potency to elicit anti-tumor memory effects of T lymphocytes, hence effectively suppressing the tumor recurrence after RFA.
Collapse
Affiliation(s)
- Huanling Guo
- Department of Medical Ultrasonics, Institute of Diagnostic and Interventional Ultrasound, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Guangliang Huang
- Department of Medical Ultrasonics, Institute of Diagnostic and Interventional Ultrasound, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Haiyi Long
- Department of Medical Ultrasonics, Institute of Diagnostic and Interventional Ultrasound, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Wenxin Wu
- Department of Medical Ultrasonics, Institute of Diagnostic and Interventional Ultrasound, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Ke Lin
- Department of Medical Ultrasonics, Institute of Diagnostic and Interventional Ultrasound, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Bin Qiao
- Department of Medical Ultrasonics, Institute of Diagnostic and Interventional Ultrasound, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Nan Zhang
- Department of Medical Ultrasonics, Institute of Diagnostic and Interventional Ultrasound, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Tongyi Huang
- Department of Medical Ultrasonics, Institute of Diagnostic and Interventional Ultrasound, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Yang Tan
- Department of Medical Ultrasonics, Institute of Diagnostic and Interventional Ultrasound, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Qi Zhang
- Department of Medical Ultrasonics, Institute of Diagnostic and Interventional Ultrasound, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Minru Zhang
- Department of Medical Ultrasonics, Institute of Diagnostic and Interventional Ultrasound, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Xiaoyan Xie
- Department of Medical Ultrasonics, Institute of Diagnostic and Interventional Ultrasound, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China.
| | - Xintao Shuai
- Nanomedicine Research Center, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510006, China; PCFM Lab of Ministry of Education School of Materials Science and Engineering, Sun Yat-sen University, Guangzhou, China.
| | - Ming Xu
- Department of Medical Ultrasonics, Institute of Diagnostic and Interventional Ultrasound, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China.
| | - Chunyang Zhang
- Department of Medical Ultrasonics, Institute of Diagnostic and Interventional Ultrasound, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China; Institute of Precision Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China.
| |
Collapse
|
45
|
Jaichand V, Lanrewaju AA, Baijnath H, Sabiu S, Mohanlall V. Profiling and cheminformatics bioprospection of curcurbitacin I and momordin Ic from Momordica balsamina on α-amylase and α-glucosidase. J Enzyme Inhib Med Chem 2025; 40:2492706. [PMID: 40302171 PMCID: PMC12044915 DOI: 10.1080/14756366.2025.2492706] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2025] [Revised: 03/18/2025] [Accepted: 04/08/2025] [Indexed: 05/01/2025] Open
Abstract
Momordica spp. has been traditionally used to manage type 2 diabetes mellitus, but the mechanisms and metabolites remain unclear. This study evaluated the inhibitory potential of Momordica balsamina extracts on α-amylase and α-glucosidase in vitro, identifying cucurbitacin I and momordin Ic via high-performance liquid chromatography-photo diode array, and their inhibitory potential in silico. Ethyl acetate seed extract (14.46 µg/ml) and hexane fruit flesh extract (16.79 µg/ml) exhibited lower IC50 values against α-amylase and α-glucosidase, respectively, compared to acarbose (reference standard). Comparatively, momordin Ic concentrations (36.57-605.98 µg/ml) were higher than cucurbitacin I (17.08-44.34 µg/ml). A 140 ns simulation showed that cucurbitacin I (-63.06 kcal/mol) and momordin Ic (-66.53 kcal/mol) exhibited stronger binding to α-amylase than acarbose (-36.46 kcal/mol), whereas cucurbitacin I (-38.08 kcal/mol) and momordin Ic (-54.87 kcal/mol) displayed weaker binding to α-glucosidase, relative to acarbose (-63.73 kcal/mol). Generally, momordin Ic demonstrated better thermodynamic properties, hence further in vitro and in vivo studies are needed to validate their antidiabetic potential.
Collapse
Affiliation(s)
- Viruska Jaichand
- Department of Biotechnology and Food Science, Faculty of Applied Science, Durban University of Technology, Durban, South Africa
| | - Adedayo Ayodeji Lanrewaju
- Department of Biotechnology and Food Science, Faculty of Applied Science, Durban University of Technology, Durban, South Africa
| | - Himansu Baijnath
- Ward Herbarium, School of Life Sciences, University of KwaZulu-Natal, Durban, South Africa
| | - Saheed Sabiu
- Department of Biotechnology and Food Science, Faculty of Applied Science, Durban University of Technology, Durban, South Africa
| | - Viresh Mohanlall
- Department of Biotechnology and Food Science, Faculty of Applied Science, Durban University of Technology, Durban, South Africa
| |
Collapse
|
46
|
Wen M, Yu A, Park Y, Calarese D, Gerber HP, Yin G. Homogeneous antibody-drug conjugates with dual payloads: potential, methods and considerations. MAbs 2025; 17:2498162. [PMID: 40322862 PMCID: PMC12054377 DOI: 10.1080/19420862.2025.2498162] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2025] [Revised: 04/18/2025] [Accepted: 04/21/2025] [Indexed: 05/08/2025] Open
Abstract
The development of site-specific dual-payload antibody-drug conjugates (ADCs) represents a potential advancement in targeted cancer therapy, enabling the simultaneous delivery of two distinct drugs into the same cancer cells to overcome payload resistance and enhance therapeutic efficacy. Here, we examine various methodologies for achieving site-specific dual-payload conjugation, including the use of multi-functional linkers, canonical amino acids, non-canonical amino acids, and enzyme-mediated methods, all of which facilitate precise control over payload attachment while ensuring homogeneity. We explore the implications of different conjugation techniques on drug-to-antibody ratios and the ratios of the two payloads, as well as their impact on process complexity and manufacturability. Additionally, we address the potential advantages of dual-payload ADCs compared to ADCs combined with traditional chemotherapy or single-payload ADC/ADC combinations. By evaluating these innovative methods, we aim to provide a comprehensive understanding of the current landscape in dual-payload ADC development and outline emerging directions necessary for further advancement of this promising therapeutic strategy.
Collapse
Affiliation(s)
- Miao Wen
- Sutro Biopharma Inc, South San Francisco, CA, USA
| | - Abigail Yu
- Sutro Biopharma Inc, South San Francisco, CA, USA
| | - Young Park
- Sutro Biopharma Inc, South San Francisco, CA, USA
| | | | | | - Gang Yin
- Sutro Biopharma Inc, South San Francisco, CA, USA
| |
Collapse
|
47
|
Li X, Xiao X, Wang S, Wu B, Zhou Y, Deng P. Uncovering de novo polyamine biosynthesis in the gut microbiome and its alteration in inflammatory bowel disease. Gut Microbes 2025; 17:2464225. [PMID: 39924644 PMCID: PMC11812404 DOI: 10.1080/19490976.2025.2464225] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/01/2024] [Revised: 01/17/2025] [Accepted: 01/26/2025] [Indexed: 02/11/2025] Open
Abstract
Polyamines are important gut microbial metabolites known to affect host physiology, yet the mechanisms behind their microbial production remain incompletely understood. In this study, we developed a stable isotope-resolved metabolomic (SIRM) approach to track polyamine biosynthesis in the gut microbiome. Viable microbial cells were extracted from fresh human and mouse feces and incubated anaerobically with [U-13C]-labeled inulin (tracer). Liquid chromatography-high resolution mass spectrometry analysis revealed distinct 13C enrichment profiles for spermidine (SPD) and putrescine (PUT), indicating that the arginine-agmatine-SPD pathway contributes to SPD biosynthesis in addition to the well-known spermidine synthase pathway (PUT aminopropylation). Species differences were observed in the 13C enrichments of polyamines and related metabolites between the human and mouse microbiome. By analyzing the fecal metabolomics and metatranscriptomic data from an inflammatory bowel disease (IBD) cohort, we found significantly higher polyamine levels in IBD patients compared to healthy controls. Further investigations using single-strain SIRM and in silico analyses identified Bacteroides spp. as key contributors to polyamine biosynthesis, harboring essential genes for this process and potentially driving the upregulation of polyamines in IBD. Taken together, this study expands our understanding of polyamine biosynthesis in the gut microbiome and will facilitate the development of precision therapies to target polyamine-associated diseases.
Collapse
Affiliation(s)
- Xinwei Li
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and College of Pharmaceutical Sciences, Soochow University, Suzhou, Jiangsu, China
- Department of Pharmaceutical Analysis, Soochow University, Suzhou, Jiangsu, China
| | - Xia Xiao
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and College of Pharmaceutical Sciences, Soochow University, Suzhou, Jiangsu, China
- Department of Pharmaceutical Analysis, Soochow University, Suzhou, Jiangsu, China
| | - Shengnan Wang
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and College of Pharmaceutical Sciences, Soochow University, Suzhou, Jiangsu, China
- Department of Pharmaceutical Analysis, Soochow University, Suzhou, Jiangsu, China
| | - Biyu Wu
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and College of Pharmaceutical Sciences, Soochow University, Suzhou, Jiangsu, China
- Department of Pharmaceutical Analysis, Soochow University, Suzhou, Jiangsu, China
| | - Yixuan Zhou
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and College of Pharmaceutical Sciences, Soochow University, Suzhou, Jiangsu, China
- Department of Pharmaceutical Analysis, Soochow University, Suzhou, Jiangsu, China
| | - Pan Deng
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and College of Pharmaceutical Sciences, Soochow University, Suzhou, Jiangsu, China
- Department of Pharmaceutical Analysis, Soochow University, Suzhou, Jiangsu, China
| |
Collapse
|
48
|
Lu Y, Partleton D, Gugu FM, Alhejaili AYG, Norris S, Harburn JJ, Gill JH, Sellars JD, Brown AK. Structural isomerisation affects the antitubercular activity of adamantyl-isoxyl adducts. J Enzyme Inhib Med Chem 2025; 40:2502600. [PMID: 40396606 PMCID: PMC12096669 DOI: 10.1080/14756366.2025.2502600] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2025] [Revised: 04/07/2025] [Accepted: 04/23/2025] [Indexed: 05/22/2025] Open
Abstract
Despite efforts to discover effective treatments to eradicate tuberculosis (TB), it remains a global threat. The increase in drug-resistant bacterial species has made the discovery of new drugs highly coveted. The utilisation of previous efficacious structures is one approach that can be employed to developing novel series of compounds to combat this ever-growing problem. This study sought to re-examine two such compounds, isoxyl (ISO) and SQ109, previously shown to be efficacious in TB treatment. SQ109-ISO hybrid compounds were shown to have demonstrable activity against both drug-sensitive and drug-resistant Mtb whilst displaying limited toxicity in vitro in comparison to other antitubercular agents. Indications from our genetic and biochemical studies suggest that these structurally similar pharmacophores bind to different proteins within Mtb, highlighting the need for careful consideration when producing regioisomeric analogues and that the utilisation of previous efficacious structures is a valid approach to developing promising novel drugs against Mtb.
Collapse
Affiliation(s)
- Yucheng Lu
- Biosciences Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, UK
| | - Daniel Partleton
- Biosciences Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, UK
| | - Filibus M. Gugu
- Biosciences Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, UK
- Department of Microbiology, Plateau State University Bokkos, Jos, Nigeria
| | - Ahmed Y. G. Alhejaili
- Biosciences Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, UK
| | - Samuel Norris
- Chemistry, School of Natural and Environmental Sciences, Newcastle University, Newcastle upon Tyne, UK
| | - J. Jonathan Harburn
- School of Pharmacy, Faculty of Medical Sciences, King George VI Building, Newcastle upon Tyne, UK
| | - Jason H. Gill
- School of Pharmacy, Faculty of Medical Sciences, King George VI Building, Newcastle upon Tyne, UK
- Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, UK
| | - Jonathan D. Sellars
- Biosciences Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, UK
- School of Pharmacy, Faculty of Medical Sciences, King George VI Building, Newcastle upon Tyne, UK
| | - Alistair K. Brown
- Biosciences Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, UK
| |
Collapse
|
49
|
Dong Y, Liu X, Xiong S, Cao M, Wu H, Chen L, Zhao M, Zheng Y, Zhang Z, Liu Y, Li Y, Qu Q, Dong C. Guanosine enhances the bactericidal effect of ceftiofur sodium on Streptococcus suis by activating bacterial metabolism. Virulence 2025; 16:2453525. [PMID: 39915976 PMCID: PMC11810099 DOI: 10.1080/21505594.2025.2453525] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2024] [Revised: 12/09/2024] [Accepted: 12/29/2024] [Indexed: 02/12/2025] Open
Abstract
The emergence and rapid development of antibiotic resistance poses a serious threat to global public health. Streptococcus suis (S. suis) is an important zoonotic pathogen, and the development of its antibiotic resistance has made the infections difficult to treat. The combination of non-antibiotic compounds with antibiotics is considered a promising strategy against multidrug-resistant bacteria. However, the mechanism by which metabolites act as antibiotic adjuvant remains unclear. Here, we found that guanosine metabolism was repressed in multidrug-resistant S. suis. Exogenous guanosine promoted the antibacterial effects of ceftiofur sodium (CEF) in vitro and in vivo. Furthermore, we demonstrated that exogenous guanosine promoted the biosynthesis of purine pathway, TCA cycle and bacterial respiration, which make bacteria more sensitive to the killing effect of antibacterial. In addition, the function of the cell membrane is affected by guanosine and the accumulation of antimicrobials in the bacteria increased. Bacterial-oxidative stress and DNA damage induced by guanosine is also one of the mechanisms by which the antibacterial effect is enhanced. These results suggest that guanosine is a promising adjuvant for antibacterial drugs and provide new theoretical basis for the clinical treatment of S. suis infection.
Collapse
Affiliation(s)
- Yue Dong
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, Heilongjiang, China
- Heilongjiang Key Laboratory for Animal Disease Control and Pharmaceutical Development, Northeast Agricultural University, Harbin, P R China
| | - Xiaona Liu
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, Heilongjiang, China
- Heilongjiang Key Laboratory for Animal Disease Control and Pharmaceutical Development, Northeast Agricultural University, Harbin, P R China
| | - Shanshan Xiong
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, Heilongjiang, China
- Heilongjiang Key Laboratory for Animal Disease Control and Pharmaceutical Development, Northeast Agricultural University, Harbin, P R China
| | - Mingyu Cao
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, Heilongjiang, China
- Heilongjiang Key Laboratory for Animal Disease Control and Pharmaceutical Development, Northeast Agricultural University, Harbin, P R China
| | - Haojie Wu
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, Heilongjiang, China
- Heilongjiang Key Laboratory for Animal Disease Control and Pharmaceutical Development, Northeast Agricultural University, Harbin, P R China
| | - Long Chen
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, Heilongjiang, China
- Heilongjiang Key Laboratory for Animal Disease Control and Pharmaceutical Development, Northeast Agricultural University, Harbin, P R China
| | - Mengmeng Zhao
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, Heilongjiang, China
- Heilongjiang Key Laboratory for Animal Disease Control and Pharmaceutical Development, Northeast Agricultural University, Harbin, P R China
| | - Yadan Zheng
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, Heilongjiang, China
- Heilongjiang Key Laboratory for Animal Disease Control and Pharmaceutical Development, Northeast Agricultural University, Harbin, P R China
| | - Zhiyun Zhang
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, Heilongjiang, China
- Heilongjiang Key Laboratory for Animal Disease Control and Pharmaceutical Development, Northeast Agricultural University, Harbin, P R China
| | - Yanyan Liu
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, Heilongjiang, China
- Heilongjiang Key Laboratory for Animal Disease Control and Pharmaceutical Development, Northeast Agricultural University, Harbin, P R China
| | - Yanhua Li
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, Heilongjiang, China
- Heilongjiang Key Laboratory for Animal Disease Control and Pharmaceutical Development, Northeast Agricultural University, Harbin, P R China
| | - Qianwei Qu
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, Heilongjiang, China
- Heilongjiang Key Laboratory for Animal Disease Control and Pharmaceutical Development, Northeast Agricultural University, Harbin, P R China
| | - Chunliu Dong
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, Heilongjiang, China
- Heilongjiang Key Laboratory for Animal Disease Control and Pharmaceutical Development, Northeast Agricultural University, Harbin, P R China
| |
Collapse
|
50
|
Toivanen K, De Sutter L, Wozniak A, Wyns K, Merikoski N, Salmikangas S, Duan J, Maksimow M, Lahtinen M, Böhling T, Schöffski P, Sihto H. Pharmacokinetic profile and in vivo anticancer efficacy of anagrelide administered subcutaneously in rodents. Drug Deliv 2025; 32:2463433. [PMID: 39930717 PMCID: PMC11816618 DOI: 10.1080/10717544.2025.2463433] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Revised: 12/18/2024] [Accepted: 01/31/2025] [Indexed: 02/14/2025] Open
Abstract
Anagrelide (ANA) is a phosphodiesterase 3A (PDE3A) inhibitor, commonly prescribed for essential thrombocythemia. It also functions as a molecular glue, inducing complex formation between PDE3A and Schlafen 12. This association either triggers apoptosis or inhibits proliferation in tumor cells, supporting its use in cancer therapy. Conventionally administered orally, ANA undergoes rapid metabolism and elimination, resulting in a short drug exposure time at the site of action. Here, we explored the pharmacokinetic profile of a subcutaneously (SC) injected ANA formulation in Sprague-Dawley rats by quantifying plasma ANA and metabolite concentrations using liquid-chromatography-tandem mass spectrometry. We further evaluated the in vivo tumor regression efficacy of orally and SC administered ANA in a patient-derived gastrointestinal stromal xenograft mouse model - UZLX-GIST2B - characterized by a KIT exon 9 driver mutation. The SC ANA exhibited extended-release plasma concentration-time profiles compared to intravenous and oral administrations. After a single administration in rats, plasma concentrations of ANA were detected up to 56 days later, and ANA metabolites up to 30 days later. The SC formulation also significantly reduced tumor volumes and demonstrated dose-dependent histological responses, nearly eradicating tumor tissue in 11 days with the highest dose. These findings suggest that the SC slow-release formulation maintains stable drug concentrations during treatment, potentially improving therapeutic efficacy at the target site.
Collapse
Affiliation(s)
- Kirsi Toivanen
- Department of Pathology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Luna De Sutter
- Department of Oncology, Laboratory of Experimental Oncology, KU Leuven, Leuven Cancer Institute, Leuven, Belgium
| | - Agnieszka Wozniak
- Department of Oncology, Laboratory of Experimental Oncology, KU Leuven, Leuven Cancer Institute, Leuven, Belgium
| | - Karo Wyns
- Department of Oncology, Laboratory of Experimental Oncology, KU Leuven, Leuven Cancer Institute, Leuven, Belgium
| | - Nanna Merikoski
- Department of Pathology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Sami Salmikangas
- Department of Pathology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Jianmin Duan
- Duan Pharmaceutical Consulting Inc., Laval, Canada
| | | | | | - Tom Böhling
- Department of Pathology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Patrick Schöffski
- Department of Oncology, Laboratory of Experimental Oncology, KU Leuven, Leuven Cancer Institute, Leuven, Belgium
| | - Harri Sihto
- Department of Pathology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| |
Collapse
|