1
|
Wen C, Zhang X, Kantapan J, Yu Z, Yuan L, Liu S, Li H, Liang S, Wei Y, Luo G, Xiao W, Dechsupa N, Lü M. Pentagalloyl glucose targets the JAK1/JAK3-STAT3 pathway to inhibit cancer stem cells and epithelial-mesenchymal transition in 5-fluorouracil-resistant colorectal cancer. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 142:156773. [PMID: 40378534 DOI: 10.1016/j.phymed.2025.156773] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/03/2025] [Revised: 03/25/2025] [Accepted: 04/13/2025] [Indexed: 05/19/2025]
Abstract
BACKGROUND Colorectal cancer (CRC) resistance to 5-fluorouracil (5-FU), primarily driven by cancer stem cells (CSCs) and epithelial-mesenchymal transition (EMT), remains a major clinical challenge, necessitating novel therapeutic strategies. PURPOSE This study aims to evaluate the therapeutic potential of pentagalloyl glucose (PGG), a bioactive compound derived from Bouea macrophylla seeds, in overcoming 5-FU resistance in CRC. METHOD Anti-tumor effects of PGG were investigated using two- and three-dimensional (2D and 3D) cell culture models and subcutaneous xenograft and metastatic mouse models. Transcriptome sequencing, western blotting, and pharmacological inhibitors were employed to elucidate the underlying molecular mechanisms. RESULTS PGG demonstrated potent anti-CSC activity; suppressed EMT-driven invasion and metastasis; and induced apoptosis in 2D monolayers, 3D spheroid models, and xenograft tumor models. Mechanistically, PGG selectively inhibited the JAK1/JAK3-STAT3 signaling pathway, considerably reducing STAT3 phosphorylation. This disruption downregulated the expression of CSC markers (CD133 and CD44), EMT regulators (N-cadherin and vimentin), and anti-apoptotic proteins (Bcl-2), effectively sensitizing 5-FU-resistant CRC to therapy. CONCLUSION PGG inhibit dual-target of CSCs and EMT via JAK1/JAK3-STAT3 signaling pathway in 5-FU-resistant CRC, providing a novel therapeutic approach to overcome chemoresistance.
Collapse
Affiliation(s)
- Chengli Wen
- Molecular Imaging and Therapy Research Unit, Department of Radiologic Technology, Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai 50200, Thailand; Department of Critical Care Medicine, The Affiliated Hospital, Southwest Medical University, Luzhou 646000, China.
| | - Xu Zhang
- Department of Gastroenterology, Honghuagang District People's Hospital of Zunyi City, Zunyi 563000, China.
| | - Jiraporn Kantapan
- Molecular Imaging and Therapy Research Unit, Department of Radiologic Technology, Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai 50200, Thailand.
| | - Zehui Yu
- Laboratory Animal Center, Southwest Medical University, Luzhou 646000, China; Human Microecology and Precision Diagnosis and Treatment of Luzhou Key Laboratory, Luzhou 646000, China.
| | - Liping Yuan
- Department of Gastroenterology, The Affiliated Hospital, Southwest Medical University, Luzhou 646000, China; Department of Gastroenterology, The First People's Hospital of Liangshan Yi Autonomous Prefecture, Xichang 615000, China.
| | - Sha Liu
- Human Microecology and Precision Diagnosis and Treatment of Luzhou Key Laboratory, Luzhou 646000, China; The Clinical Medicine Research Center, The Affiliated Hospital of Southwest Medical University, Luzhou 646000, China.
| | - Hao Li
- Human Microecology and Precision Diagnosis and Treatment of Luzhou Key Laboratory, Luzhou 646000, China; Department of Gastroenterology, The Affiliated Hospital, Southwest Medical University, Luzhou 646000, China.
| | - Sicheng Liang
- Human Microecology and Precision Diagnosis and Treatment of Luzhou Key Laboratory, Luzhou 646000, China; Department of Gastroenterology, The Affiliated Hospital, Southwest Medical University, Luzhou 646000, China.
| | - Yan Wei
- Key Laboratory of Medical Electrophysiology of Ministry of Education and Medical Electrophysiological Key Laboratory of Sichuan Province, Collaborative Innovation Center for Prevention and Treatment of Cardiovascular Disease, Institute of Cardiovascular Research, Southwest Medical University, Luzhou 646000, China.
| | - Gang Luo
- Department of Gastroenterology, The Affiliated Hospital, Southwest Medical University, Luzhou 646000, China.
| | - Wanmeng Xiao
- Human Microecology and Precision Diagnosis and Treatment of Luzhou Key Laboratory, Luzhou 646000, China; Department of Gastroenterology, The Affiliated Hospital, Southwest Medical University, Luzhou 646000, China; The Clinical Medicine Research Center, The Affiliated Hospital of Southwest Medical University, Luzhou 646000, China.
| | - Nathupakorn Dechsupa
- Molecular Imaging and Therapy Research Unit, Department of Radiologic Technology, Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai 50200, Thailand.
| | - Muhan Lü
- Human Microecology and Precision Diagnosis and Treatment of Luzhou Key Laboratory, Luzhou 646000, China; Department of Gastroenterology, The Affiliated Hospital, Southwest Medical University, Luzhou 646000, China.
| |
Collapse
|
2
|
Sepehri S, De Win D, Heymans A, Van Goethem F, Rodrigues RM, Rogiers V, Vanhaecke T. Next generation risk assessment of hair dye HC yellow no. 13: Ensuring protection from liver steatogenic effects. Regul Toxicol Pharmacol 2025; 159:105794. [PMID: 40024558 DOI: 10.1016/j.yrtph.2025.105794] [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: 10/09/2024] [Revised: 02/17/2025] [Accepted: 02/27/2025] [Indexed: 03/04/2025]
Abstract
This study employs animal-free Next Generation Risk Assessment (NGRA) principles to evaluate the safety of repeated dermal exposure to 2.5% (w/w) HC Yellow No. 13 (HCY13) hair dye. As multiple in silico tools consistently flagged hepatotoxic potential, likely due to HCY13's trifluoromethyl group, which is known to interfere with hepatic lipid metabolism, liver steatosis was chosen as the primary mode of action for evaluation. AOP-guided in vitro tests were conducted, exposing human stem cell-derived hepatic cells to varying HCY13 concentrations over 72 h. The expression of 11 lipid metabolism-related marker genes (AHR, PPARA, LXRA, APOB, ACOX1, CPT1A, FASN, SCD1, DGAT2, CD36, and PPARG) and triglyceride accumulation, a phenotypic hallmark of steatosis, were measured. PROAST software was used to calculate in vitro Points of Departure (PoDNAM) for each biomarker. Using GastroPlus 9.9, physiologically-based pharmacokinetic (PBPK) models estimated internal liver concentrations (Cmax liver) of HCY13, ranging from 4 to 20 pM. All PoDNAM values significantly exceeded the predicted Cmax liver, indicating that HCY13 at 2.5% (w/w) is unlikely to induce liver steatosis under the assumed conditions. This research demonstrates the utility of NGRA, integrating AOP-based in vitro assays and computational models to protect human health and support regulatory decision-making without animal testing.
Collapse
Affiliation(s)
- Sara Sepehri
- Department of In Vitro Toxicology and Dermato-Cosmetology (IVTD), Vrije Universiteit Brussel, Brussels, Belgium.
| | - Dinja De Win
- Department of In Vitro Toxicology and Dermato-Cosmetology (IVTD), Vrije Universiteit Brussel, Brussels, Belgium.
| | - Anja Heymans
- Department of In Vitro Toxicology and Dermato-Cosmetology (IVTD), Vrije Universiteit Brussel, Brussels, Belgium.
| | - Freddy Van Goethem
- Department of In Vitro Toxicology and Dermato-Cosmetology (IVTD), Vrije Universiteit Brussel, Brussels, Belgium.
| | - Robim M Rodrigues
- Department of In Vitro Toxicology and Dermato-Cosmetology (IVTD), Vrije Universiteit Brussel, Brussels, Belgium.
| | - Vera Rogiers
- Department of In Vitro Toxicology and Dermato-Cosmetology (IVTD), Vrije Universiteit Brussel, Brussels, Belgium.
| | - Tamara Vanhaecke
- Department of In Vitro Toxicology and Dermato-Cosmetology (IVTD), Vrije Universiteit Brussel, Brussels, Belgium.
| |
Collapse
|
3
|
Wei X, Liu R, Li W, Yu Q, Yang QT, Li T. Advances in research regarding epithelial-mesenchymal transition and prostate cancer. Front Cell Dev Biol 2025; 13:1583255. [PMID: 40519259 PMCID: PMC12162976 DOI: 10.3389/fcell.2025.1583255] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2025] [Accepted: 05/09/2025] [Indexed: 06/18/2025] Open
Abstract
Prostate cancer (PCa) is the most prevalent cancer in men and the fifth leading cause of cancer-related mortality among men globally. Despite substantial advancements in patient prognosis attributable to improvements in PCa treatment, individuals with metastatic castration-resistant prostate cancer continue to experience poor outcomes. Epithelial-mesenchymal transition (EMT) is characterized as a cellular event in which epithelial cells adopt a mesenchymal phenotype while simultaneously losing their epithelial characteristics. EMT has been demonstrated to be associated with the progression of PCa, encompassing tumor metastasis, recurrence, drug resistance, and the development of an immunosuppressive microenvironment. Consequently, this review synthesizes recent studies on EMT in PCa, consolidating the events mediated by EMT in the progression of PCa and the molecular mechanisms linked to EMT activation in this context.
Collapse
Affiliation(s)
| | | | | | | | | | - Tao Li
- Department of Urology, The Affiliated Hospital of Guizhou Medical University, Guiyang, China
| |
Collapse
|
4
|
Wood A, Atienzar F, Basili D, Coulet M, Fernandez R, Galano M, Marin-Kuan M, Montoya G, Piechota P, Punt A, Reale E, Wang S, Hepburn P. Countdown to 2027 - maximising use of NAMs in food safety assessment: closing the gap for regulatory assessments in Europe. Regul Toxicol Pharmacol 2025:105863. [PMID: 40449716 DOI: 10.1016/j.yrtph.2025.105863] [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: 03/12/2025] [Revised: 05/15/2025] [Accepted: 05/26/2025] [Indexed: 06/03/2025]
Abstract
Safety assessments of regulated food products in the European Union (EU) largely rely on experimental animal studies. Currently, the European Commission is developing a roadmap to phase out animal testing for chemical safety assessment across all relevant pieces of legislation, including foods, while the ambition of the European Food Safety Authority (EFSA) is that by 2027, new scientific developments, i.e., new approach/non-animal methods (NAMs), will be integrated into assessments leading to "the minimisation of animal testing". However, considering recent requests that have been made to conduct new animal studies for some regulated products, significant progress is required to minimise further and ultimately replace animal testing in the food safety environment. To advance this, we review several NAMs amenable for use in food safety assessment and reflect on their presence in EU food safety regulation and sectoral guidance. For many years, proposals to incorporate NAMs into food safety assessments have been made with questionable regulatory impact. Therefore, we present several amendments which could be made to the EU food regulatory system and current strategies towards phasing out animal testing which, if taken up, could lead to a tangible difference in the extent of animal testing within the food safety environment. Recognising that research may be required for some of these NAMs to enhance regulatory uptake, we propose potential follow-up projects that complement recent research & innovation (R&I) needs published by EFSA which food safety stakeholders could coordinate or participate in.
Collapse
Affiliation(s)
- Adam Wood
- Unilever Safety and Environmental Assurance Centre, Colworth Science Park, Sharnbrook, Bedfordshire MK44 1LQ, UK.
| | - Franck Atienzar
- Coca-Cola Services SA/NV, Chaussée De Mons 1424, 1070 Anderlecht, Belgium
| | - Danilo Basili
- Société des Produits Nestlé S.A. Nestlé Research - Rte du Jorat 57, 1000 Lausanne 26, Switzerland
| | - Myriam Coulet
- Société des Produits Nestlé S.A. Nestlé Research - Rte du Jorat 57, 1000 Lausanne 26, Switzerland
| | - Rebeca Fernandez
- FoodDrinkEurope, Avenue des Nerviens 9-31 - 1040 Brussels, Belgium
| | - Melina Galano
- dsm-firmenich, Alexander Fleminglaan 1, 2613 AX, Delft, the Netherlands
| | - Maricel Marin-Kuan
- Société des Produits Nestlé S.A. Nestlé Research - Rte du Jorat 57, 1000 Lausanne 26, Switzerland
| | - Gina Montoya
- Société des Produits Nestlé S.A. Nestlé Research - Rte du Jorat 57, 1000 Lausanne 26, Switzerland
| | - Przemyslaw Piechota
- Société des Produits Nestlé S.A. Nestlé Research - Rte du Jorat 57, 1000 Lausanne 26, Switzerland
| | - Ans Punt
- Unilever Safety and Environmental Assurance Centre, Colworth Science Park, Sharnbrook, Bedfordshire MK44 1LQ, UK
| | - Elena Reale
- Société des Produits Nestlé S.A. Nestlé Research - Rte du Jorat 57, 1000 Lausanne 26, Switzerland
| | - Si Wang
- PepsiCo International, Beaumont Park, 4 Leycroft Road Leicester LE4 1ET, UK
| | - Paul Hepburn
- Unilever Safety and Environmental Assurance Centre, Colworth Science Park, Sharnbrook, Bedfordshire MK44 1LQ, UK
| |
Collapse
|
5
|
Mizuno K, Takeshita JI, Harakawa Y, Hosaka T, Shizu R, Yoshinari K. Utility of in vitro assay data in read-across prediction of nongenotoxic carcinogenicity of pesticides for cytotoxicity-related rat tumors. Food Chem Toxicol 2025; 202:115566. [PMID: 40403954 DOI: 10.1016/j.fct.2025.115566] [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: 02/22/2025] [Revised: 04/30/2025] [Accepted: 05/19/2025] [Indexed: 05/24/2025]
Abstract
Developing alternative methods for rat carcinogenicity studies remains challenging. This study aimed to establish a read-across method to predict nongenotoxic carcinogenicity in rats using molecular descriptors and in vitro assays. Based on 2-year rat carcinogenicity study results of agrochemicals, 80 compounds that caused benign or malignant tumors in the liver, thyroid, testis, uterus, ovary, breast, nasal cavity, stomach, or bladder/urethra and 46 compounds that did not were selected and subjected to cell-based cytotoxicity assays. Here, we focused on tumors associated with epithelial cell injury (nasal cavity, stomach, and bladder/urethra tumors). The read-across prediction was performed using neighboring substances, which were selected based on the Euclidean distance between the substances calculated using molecular descriptors. In some cases, neighboring substances were further selected based on the concordance of the in vitro assay results. The selection of neighboring substances based on the carcinogenicity-relevant descriptors and then on the cytotoxicity assay data improved the prediction accuracy (balanced accuracy: 0.752-0.821) compared to the accuracy with substances selected based on unselected descriptors alone (balanced accuracy: 0.294-0.582). These results suggest that the combined use of carcinogenicity-relevant descriptors and in vitro assays related to carcinogenic mechanisms is useful for read-across prediction of cytotoxicity-related tumors in rats.
Collapse
Affiliation(s)
- Kosuke Mizuno
- School of Pharmaceutical Sciences, University of Shizuoka, Shizuoka, Japan
| | - Jun-Ichi Takeshita
- School of Pharmaceutical Sciences, University of Shizuoka, Shizuoka, Japan; Research Institute of Science for Safety and Sustainability, National Institute of Advanced Industrial Science and Technology (AIST), Tsukuba, Japan
| | - Yu Harakawa
- School of Pharmaceutical Sciences, University of Shizuoka, Shizuoka, Japan
| | - Takuomi Hosaka
- School of Pharmaceutical Sciences, University of Shizuoka, Shizuoka, Japan
| | - Ryota Shizu
- School of Pharmaceutical Sciences, University of Shizuoka, Shizuoka, Japan
| | - Kouichi Yoshinari
- School of Pharmaceutical Sciences, University of Shizuoka, Shizuoka, Japan.
| |
Collapse
|
6
|
Kunikullaya U K. An integrated approach to understanding the effects of exposome on neuroplasticity. Behav Brain Res 2025; 485:115516. [PMID: 40024484 DOI: 10.1016/j.bbr.2025.115516] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2024] [Revised: 02/08/2025] [Accepted: 02/27/2025] [Indexed: 03/04/2025]
Abstract
Anthropogenic factors are those that occur due to human activities. The exposome is proposed to complement the genome, wherein an individual's exposure begins before birth. The range of exposures includes physical, chemical, dietary, lifestyle, biological, and occupational sources. Exposome has a positive or negative influence on neuroplasticity during different stages of life. A comprehensive study of the exposome is thus necessary to incorporate these factors and their influence on the individual, community, and the population as a whole. Exposomic research and global health present significant opportunities for interdisciplinary research. This review gives an overview of the exposome and its influence on neuroplasticity. It proposes methods to study the exposome on neuroplasticity across the lifespan of the individual. This is possible with the use of self-reported data, large-scale cohort formation, physiological sensors, neuroimaging, omics, molecular biology, and systems approaches. These approaches aim to provide a holistic understanding of an individual's neurological well-being and its implications for the population at large. This will also enable the designing of novel preventive and treatment strategies for managing neurological disorders.
Collapse
Affiliation(s)
- Kirthana Kunikullaya U
- MeDH, Department of Medicine, Huddinge, Karolinska Universitetssjukhuset Huddinge, Stockholm 14186, Sweden.
| |
Collapse
|
7
|
Mo F, Wang M, Gao R, Gu T, Zheng K, Wang A, Qiu Y. Functional analysis of open stomata 1-slow anion channel associated 1-6 protein module in enhancing drought tolerance in tomato through stomatal regulation mechanisms. Int J Biol Macromol 2025; 308:142591. [PMID: 40157666 DOI: 10.1016/j.ijbiomac.2025.142591] [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: 12/29/2024] [Revised: 03/22/2025] [Accepted: 03/26/2025] [Indexed: 04/01/2025]
Abstract
Tomato (Solanum lycopersicum) is an important vegetable crop, whose growth and development are frequently subjected to drought stress, which severely limits its growth and yield. Identifying key drought-resistance genes in tomato is crucial for elucidating the mechanisms of drought resistance and improving tomato's drought tolerance, which has practical implications for agricultural production. The results of this study demonstrate that silencing SlSLAC1-6 (Slow anion channel associated 1-6) reduces tomato's drought tolerance. SnRK2.6/OST1 (Open stomata 1) protein kinase is a key component in plants' resistance to abiotic stress. Interactions between SlOST1 and SlSLAC1-6 were confirmed through Y2H, BiFC, LCI, Co-IP, and Pull-down assays. Simultaneously, overexpression and knockout of SlOST1 proved that it positively regulates tomato's drought tolerance by influencing reactive oxygen species (ROS) homeostasis, photosynthetic capacity, stomatal closure, and other mechanisms. Silencing SlSLAC1-6 in SlOST1 knockout plants further reduced tomato's drought tolerance. The regulation of tomato drought tolerance by SlOST1 and SlSLAC1-6 highlights the complexity of plant adaptation to drought. These findings provide new insights into the regulatory network of the SlOST1-SlSLAC1 protein module in tomato drought tolerance and offer gene resources for future tomato drought-resistance breeding.
Collapse
Affiliation(s)
- Fulei Mo
- College of Life Sciences, Northeast Agricultural University, Harbin 150030, China
| | - Meiliang Wang
- College of Life Sciences, Northeast Agricultural University, Harbin 150030, China
| | - Ruihua Gao
- College of Life Sciences, Northeast Agricultural University, Harbin 150030, China
| | - Tingting Gu
- College of Life Sciences, Northeast Agricultural University, Harbin 150030, China
| | - Kaiqi Zheng
- College of Life Sciences, Northeast Agricultural University, Harbin 150030, China
| | - Aoxue Wang
- College of Life Sciences, Northeast Agricultural University, Harbin 150030, China; College of Horticulture and landscape architecture, Northeast Agricultural University, Harbin 150030, China.
| | - Youwen Qiu
- College of Life Sciences, Northeast Agricultural University, Harbin 150030, China.
| |
Collapse
|
8
|
Gutleb AC, Murugadoss S, Stępnik M, SenGupta T, El Yamani N, Longhin EM, Olsen AKH, Wyrzykowska E, Jagiello K, Judzinska B, Cambier S, Honza T, McFadden E, Shaposhnikov S, Puzyn T, Serchi T, Weber P, Arnesdotter E, Skakalova V, Jirsova K, Grudzinski IP, Collins A, Rundén-Pran E, Dusinska M. New Approach Methods (NAMs) for genotoxicity assessment of nano- and advanced materials; Advantages and challenges. MUTATION RESEARCH. GENETIC TOXICOLOGY AND ENVIRONMENTAL MUTAGENESIS 2025; 904:503867. [PMID: 40382189 DOI: 10.1016/j.mrgentox.2025.503867] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Revised: 03/07/2025] [Accepted: 03/26/2025] [Indexed: 05/20/2025]
Abstract
Genotoxicity assessment is essential for ensuring chemical safety and mitigating risks to human health and the environment. Traditional methods, reliant on animal models, are time-consuming, costly, and raise ethical concerns. New Approach Methods (NAMs) offer innovative, cost-effective, and ethical alternatives, playing a pivotal role in both traditional and next-generation risk assessment (NGRA) by minimizing the need for animal testing, particularly in genotoxicity evaluations. However, the development of NAMs often overlooks the particular physicochemical properties of nanomaterials (NMs), which significantly influence their toxicological behaviour and can interfere with genotoxicity evaluation. This underscores an urgent need for the standardization and adaptation of NAMs to address nano- and advanced material-specific genotoxicity challenges. In this review, we summarize the challenges associated with genotoxicity testing of NMs and highlight the suitability of existing in vitro and in silico NAMs for NMs and advanced materials, enabling genotoxicity testing across various exposure routes and organ systems. Despite considerable progress, regulatory validation remains constrained by the absence of approved test guidelines and standardized protocols. To achieve regulatory acceptance, it is crucial to adapt NAMs to NM-specific exposure scenarios, refine test systems to better mimic human biology, develop tailored in vitro protocols, and ensure thorough characterisation of NMs both in pristine form and dispersed in culture medium. Collaborative efforts among scientists, regulators, industry, and advocacy groups are vital to improving the reliability and regulatory acceptance of NAMs. By addressing these challenges, NAMs have the potential to revolutionize genotoxicity risk assessment, advancing it towards a more sustainable, efficient and ethical framework.
Collapse
Affiliation(s)
- Arno C Gutleb
- Environmental Sustainability Assessment and Circularity (SUSTAIN) Unit, Luxembourg Institute of Science and Technology (LIST), Luxembourg; INVITROLIZE sarl, Wellenstein, Luxembourg.
| | - Sivakumar Murugadoss
- Health Effects Laboratory, Department of Environmental Chemistry and Health Effects, NILU The Climate and Environmental Research Institute, Norway.
| | - Maciej Stępnik
- QSAR Lab Ltd, Trzy Lipy 3, Gdańsk, Poland; Department of Toxicology, Faculty of Pharmacy, Medical University of Łódź, Poland.
| | - Tanima SenGupta
- Health Effects Laboratory, Department of Environmental Chemistry and Health Effects, NILU The Climate and Environmental Research Institute, Norway.
| | - Naouale El Yamani
- Health Effects Laboratory, Department of Environmental Chemistry and Health Effects, NILU The Climate and Environmental Research Institute, Norway.
| | - Eleonora Marta Longhin
- Health Effects Laboratory, Department of Environmental Chemistry and Health Effects, NILU The Climate and Environmental Research Institute, Norway.
| | - Ann-Karin Hardie Olsen
- Health Effects Laboratory, Department of Environmental Chemistry and Health Effects, NILU The Climate and Environmental Research Institute, Norway.
| | | | - Karolina Jagiello
- QSAR Lab Ltd, Trzy Lipy 3, Gdańsk, Poland; University of Gdańsk, Faculty of Chemistry, Laboratory of Environmental Chemoinformatics, Wita Stwosza, Gdańsk 63, 80-308, Poland.
| | - Beata Judzinska
- QSAR Lab Ltd, Trzy Lipy 3, Gdańsk, Poland; University of Gdańsk, Faculty of Chemistry, Laboratory of Environmental Chemoinformatics, Wita Stwosza, Gdańsk 63, 80-308, Poland.
| | - Sebastien Cambier
- Environmental Sustainability Assessment and Circularity (SUSTAIN) Unit, Luxembourg Institute of Science and Technology (LIST), Luxembourg.
| | - Tatiana Honza
- Health Effects Laboratory, Department of Environmental Chemistry and Health Effects, NILU The Climate and Environmental Research Institute, Norway.
| | - Erin McFadden
- Health Effects Laboratory, Department of Environmental Chemistry and Health Effects, NILU The Climate and Environmental Research Institute, Norway.
| | | | - Tomasz Puzyn
- QSAR Lab Ltd, Trzy Lipy 3, Gdańsk, Poland; University of Gdańsk, Faculty of Chemistry, Laboratory of Environmental Chemoinformatics, Wita Stwosza, Gdańsk 63, 80-308, Poland.
| | - Tommaso Serchi
- Environmental Sustainability Assessment and Circularity (SUSTAIN) Unit, Luxembourg Institute of Science and Technology (LIST), Luxembourg.
| | - Pamina Weber
- Environmental Sustainability Assessment and Circularity (SUSTAIN) Unit, Luxembourg Institute of Science and Technology (LIST), Luxembourg
| | - Emma Arnesdotter
- Environmental Sustainability Assessment and Circularity (SUSTAIN) Unit, Luxembourg Institute of Science and Technology (LIST), Luxembourg
| | | | - Katerina Jirsova
- Laboratory of Biology and Pathology of the Eye, Institute of Biology and Medical Genetics, 1st Faculty of Medicine and General Teaching Hospital, Charles University, Prague, Czech Republic.
| | - Ireneusz P Grudzinski
- Department of Toxicology and Food Science, Faculty of Pharmacy, Medical University of Warsaw, Banacha Str. 1, Warsaw 02-097, Poland.
| | - Andrew Collins
- NorGenoTech, Norway; Department of Nutrition, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway.
| | - Elise Rundén-Pran
- Health Effects Laboratory, Department of Environmental Chemistry and Health Effects, NILU The Climate and Environmental Research Institute, Norway.
| | - Maria Dusinska
- Health Effects Laboratory, Department of Environmental Chemistry and Health Effects, NILU The Climate and Environmental Research Institute, Norway.
| |
Collapse
|
9
|
Rodríguez-Rodríguez R, Baena M, Zagmutt S, Paraiso WK, Reguera AC, Fadó R, Casals N. International Union of Basic and Clinical Pharmacology. CXIX. Fundamental insights and clinical relevance regarding the carnitine palmitoyltransferase family of enzymes. Pharmacol Rev 2025; 77:100051. [PMID: 40106976 DOI: 10.1016/j.pharmr.2025.100051] [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: 07/09/2024] [Accepted: 02/14/2025] [Indexed: 03/22/2025] Open
Abstract
The carnitine palmitoyltransferases (CPTs) play a key role in controlling the oxidation of long-chain fatty acids and are potential therapeutic targets for diseases with a strong metabolic component, such as obesity, diabetes, and cancer. Four distinct proteins are CPT1A, CPT1B, CPT1C, and CPT2, differing in tissue expression and catalytic activity. CPT1s are finely regulated by malonyl-CoA, a metabolite whose intracellular levels reflect the cell's nutritional state. Although CPT1C does not exhibit significant catalytic activity, it is capable of modulating the functioning of other neuronal proteins. Structurally, all CPTs share a Y-shaped catalytic tunnel that allows the entry of 2 substrates and accommodation of the acyl group in a hydrophobic pocket. Several molecules targeting these enzymes have been described, some showing potential in normalizing blood glucose levels in diabetic patients, and others that, through a central mechanism, are anorexigenic and enhance energy expenditure. However, given the critical roles that CPTs play in certain tissues, such as the heart, liver, and brain, it is essential to fully understand the differences between the various isoforms. We analyze in detail the structure of these proteins, their cellular and physiological functions, and their potential as therapeutic targets in diseases such as obesity, diabetes, and cancer. We also describe drugs identified to date as having inhibitory or activating capabilities for these proteins. This knowledge will support the design of new drugs specific to each isoform, and the development of nanomedicines that can selectively target particular tissues or cells. SIGNIFICANCE STATEMENT: Carnitine palmitoyltransferase (CPT) proteins, as gatekeepers of fatty acid oxidation, have great potential as pharmacological targets to treat metabolic diseases like obesity, diabetes, and cancer. In recent years, significant progress has been made in understanding the 3-dimensional structure of CPTs and their pathophysiological functions. A deeper understanding of the differences between the various CPT family members will enable the design of selective drugs and therapeutic approaches with fewer side effects.
Collapse
Affiliation(s)
- Rosalía Rodríguez-Rodríguez
- Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Universitat Internacional de Catalunya (UIC), Sant Cugat del Vallès, Spain; Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y la Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain.
| | - Miguel Baena
- Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Universitat Internacional de Catalunya (UIC), Sant Cugat del Vallès, Spain
| | - Sebastián Zagmutt
- Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Universitat Internacional de Catalunya (UIC), Sant Cugat del Vallès, Spain
| | - West Kristian Paraiso
- Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Universitat Internacional de Catalunya (UIC), Sant Cugat del Vallès, Spain
| | - Ana Cristina Reguera
- Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Universitat Internacional de Catalunya (UIC), Sant Cugat del Vallès, Spain
| | - Rut Fadó
- Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Universitat Internacional de Catalunya (UIC), Sant Cugat del Vallès, Spain
| | - Núria Casals
- Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Universitat Internacional de Catalunya (UIC), Sant Cugat del Vallès, Spain; Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y la Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain.
| |
Collapse
|
10
|
Han J, Jeong H, Choi J, Kim H, Kwon T, Myung K, Park K, Park JI, Sánchez S, Jung D, Yu CS, Song IH, Shim J, Myung S, Kang H, Park T. Bioprinted Patient-Derived Organoid Arrays Capture Intrinsic and Extrinsic Tumor Features for Advanced Personalized Medicine. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2407871. [PMID: 40151904 PMCID: PMC12120747 DOI: 10.1002/advs.202407871] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/11/2024] [Revised: 02/11/2025] [Indexed: 03/29/2025]
Abstract
Heterogeneity and the absence of a tumor microenvironment (TME) in traditional patient-derived organoid (PDO) cultures limit their effectiveness for clinical use. Here, Embedded Bioprinting-enabled Arrayed PDOs (Eba-PDOs) featuring uniformly arrayed colorectal cancer (CRC) PDOs within a recreated TME is presented. This model faithfully reproduces critical TME attributes, including elevated matrix stiffness (≈7.5 kPa) and hypoxic conditions found in CRC. Transcriptomic and immunofluorescence microscopy analysis reveal that Eba-PDOs more accurately represent actual tissues compared to traditional PDOs. Furthermore, Eba-PDO effectively capture the variability of CEACAM5 expression-a critical CRC marker-across different patients, correlating with patient classification and differential responses to 5-fluorouracil treatment. This method achieves an uniform size and shape within PDOs from the same patient while preserving distinct morphological features among those from different individuals. These features of Eba-PDO enable the efficient development of a label-free, morphology-based predictive model using supervised learning, enhancing its suitability for clinical applications. Thus, this approach to PDO bioprinting is a promising tool for generating personalized tumor models and advancing precision medicine.
Collapse
Affiliation(s)
- Jonghyeuk Han
- Department of Biomedical EngineeringUlsan National Institute of Science and Technology (UNIST)Ulsan44919Republic of Korea
- Wallace H. Coulter Department of Biomedical EngineeringEmory University School of Medicine & Georgia Institute of TechnologyAtlantaGA30332USA
| | - Hye‐Jin Jeong
- Department of Biomedical EngineeringUlsan National Institute of Science and Technology (UNIST)Ulsan44919Republic of Korea
- Center for Genome EngineeringInstitute for Basic ScienceDaejeon34126Republic of Korea
| | - Jeonghan Choi
- Department of Biomedical EngineeringUlsan National Institute of Science and Technology (UNIST)Ulsan44919Republic of Korea
| | - Hyeonseo Kim
- Department of Biomedical EngineeringUlsan National Institute of Science and Technology (UNIST)Ulsan44919Republic of Korea
| | - Taejoon Kwon
- Department of Biomedical EngineeringUlsan National Institute of Science and Technology (UNIST)Ulsan44919Republic of Korea
- Center for Genomic IntegrityInstitute for Basic ScienceUlsan44919Republic of Korea
| | - Kyungjae Myung
- Department of Biomedical EngineeringUlsan National Institute of Science and Technology (UNIST)Ulsan44919Republic of Korea
- Center for Genomic IntegrityInstitute for Basic ScienceUlsan44919Republic of Korea
| | - Kyemyung Park
- Graduate School of Health Science and Technology and Department of Biomedical EngineeringUlsan National Institute of Science and TechnologyUlsan44919Republic of Korea
| | - Jung In Park
- Department of Biomedical EngineeringUlsan National Institute of Science and Technology (UNIST)Ulsan44919Republic of Korea
| | - Samuel Sánchez
- Institute for Bioengineering of Catalonia (IBEC)The Barcelona Institute for Science and Technology (BIST)Barcelona08028Spain
- Catalan Institute for Research and Advanced Studies (ICREA)Barcelona08010Spain
| | - Deok‐Beom Jung
- Digestive Diseases Research CenterUniversity of Ulsan College of MedicineSeoul05505Republic of Korea
| | - Chang Sik Yu
- Division of Colon and Rectal SurgeryDepartment of SurgeryAsan Medical CenterUniversity of Ulsan College of MedicineSeoul05505Republic of Korea
| | - In Ho Song
- Division of Colon and Rectal SurgeryDepartment of SurgeryAsan Medical CenterUniversity of Ulsan College of MedicineSeoul05505Republic of Korea
| | - Jin‐Hyung Shim
- Research InstituteT&R Biofab Co. Ltd.Siheung15111Republic of Korea
- Department of Mechanical EngineeringTech University of KoreaSiheung15073Republic of Korea
| | - Seung‐Jae Myung
- Digestive Diseases Research CenterUniversity of Ulsan College of MedicineSeoul05505Republic of Korea
- Department of GastroenterologyAsan Medical CenterUniversity of Ulsan College of MedicineSeoul05505Republic of Korea
- EDIS BiotechSeoul05505Republic of Korea
| | - Hyun‐Wook Kang
- Department of Biomedical EngineeringUlsan National Institute of Science and Technology (UNIST)Ulsan44919Republic of Korea
| | - Tae‐Eun Park
- Department of Biomedical EngineeringUlsan National Institute of Science and Technology (UNIST)Ulsan44919Republic of Korea
| |
Collapse
|
11
|
Wang D, Suzuki A, Tong W. The connection between Bayesian networks and adverse outcome pathway (AOP) networks and how to use it for predicting drug toxicity. Drug Discov Today 2025; 30:104350. [PMID: 40187482 DOI: 10.1016/j.drudis.2025.104350] [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: 07/16/2024] [Revised: 03/20/2025] [Accepted: 03/31/2025] [Indexed: 04/07/2025]
Abstract
There is significant interest in combining adverse outcome pathways (AOPs) with Bayesian networks (BNs) because of their shared representation using directed acyclic graphs (DAGs). However, it has not been verified empirically whether AOP networks are mathematically congruent with BNs. Furthermore, important properties for BNs, such as Markov blankets, have not been emphasized, which is a missed opportunity for simplifying and optimizing the model. Here, we summarize the connection between AOP networks and BNs and explore the implications for toxicity modeling. We also present a case study in drug-related liver toxicity. Our results confirm that AOP networks are congruent mathematically with BNs, with incorporation of the mathematical properties of BN leading to significantly simplified and more efficient models.
Collapse
Affiliation(s)
- Dong Wang
- Division of Bioinformatics and Biostatistics, National Center for Toxicological Research, U.S. Food and Drug Administration, Jefferson, AR, USA.
| | - Ayako Suzuki
- Division of Gastroenterology, Duke University, Durham, NC, USA; Department of Medicine, Durham VA Medical Center, Durham, NC, USA
| | - Weida Tong
- Division of Bioinformatics and Biostatistics, National Center for Toxicological Research, U.S. Food and Drug Administration, Jefferson, AR, USA
| |
Collapse
|
12
|
Ge X, Wekselblatt JB, Elmore S, Wang B, Wang T, Dai R, Zhang T, Dave H, Ghaderi M, Anilkumar AR, Wang B, Sirsi SR, Ahn JM, Shapiro MG, Oka Y, Lois C, Qin Z. In Vivo Cytosolic Delivery of Biomolecules into Neurons for Super-Resolution Imaging and Genome Modification. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025:e2501033. [PMID: 40285608 DOI: 10.1002/advs.202501033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/16/2025] [Revised: 03/27/2025] [Indexed: 04/29/2025]
Abstract
Efficient delivery of biomolecules into neurons has significant impacts on therapeutic applications in the central nervous system (CNS) and fundamental neuroscience research. Existing viral and non-viral delivery methods often suffer from inefficient intracellular access due to the endocytic pathway. Here, a neuron-targeting and direct cytosolic delivery platform is discovered by using a 15-amino-acid peptide, termed the N1 peptide, which enables neuron-specific targeting and cytosolic delivery of functional biomolecules. The N1 peptide initially binds hyaluronan in the extracellular matrix and subsequently passes the membrane of neurons without being trapped into endosome. This mechanism facilitates the efficient delivery of cell-impermeable and photo-stable fluorescent dye for super-resolution imaging of dendritic spines, and functional proteins, such as Cre recombinase, for site-specific genome modification. Importantly, the N1 peptide exhibits robust neuronal specificity across diverse species, including mice, rats, tree shrews, and zebra finches. Its targeting capability is further demonstrated through various administration routes, including intraparenchymal, intrathecal, and intravenous (i.v.) injections after blood-brain barrier (BBB) opening with focused ultrasound (FUS). These findings establish the N1 peptide as a versatile and functional platform with significant potential for bioimaging and advanced therapeutic applications.
Collapse
Affiliation(s)
- Xiaoqian Ge
- Department of Biomedical Engineering, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
- Department of Mechanical Engineering, University of Texas at Dallas, Richardson, TX, 75080-3021, USA
| | - Joseph B Wekselblatt
- Division of Chemistry and Chemical Engineering, California Institute of Technology, Pasadena, CA, 91125, USA
- Department of Opthalmology, David Geffen School of Medicine, University of California, Los Angeles, CA, 90095, USA
| | - Scott Elmore
- Department of Chemistry and Biochemistry, University of Texas at Dallas, Richardson, TX, 75080-3021, USA
| | - Bo Wang
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, 91125, USA
| | - Tongtong Wang
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, 91125, USA
| | - Renjinming Dai
- Department of Bioengineering, University of Texas at Dallas, Richardson, TX, 75080-3021, USA
| | - Tingting Zhang
- Department of Mechanical Engineering, University of Texas at Dallas, Richardson, TX, 75080-3021, USA
| | - Harsh Dave
- Department of Bioengineering, University of Texas at Dallas, Richardson, TX, 75080-3021, USA
| | - Mohammadaref Ghaderi
- Department of Bioengineering, University of Texas at Dallas, Richardson, TX, 75080-3021, USA
| | - Athul Raj Anilkumar
- Department of Bioengineering, University of Texas at Dallas, Richardson, TX, 75080-3021, USA
| | - Bill Wang
- Department of Bioengineering, University of Texas at Dallas, Richardson, TX, 75080-3021, USA
| | - Shashank R Sirsi
- Department of Bioengineering, University of Texas at Dallas, Richardson, TX, 75080-3021, USA
| | - Jung-Mo Ahn
- Department of Chemistry and Biochemistry, University of Texas at Dallas, Richardson, TX, 75080-3021, USA
| | - Mikhail G Shapiro
- Division of Chemistry and Chemical Engineering, California Institute of Technology, Pasadena, CA, 91125, USA
- Andrew and Peggy Cherng Department of Medical Engineering, California Institute of Technology, Pasadena, CA, 91125, USA
- Howard Hughes Medical Institute, Pasadena, CA, 91125, USA
| | - Yuki Oka
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, 91125, USA
| | - Carlos Lois
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, 91125, USA
| | - Zhenpeng Qin
- Department of Biomedical Engineering, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
- Department of Mechanical Engineering, University of Texas at Dallas, Richardson, TX, 75080-3021, USA
- Department of Bioengineering, University of Texas at Dallas, Richardson, TX, 75080-3021, USA
- Center for Advanced Pain Studies, University of Texas at Dallas, Richardson, TX, 75080-3021, USA
| |
Collapse
|
13
|
Gookin TE, Chakravorty D, Assmann SM. Influence of expression and purification protocols on Gα biochemical activity: kinetics of plant and mammalian G protein cycles. Front Mol Biosci 2025; 12:1513660. [PMID: 40260404 PMCID: PMC12009698 DOI: 10.3389/fmolb.2025.1513660] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2024] [Accepted: 03/21/2025] [Indexed: 04/23/2025] Open
Abstract
Heterotrimeric G proteins, composed of Gα, Gβ, and Gγ subunits, are a class of signal transduction complexes with broad roles in human health and agriculturally relevant plant physiological and developmental traits. In the classic paradigm, guanine nucleotide binding to the Gα subunit regulates the activation status of the complex. We sought to develop improved methods for heterologous expression and rapid purification of Gα subunits, initially targeting GPA1, the sole canonical Gα subunit of the model plant species, Arabidopsis thaliana. Compared to conventional methods, our expression methodology and rapid StrepII-tag mediated purification facilitates substantially higher yield, and isolation of protein with increased GTP binding and hydrolysis activities. Human GNAI1 purified using our approach displayed the expected binding and hydrolysis activities, indicating our protocol is applicable to mammalian Gα subunits, potentially including those for which purification of enzymatically active protein has been historically problematic. We subsequently utilized domain swaps of GPA1 and human GNAO1 to demonstrate that the inherent instability of GPA1 is a function of the interaction between the Ras and helical domains. Additionally, we found that GPA1-GNAO1 domain swaps partially uncouple the instability from the rapid nucleotide binding kinetics displayed by GPA1. In summary, our work provides insights into methods to optimally study heterotrimeric G proteins, and reveals roles of the helical domain in Gα kinetics and stability.
Collapse
|
14
|
Zheng R, Zhang S, Chen S, Zha W, Li X, Li Q, He J, He S, Feng M, Shen Y. Sunlight-mediated environmental risks of tinidazole in seawater: A neglected ocular toxicity of photolysis mixtures. JOURNAL OF HAZARDOUS MATERIALS 2025; 487:137217. [PMID: 39823881 DOI: 10.1016/j.jhazmat.2025.137217] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Revised: 12/26/2024] [Accepted: 01/12/2025] [Indexed: 01/20/2025]
Abstract
Tinidazole (TNZ), a common nitroimidazole antibiotic, is pervasive in aquatic ecosystems, posing potential threats to marine organisms. The environmental fate of TNZ, particularly under solar irradiation, and the associated secondary risks are not well characterized. Herein, the photochemical reactivity of TNZ and four other typical nitroimidazoles (i.e., metronidazole, ornidazole, dimetridazole, and secnidazole) were quantified for multiple photoreactive species. The photolysis products of these nitroimidazoles were identified under solar irradiation, from which the reaction pathways were tentatively proposed. Furthermore, the photo-induced toxicity evolution mechanisms of TNZ were investigated by comparing phenotypic, transcriptomic, and metabolomic changes in marine medaka embryos (Oryzias melastigma) after exposure to TNZ and its photo-irradiated mixtures. Our results indicated that the photo-irradiated TNZ enhanced visual toxicity to marine medaka embryos compared to the parent compound. The photolysis mixtures induced embryonic ocular malformation and significantly affected the expression of the associated genes with the initiation/termination of the phototransduction cascade, leading to metabolite changes related to visual impairment. This work reported the first comprehensive assessment of the photolysis-mediated environmental fate and secondary risks of TNZ in seawater. The findings highlighted the necessity of including complex photolysis mixtures under solar irradiation in future chemical risk assessments of marine environments.
Collapse
Affiliation(s)
- Ruping Zheng
- Key Laboratory of the Ministry of Education for Coastal and Wetland Ecosystems, College of the Environment and Ecology, Xiamen University, Xiamen 361102, China
| | - Shengqi Zhang
- Key Laboratory of the Ministry of Education for Coastal and Wetland Ecosystems, College of the Environment and Ecology, Xiamen University, Xiamen 361102, China
| | - Shengyue Chen
- Key Laboratory of the Ministry of Education for Coastal and Wetland Ecosystems, College of the Environment and Ecology, Xiamen University, Xiamen 361102, China
| | - Wenqi Zha
- Key Laboratory of the Ministry of Education for Coastal and Wetland Ecosystems, College of the Environment and Ecology, Xiamen University, Xiamen 361102, China
| | - Xinyue Li
- Key Laboratory of the Ministry of Education for Coastal and Wetland Ecosystems, College of the Environment and Ecology, Xiamen University, Xiamen 361102, China
| | - Qiuru Li
- Key Laboratory of the Ministry of Education for Coastal and Wetland Ecosystems, College of the Environment and Ecology, Xiamen University, Xiamen 361102, China
| | - Jinlin He
- Key Laboratory of the Ministry of Education for Coastal and Wetland Ecosystems, College of the Environment and Ecology, Xiamen University, Xiamen 361102, China
| | - Shanshan He
- Key Laboratory of the Ministry of Education for Coastal and Wetland Ecosystems, College of the Environment and Ecology, Xiamen University, Xiamen 361102, China
| | - Mingbao Feng
- Key Laboratory of the Ministry of Education for Coastal and Wetland Ecosystems, College of the Environment and Ecology, Xiamen University, Xiamen 361102, China.
| | - Yingjia Shen
- Key Laboratory of the Ministry of Education for Coastal and Wetland Ecosystems, College of the Environment and Ecology, Xiamen University, Xiamen 361102, China; State Key Laboratory of Mariculture Breeding, Xiamen University, Xiamen 361102, China.
| |
Collapse
|
15
|
Li K, Dai YJ, Zhang H, Zhang Z. YAP1 activates SLC2A1 transcription and augments the malignant behavior of colorectal cancer cells by activating the Wnt/β-catenin signaling pathway. Cell Div 2025; 20:8. [PMID: 40186232 PMCID: PMC11969700 DOI: 10.1186/s13008-025-00148-y] [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: 01/05/2025] [Accepted: 03/18/2025] [Indexed: 04/07/2025] Open
Abstract
OBJECTIVE This paper examined the role of solute carrier family 2 member 1 (SLC2A1) in colorectal cancer (CRC) progression, focusing on its expression levels, functional implications, and regulatory mechanisms involving Yes-associated protein 1 (YAP1) and the Wnt signaling pathway. METHODS GEO datasets (GSE14297, GSE18462, GSE40367) were analyzed to identify genes linked to metastasis in CRC, and TCGA-COAD system was used to analyze the expression pattern and prognostic values of SLC2A1 in CRC. Functional studies were conducted using CRC cell lines (Caco-2 and SW480). Cell viability, migration and invasion, and apoptosis were examined using EdU assays, Transwell assays, and flow cytometry. YAP1's regulatory role on SLC2A1 was investigated using ChIP-qPCR and luciferase reporter assays. The Wnt/β-catenin agonist SKL2001 was used for functional rescue experiments. RESULTS SLC2A1 was upregulated in CRC cells, and its upregulation was associated with tumor metastasis and unfavorable outcomes according to bioinformatics. Knockdown of SLC2A1 resulted in reduced cell viability, decreased migration, and increased apoptosis in Caco-2 and SW480 cells. Additionally, YAP1 was identified as a transcriptional activator of SLC2A1. Knockdown of YAP1 decreased SLC2A1 expression and reduced expression of Wnt target genes, thus suppressing malignant behavior of tumor cells. However, further overexpression of SLC2A1 restored cell viability and migration in YAP1-deficient cells. The YAP1- SLC2A1 axis activated the Wnt/β-catenin by reducing GSK3β activity. CONCLUSION SLC2A1 is critical in CRC progression, with YAP1 serving as a key regulator of its expression and function. The YAP1-SLC2A1-Wnt axis represents a potential therapeutic target for CRC, providing insights into metabolic adaptations that support tumor growth and metastasis.
Collapse
Affiliation(s)
- Kunpeng Li
- Zhongda Hospital of Southeast University, No 87 Dingjiaqiao, Nanjing, 210009, Jiangsu, PR China
| | - Ya-Jie Dai
- Department of General Surgery, Zhongda Hospital, Southeast University, Nanjing, 210009, Jiangsu, PR China
| | - Haifeng Zhang
- Department of General Surgery, Zhongda Hospital, Southeast University, Nanjing, 210009, Jiangsu, PR China
| | - Zhigang Zhang
- Department of General Surgery, Zhongda Hospital, Southeast University, Nanjing, 210009, Jiangsu, PR China.
| |
Collapse
|
16
|
Bhoumik S, Lee Y. Core Molecular Clock Factors Regulate Osteosarcoma Stem Cell Survival and Behavior via CSC/EMT Pathways and Lipid Droplet Biogenesis. Cells 2025; 14:517. [PMID: 40214471 PMCID: PMC11988071 DOI: 10.3390/cells14070517] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2025] [Revised: 03/26/2025] [Accepted: 03/27/2025] [Indexed: 04/14/2025] Open
Abstract
The circadian clock, an intrinsic 24 h cellular timekeeping system, regulates fundamental biological processes, including tumor physiology and metabolism. Cancer stem cells (CSCs), a subpopulation of cancer cells with self-renewal and tumorigenic capacities, are implicated in tumor initiation, recurrence, and metastasis. Despite growing evidence for the circadian clock's involvement in regulating CSC functions, its precise regulatory mechanisms remain largely unknown. Here, using a human osteosarcoma (OS) model (143B), we have shown that core molecular clock factors are critical for OS stem cell survival and behavior via direct modulation of CSC and lipid metabolic pathways. In single-cell-derived spheroid formation assays, 143B OS cells exhibited robust spheroid-forming capacity under 3D culture conditions. Furthermore, siRNA-mediated depletion of core clock components (i.e., BMAL1, CLOCK, CRY1/2, PER1/2)-essential positive and negative elements of the circadian clock feedback loop-significantly reduced spheroid formation in 143B CSCs isolated from in vivo OS xenografts. In contrast, knockdown of the secondary clock-stabilizing factor genes NR1D1 and NR1D2 had little effect. We also found that knockdown of BMAL1, CLOCK, or CRY1/2 markedly impaired the migration and invasion capacities of 143B CSCs. At the molecular level, silencing of BMAL1, CLOCK, or CRY1/2 distinctly altered the expression of genes associated with stem cell properties and the epithelial-mesenchymal transition (EMT) in 143B CSCs. In addition, disruption of BMAL1, CLOCK, or CRY1/2 expression significantly reduced lipid droplet formation by downregulating the expression of genes involved in lipogenesis (e.g., DGAT1, FASN, ACSL4, PKM2, CHKA, SREBP1), which are closely linked to CSC/EMT processes. Furthermore, transcriptomic analysis of human OS patient samples revealed that compared with other core clock genes, CRY1 was highly expressed in OS tumors relative to controls, and its expression exhibited strong positive correlations with patient prognosis, survival, and LD biogenesis gene expression. These findings highlight the critical role of the molecular circadian clock in regulating CSC properties and metabolism, underscoring the therapeutic potential of targeting the core clock machinery to enhance OS treatment outcomes.
Collapse
Affiliation(s)
- Sukanya Bhoumik
- Department of Translational Medicine and Physiology, Elson S. Floyd College of Medicine, Washington State University, Spokane, WA 99202, USA;
| | - Yool Lee
- Department of Translational Medicine and Physiology, Elson S. Floyd College of Medicine, Washington State University, Spokane, WA 99202, USA;
- Department of Integrative Physiology and Neuroscience, College of Veterinary Medicine, Washington State University, Pullman, WA 99164, USA
- Sleep and Performance Research Center, Washington State University, Spokane, WA 99202, USA
- Steve Gleason Institute for Neuroscience, Washington State University, Spokane, WA 99202, USA
| |
Collapse
|
17
|
Serra A, Zouraris D, Schaffert A, Torres Maia M, Tsiros P, Virmani I, Di Lieto E, Saarimäki LA, Morikka J, Riudavets-Puig R, Varsou DD, Papavasileiou KD, Kolokathis PD, Mintis DG, Tzoupis H, Tsoumanis A, Melagraki G, Arvanitidis A, Doganis P, Minadakis V, Savvas G, Perello-y-bestard A, Cucurachi S, Buljan M, Nikiforou F, Karakoltzidis A, Karakitsios S, Sarigiannis DA, Friedrichs S, Seitz C, Gutierrez TN, Isigonis P, Cambier S, Marvuglia A, Lindner GG, Sergent JA, Gheorghe LC, Bradford LJA, Park SG, Ha SM, Gerelkhuu Z, Yoon TH, Petry R, Martinez DST, Winkler DA, Wick P, Exner TE, Dondero F, Serchi T, Peijnenburg W, Sarimveis H, Paparella M, Lynch I, Afantitis A, Greco D. INSIGHT: An integrated framework for safe and sustainable chemical and material assessment. Comput Struct Biotechnol J 2025; 29:125-137. [PMID: 40241814 PMCID: PMC12002752 DOI: 10.1016/j.csbj.2025.03.042] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2025] [Revised: 03/25/2025] [Accepted: 03/25/2025] [Indexed: 04/18/2025] Open
Abstract
The assessment of chemicals and materials has traditionally been fragmented, with health, environmental, social, and economic impacts evaluated independently. This disjointed approach limits the ability to capture trade-offs and synergies necessary for comprehensive decision-making under the Safe and Sustainable by Design (SSbD) framework. The EU INSIGHT project addresses this challenge by developing a novel computational framework for integrated impact assessment, based on the Impact Outcome Pathway (IOP) approach. Extending the Adverse Outcome Pathway (AOP) concept, IOPs establish mechanistic links between chemical and material properties and their environmental, health, and socio-economic consequences. The project integrates multi-source datasets (including omics, life cycle inventories, and exposure models) into a structured knowledge graph (KG), ensuring FAIR (Findable, Accessible, Interoperable, Reusable) data principles are met. INSIGHT is being developed and validated through four case studies targeting per- and polyfluoroalkyl substances (PFAS), graphene oxide (GO), bio-based synthetic amorphous silica (SAS), and antimicrobial coatings. These studies demonstrate how multi-model simulations, decision-support tools, and artificial intelligence-driven knowledge extraction can enhance the predictability and interpretability of chemical and material impacts. Additionally, INSIGHT incorporates interactive, web-based decision maps to provide stakeholders with accessible, regulatory-compliant risk and sustainability assessments. By bridging mechanistic toxicology, exposure modeling, life cycle assessment, and socio-economic analysis, INSIGHT advances a scalable, transparent, and data-driven approach to SSbD. This project aligns with the European Green Deal and global sustainability goals, promoting safer, more sustainable innovation in chemicals and materials through an integrated, mechanistic, and computationally advanced framework.
Collapse
Affiliation(s)
- Angela Serra
- Finnish Hub for Development and Validation of Integrated Approaches (FHAIVE), Faculty of Medicine and Health Technology, Tampere University, Tampere 33100, Finland
- Division of Pharmaceutical Biosciences, Faculty of Pharmacy, University of Helsinki, Helsinki 00790, Finland
| | - Dimitrios Zouraris
- NovaMechanics Ltd, Nicosia 1070, Cyprus
- Entelos Institute, Larnaca 6059, Cyprus
| | - Alexandra Schaffert
- Finnish Hub for Development and Validation of Integrated Approaches (FHAIVE), Faculty of Medicine and Health Technology, Tampere University, Tampere 33100, Finland
| | - Marcella Torres Maia
- Finnish Hub for Development and Validation of Integrated Approaches (FHAIVE), Faculty of Medicine and Health Technology, Tampere University, Tampere 33100, Finland
| | - Periklis Tsiros
- School of Chemical Engineering, National Technical University of Athens, Attiki 15772, Greece
| | - Ishita Virmani
- Medical University Innsbruck, Institute for Medical Biochemistry, Innsbruck 6020, Austria
| | - Emanuele Di Lieto
- Finnish Hub for Development and Validation of Integrated Approaches (FHAIVE), Faculty of Medicine and Health Technology, Tampere University, Tampere 33100, Finland
| | - Laura Aliisa Saarimäki
- Finnish Hub for Development and Validation of Integrated Approaches (FHAIVE), Faculty of Medicine and Health Technology, Tampere University, Tampere 33100, Finland
| | - Jack Morikka
- Finnish Hub for Development and Validation of Integrated Approaches (FHAIVE), Faculty of Medicine and Health Technology, Tampere University, Tampere 33100, Finland
| | - Rafael Riudavets-Puig
- EMPA Materials Science and Technology, Lerchenfeldstrasse 5, St. Gallen CH-9014, Switzerland
| | - Dimitra-Danai Varsou
- Entelos Institute, Larnaca 6059, Cyprus
- NovaMechanics MIKE, Piraeus 18545, Greece
| | | | | | - Dimitris G. Mintis
- Entelos Institute, Larnaca 6059, Cyprus
- NovaMechanics MIKE, Piraeus 18545, Greece
| | - Haralampos Tzoupis
- NovaMechanics Ltd, Nicosia 1070, Cyprus
- Entelos Institute, Larnaca 6059, Cyprus
| | - Andreas Tsoumanis
- NovaMechanics Ltd, Nicosia 1070, Cyprus
- NovaMechanics MIKE, Piraeus 18545, Greece
| | - Georgia Melagraki
- Division of Physical Sciences and Applications, Hellenic Military Academy, Vari 16672, Greece
| | - Alex Arvanitidis
- School of Chemical Engineering, National Technical University of Athens, Attiki 15772, Greece
| | - Philip Doganis
- School of Chemical Engineering, National Technical University of Athens, Attiki 15772, Greece
| | - Vasileios Minadakis
- School of Chemical Engineering, National Technical University of Athens, Attiki 15772, Greece
| | - Giannis Savvas
- School of Chemical Engineering, National Technical University of Athens, Attiki 15772, Greece
| | - Adrien Perello-y-bestard
- Institute of Environmental Sciences, Leiden University, P.O. Box 9518, Leiden 2300 RA, Netherlands
| | - Stefano Cucurachi
- Institute of Environmental Sciences, Leiden University, P.O. Box 9518, Leiden 2300 RA, Netherlands
| | - Marija Buljan
- EMPA Materials Science and Technology, Lerchenfeldstrasse 5, St. Gallen CH-9014, Switzerland
| | - Fotini Nikiforou
- Aristotle University of Thessaloniki, Department of Chemical Engineering, Environmental Engineering Laboratory, University Campus, Thessaloniki 54124, Greece
- HERACLES Research Center on the Exposome and Health, Center for Interdisciplinary Research and Innovation, Balkan Center, Bldg. B, 10th km Thessaloniki – Thermi Road, 57001, Greece
| | - Achilleas Karakoltzidis
- Aristotle University of Thessaloniki, Department of Chemical Engineering, Environmental Engineering Laboratory, University Campus, Thessaloniki 54124, Greece
- HERACLES Research Center on the Exposome and Health, Center for Interdisciplinary Research and Innovation, Balkan Center, Bldg. B, 10th km Thessaloniki – Thermi Road, 57001, Greece
| | - Spyros Karakitsios
- Aristotle University of Thessaloniki, Department of Chemical Engineering, Environmental Engineering Laboratory, University Campus, Thessaloniki 54124, Greece
- HERACLES Research Center on the Exposome and Health, Center for Interdisciplinary Research and Innovation, Balkan Center, Bldg. B, 10th km Thessaloniki – Thermi Road, 57001, Greece
| | - Dimosthenis A. Sarigiannis
- Aristotle University of Thessaloniki, Department of Chemical Engineering, Environmental Engineering Laboratory, University Campus, Thessaloniki 54124, Greece
- HERACLES Research Center on the Exposome and Health, Center for Interdisciplinary Research and Innovation, Balkan Center, Bldg. B, 10th km Thessaloniki – Thermi Road, 57001, Greece
| | | | | | | | | | | | | | - Gottlieb Georg Lindner
- Evonik Operations GmbH, Research, Development & Innovation, Bruehler Strasse 2, Wesseling 50389, Germany
| | - Jacques-Aurélien Sergent
- Solvay SA, Toxicological and Environmental Risk Assessment Unit, Rue de Ransbeek 310, Bruxelles 1120, Belgium
| | - L. Cristiana Gheorghe
- School of Geography, Earth and Environmental Sciences, University of Birmingham, Birmingham B15 2TT, United Kingdom
- Centre for Environmental Research and Justice, University of Birmingham, Edgbaston, Birmingham B15 2TT, United Kingdom
| | - Laura-Jayne A. Bradford
- School of Geography, Earth and Environmental Sciences, University of Birmingham, Birmingham B15 2TT, United Kingdom
- Centre for Environmental Research and Justice, University of Birmingham, Edgbaston, Birmingham B15 2TT, United Kingdom
| | - Seung-Geun Park
- Department of Chemistry, College of Natural Sciences, Hanyang University, Seoul 04763, South Korea
| | - Seung Min Ha
- Department of Chemistry, College of Natural Sciences, Hanyang University, Seoul 04763, South Korea
| | - Zayakhuu Gerelkhuu
- Institute of Next Generation Material Design, Hanyang University, Seoul 04763, South Korea
| | - Tae Hyun Yoon
- Department of Chemistry, College of Natural Sciences, Hanyang University, Seoul 04763, South Korea
- Institute of Next Generation Material Design, Hanyang University, Seoul 04763, South Korea
| | - Romana Petry
- Brazilian Nanotechnology National Laboratory (LNNano), Brazilian Center for Research in Energy and Materials (CNPEM), Campinas, Sao Paulo, Brazil
| | - Diego Stéfani Teodoro Martinez
- Brazilian Nanotechnology National Laboratory (LNNano), Brazilian Center for Research in Energy and Materials (CNPEM), Campinas, Sao Paulo, Brazil
| | - David A. Winkler
- Department of Biochemistry and Chemistry, La Trobe Institute for Molecular Science, La Trobe University, Bundoora, Victoria 3086, Australia
- Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria 3052, Australia
- School of Pharmacy, University of Nottingham, Nottingham NG7 2RD, United Kingdom
| | - Peter Wick
- EMPA Materials Science and Technology, Lerchenfeldstrasse 5, St. Gallen CH-9014, Switzerland
| | - Thomas E. Exner
- Seven Past Nine d.o.o., Hribljane 10, Cerknica 1380, Slovenia
| | - Francesco Dondero
- Department of Science and Technological Innovation, University of Eastern Piedmont, Alessandria 15121, Italy
| | - Tommaso Serchi
- Luxembourg Institute of Science and Technology (LIST), Luxembourg
| | - Willie Peijnenburg
- Institute of Environmental Sciences, Leiden University, P.O. Box 9518, Leiden 2300 RA, Netherlands
- National Institute for Public Health and the Environment (RIVM), Center for Safety Assessment of Substances and Products, Bilthoven, Netherlands
| | - Haralambos Sarimveis
- School of Chemical Engineering, National Technical University of Athens, Attiki 15772, Greece
| | - Martin Paparella
- Medical University Innsbruck, Institute for Medical Biochemistry, Innsbruck 6020, Austria
| | - Iseult Lynch
- School of Geography, Earth and Environmental Sciences, University of Birmingham, Birmingham B15 2TT, United Kingdom
- Centre for Environmental Research and Justice, University of Birmingham, Edgbaston, Birmingham B15 2TT, United Kingdom
| | - Antreas Afantitis
- NovaMechanics Ltd, Nicosia 1070, Cyprus
- Entelos Institute, Larnaca 6059, Cyprus
- NovaMechanics MIKE, Piraeus 18545, Greece
| | - Dario Greco
- Finnish Hub for Development and Validation of Integrated Approaches (FHAIVE), Faculty of Medicine and Health Technology, Tampere University, Tampere 33100, Finland
- Division of Pharmaceutical Biosciences, Faculty of Pharmacy, University of Helsinki, Helsinki 00790, Finland
| |
Collapse
|
18
|
Eduardo MB, Cottone G, McCloskey CW, Liu S, Palma FR, Zappia MP, Islam AB, Gao P, Setya J, Dennis S, Gao H, Zhang Q, Xuei X, Luo Y, Locasale J, Bonini MG, Khokha R, Frolov MV, Benevolenskaya EV, Chandel NS, Khan SA, Clare SE. A metabolic shift to the serine pathway induced by lipids fosters epigenetic reprogramming in nontransformed breast cells. SCIENCE ADVANCES 2025; 11:eads9182. [PMID: 40117373 PMCID: PMC11927636 DOI: 10.1126/sciadv.ads9182] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Accepted: 02/18/2025] [Indexed: 03/23/2025]
Abstract
Lipid metabolism and the serine, one-carbon, glycine (SOG) and methionine pathways are independently and significantly correlated with estrogen receptor-negative breast cancer (ERneg BC). Here, we propose a link between lipid metabolism and ERneg BC through phosphoglycerate dehydrogenase (PHGDH), the rate-limiting enzyme in the de novo serine pathway. We demonstrate that the metabolism of the paradigmatic medium-chain fatty acid octanoic acid leads to a metabolic shift toward the SOG and methionine pathways. PHGDH plays a role in both the forward direction, contributing to the production of S-adenosylmethionine, and the reverse direction, generating the oncometabolite 2-hydroxyglutarate, leading to epigenomic reprogramming and phenotypic plasticity. The methionine cycle is closely linked to the transsulfuration pathway. Consequently, we observe that the shift increases the antioxidant glutathione, which mitigates reactive oxygen species (ROS), enabling survival of a subset of cells that have undergone DNA damage. These metabolic changes contribute to several hallmarks of cancer.
Collapse
Affiliation(s)
| | - Gannon Cottone
- Department of Surgery, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Curtis W. McCloskey
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| | - Shiyu Liu
- Department of Pharmacology and Cancer Biology, Duke University, Durham, NC, USA
| | - Flavio R. Palma
- Department of Medicine/Division of Hematology Oncology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Maria Paula Zappia
- Department of Biochemistry and Molecular Genetics, University of Illinois at Chicago, Chicago, IL, USA
| | - Abul B.M.M.K. Islam
- Department of Biochemistry and Molecular Genetics, University of Illinois at Chicago, Chicago, IL, USA
| | - Peng Gao
- Robert H. Lurie Cancer Center Metabolomics Core, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Joel Setya
- Department of Surgery, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Saya Dennis
- Department of Preventive Medicine, Northwestern University, Chicago, IL, USA
| | - Hongyu Gao
- Center for Medical Genomics, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Qian Zhang
- Robert H. Lurie Cancer Center Metabolomics Core, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Xiaoling Xuei
- Center for Medical Genomics, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Yuan Luo
- Department of Preventive Medicine, Northwestern University, Chicago, IL, USA
- Robert H. Lurie Cancer Center of Northwestern University, Chicago, IL, USA
| | - Jason Locasale
- Department of Pharmacology and Cancer Biology, Duke University, Durham, NC, USA
| | - Marcelo G. Bonini
- Department of Medicine/Division of Hematology Oncology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
- Robert H. Lurie Cancer Center of Northwestern University, Chicago, IL, USA
| | - Rama Khokha
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| | - Maxim V. Frolov
- Department of Medicine/Division of Hematology Oncology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
- Department of Biochemistry and Molecular Genetics, University of Illinois at Chicago, Chicago, IL, USA
| | - Elizaveta V. Benevolenskaya
- Department of Medicine/Division of Hematology Oncology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
- Department of Biochemistry and Molecular Genetics, University of Illinois at Chicago, Chicago, IL, USA
| | - Navdeep S. Chandel
- Robert H. Lurie Cancer Center of Northwestern University, Chicago, IL, USA
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Northwestern University, Chicago, IL, USA
- Department of Biochemistry and Molecular Genetics, Northwestern University, Chicago, IL, USA
| | - Seema A. Khan
- Department of Surgery, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
- Robert H. Lurie Cancer Center of Northwestern University, Chicago, IL, USA
| | - Susan E. Clare
- Department of Surgery, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
- Robert H. Lurie Cancer Center of Northwestern University, Chicago, IL, USA
| |
Collapse
|
19
|
Zhang B, Song C, Zhou B, Zhang J, Dong W, Zhang Y, Zhao X, Zhang Q. CTNNB1 and CDH1 Regulate Trophoblast Cell Adhesion and Junction Formation in Yak Placental Tissue at Different Gestational Stages. Animals (Basel) 2025; 15:876. [PMID: 40150405 PMCID: PMC11939409 DOI: 10.3390/ani15060876] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2025] [Revised: 03/15/2025] [Accepted: 03/17/2025] [Indexed: 03/29/2025] Open
Abstract
Yaks (Bos grunniens), which are distributed across the Tibetan Plateau and other high-altitude regions, are vital livestock that provide essential resources for local herders and have significant economic and ecological value [...].
Collapse
Affiliation(s)
- Bohao Zhang
- College of Veterinary Medicine, Gansu Agricultural University, Lanzhou 730070, China; (B.Z.); (C.S.); (W.D.); (Y.Z.); (X.Z.)
- College of Life Science and Biotechnology, Gansu Agricultural University, Lanzhou 730070, China; (B.Z.); (J.Z.)
- Gansu Key Laboratory of Animal Generational Physiology and Reproductive Regulation, Lanzhou 730070, China
| | - Chen Song
- College of Veterinary Medicine, Gansu Agricultural University, Lanzhou 730070, China; (B.Z.); (C.S.); (W.D.); (Y.Z.); (X.Z.)
- Gansu Key Laboratory of Animal Generational Physiology and Reproductive Regulation, Lanzhou 730070, China
| | - Bin Zhou
- College of Life Science and Biotechnology, Gansu Agricultural University, Lanzhou 730070, China; (B.Z.); (J.Z.)
- Gansu Key Laboratory of Animal Generational Physiology and Reproductive Regulation, Lanzhou 730070, China
| | - Junjun Zhang
- College of Life Science and Biotechnology, Gansu Agricultural University, Lanzhou 730070, China; (B.Z.); (J.Z.)
- Gansu Key Laboratory of Animal Generational Physiology and Reproductive Regulation, Lanzhou 730070, China
| | - Weitao Dong
- College of Veterinary Medicine, Gansu Agricultural University, Lanzhou 730070, China; (B.Z.); (C.S.); (W.D.); (Y.Z.); (X.Z.)
- Gansu Key Laboratory of Animal Generational Physiology and Reproductive Regulation, Lanzhou 730070, China
| | - Yong Zhang
- College of Veterinary Medicine, Gansu Agricultural University, Lanzhou 730070, China; (B.Z.); (C.S.); (W.D.); (Y.Z.); (X.Z.)
- College of Life Science and Biotechnology, Gansu Agricultural University, Lanzhou 730070, China; (B.Z.); (J.Z.)
- Gansu Key Laboratory of Animal Generational Physiology and Reproductive Regulation, Lanzhou 730070, China
| | - Xingxu Zhao
- College of Veterinary Medicine, Gansu Agricultural University, Lanzhou 730070, China; (B.Z.); (C.S.); (W.D.); (Y.Z.); (X.Z.)
- College of Life Science and Biotechnology, Gansu Agricultural University, Lanzhou 730070, China; (B.Z.); (J.Z.)
- Gansu Key Laboratory of Animal Generational Physiology and Reproductive Regulation, Lanzhou 730070, China
| | - Quanwei Zhang
- College of Veterinary Medicine, Gansu Agricultural University, Lanzhou 730070, China; (B.Z.); (C.S.); (W.D.); (Y.Z.); (X.Z.)
- College of Life Science and Biotechnology, Gansu Agricultural University, Lanzhou 730070, China; (B.Z.); (J.Z.)
- Gansu Key Laboratory of Animal Generational Physiology and Reproductive Regulation, Lanzhou 730070, China
| |
Collapse
|
20
|
Moccia F, Totaro A, Guerra G, Testa G. Ca 2+ Signaling in Cardiac Fibroblasts: An Emerging Signaling Pathway Driving Fibrotic Remodeling in Cardiac Disorders. Biomedicines 2025; 13:734. [PMID: 40149710 PMCID: PMC11940070 DOI: 10.3390/biomedicines13030734] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2025] [Revised: 03/13/2025] [Accepted: 03/15/2025] [Indexed: 03/29/2025] Open
Abstract
Cardiac fibrosis is a scarring event that occurs in the myocardium in response to multiple cardiovascular disorders, such as acute myocardial infarction (AMI), ischemic cardiomyopathy, dilated cardiomyopathy, hypertensive heart disease, inflammatory heart disease, diabetic cardiomyopathy, and aortic stenosis. Fibrotic remodeling is mainly sustained by the differentiation of fibroblasts into myofibroblasts, which synthesize and secrete most of the extracellular matrix (ECM) proteins. An increase in the intracellular Ca2+ concentration ([Ca2+]i) in cardiac fibroblasts is emerging as a critical mediator of the fibrogenic signaling cascade. Herein, we review the mechanisms that may shape intracellular Ca2+ signals involved in fibroblast transdifferentiation into myofibroblasts. We focus our attention on the functional interplay between inositol-1,4,5-trisphosphate (InsP3) receptors (InsP3Rs) and store-operated Ca2+ entry (SOCE). In accordance with this, InsP3Rs and SOCE drive the Ca2+ response elicited by Gq-protein coupled receptors (GqPCRs) that promote fibrotic remodeling. Then, we describe the additional mechanisms that sustain extracellular Ca2+ entry, including receptor-operated Ca2+ entry (ROCE), P2X receptors, Transient Receptor Potential (TRP) channels, and Piezo1 channels. In parallel, we discuss the pharmacological manipulation of the Ca2+ handling machinery as a promising approach to mitigate or reverse fibrotic remodeling in cardiac disorders.
Collapse
Affiliation(s)
- Francesco Moccia
- Department of Medicine and Health Sciences “V. Tiberio”, University of Molise, 86100 Campobasso, Italy; (A.T.); (G.G.); (G.T.)
| | | | | | | |
Collapse
|
21
|
Patrașcu AV, Țarcă E, Lozneanu L, Ungureanu C, Moroșan E, Parteni DE, Jehac A, Bernic J, Cojocaru E. The Role of Epithelial-Mesenchymal Transition in Osteosarcoma Progression: From Biology to Therapy. Diagnostics (Basel) 2025; 15:644. [PMID: 40075892 PMCID: PMC11898898 DOI: 10.3390/diagnostics15050644] [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: 01/24/2025] [Revised: 02/24/2025] [Accepted: 02/26/2025] [Indexed: 03/14/2025] Open
Abstract
Osteosarcoma (OS) is the most common primary malignant bone tumor, predominantly affecting children, adolescents, and young adults. Epithelial-mesenchymal transition (EMT), a process in which epithelial cells lose their cell-cell adhesion and gain migratory and invasive properties, has been extensively studied in various carcinomas. However, its role in mesenchymal tumors like osteosarcoma remains less explored. EMT is increasingly recognized as a key factor in the progression of osteosarcoma, contributing to tumor invasion, metastasis, and resistance to chemotherapy. This narrative review aims to provide a comprehensive overview of the molecular mechanisms driving EMT in osteosarcoma, highlighting the involvement of signaling pathways such as TGF-β, transcription factors like Snail, Twist, and Zeb, and the role of microRNAs in modulating EMT. Furthermore, we discuss how EMT correlates with poor prognosis and therapy resistance in osteosarcoma patients, emphasizing the potential of targeting EMT for therapeutic intervention. Recent advancements in understanding EMT in osteosarcoma have opened new avenues for treatment, including EMT inhibitors and combination therapies aimed at overcoming drug resistance. By integrating biological insights with clinical implications, this review underscores the importance of EMT as a critical process in osteosarcoma progression and its potential as a therapeutic target.
Collapse
Affiliation(s)
- Andrei-Valentin Patrașcu
- Department of Morphofunctional Sciences I—Pathology, Faculty of Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania; (A.-V.P.); (C.U.); (E.M.); (D.-E.P.); (E.C.)
| | - Elena Țarcă
- Department of Surgery II—Pediatric Surgery, Faculty of Medicine, University of Medicine and Pharmacy “Gr. T. Popa”, 700115 Iasi, Romania
| | - Ludmila Lozneanu
- Department of Morphofunctional Sciences I—Histology, Faculty of Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania;
| | - Carmen Ungureanu
- Department of Morphofunctional Sciences I—Pathology, Faculty of Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania; (A.-V.P.); (C.U.); (E.M.); (D.-E.P.); (E.C.)
| | - Eugenia Moroșan
- Department of Morphofunctional Sciences I—Pathology, Faculty of Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania; (A.-V.P.); (C.U.); (E.M.); (D.-E.P.); (E.C.)
| | - Diana-Elena Parteni
- Department of Morphofunctional Sciences I—Pathology, Faculty of Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania; (A.-V.P.); (C.U.); (E.M.); (D.-E.P.); (E.C.)
| | - Alina Jehac
- Second Dental Medicine Department, Faculty of Dental Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania;
| | - Jana Bernic
- Discipline of Pediatric Surgery, “Nicolae Testemițanu” State University of Medicine and Pharmacy, MD-2001 Chisinau, Moldova;
| | - Elena Cojocaru
- Department of Morphofunctional Sciences I—Pathology, Faculty of Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania; (A.-V.P.); (C.U.); (E.M.); (D.-E.P.); (E.C.)
| |
Collapse
|
22
|
Wan X, Zhan J, Ye S, Chen C, Li R, Shen M. Construction of a prognostic model and analysis of related mechanisms in breast cancer based on multiple datasets. Transl Cancer Res 2025; 14:930-948. [PMID: 40104717 PMCID: PMC11912066 DOI: 10.21037/tcr-24-838] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Accepted: 12/26/2024] [Indexed: 03/20/2025]
Abstract
Background Breast cancer (BC) is a common tumor among women and is a heterogeneous disease with many subtypes. Each subtype shows different clinical presentations, disease trajectories and prognoses, and different responses to neoadjuvant therapy; thus, a new and universal prognostic biomarker for BC patients is urgently needed. Our goal is to identify a novel prognostic molecular biomarker that can accurately predict the outcome of all BC subtypes and guide their clinical management. Methods Utilizing data from The Cancer Genome Atlas (TCGA), we analyzed differential gene expression and patient clinical data. Weighted gene coexpression network analysis (WGCNA), Cox univariate regression and least absolute shrinkage and selection operator (LASSO) analysis were used to construct a prognostic model; the differential expression of the core genes in this model was validated via real-time quantitative polymerase chain reaction (RT-qPCR), and the reliability of the predictive model was validated in both an internal cohort and a BC patient dataset from the Gene Expression Omnibus (GEO) database. Further studies, such as gene set variation analysis (GSVA) and gene set enrichment analysis (GSEA), were performed to investigate the enrichment of signaling pathways. The CIBERSORT algorithm was used to estimate immune infiltration and tumor mutation burden (TMB), and drug sensitivity analysis was performed to evaluate the treatment response. Results A total of 1,643 differentially expressed genes were identified. After WGCNA and Cox regression combined with LASSO analysis, 15 genes were identified by screening and used to establish a prognostic gene signature. Further analysis revealed that the epithelial-mesenchymal transition (EMT) pathway gene signature was enriched in these genes. Each patient was assigned a risk score, and according to the median risk score, patients were classified into a high-risk group or a low-risk group. The prognosis of the low-risk group was better than that of the high-risk group (P<0.01), and analyses of two independent GEO validation cohorts yielded similar results. Furthermore, a nomogram was constructed and found to perform well in predicting prognosis. GSVA revealed that the EMT pathway, transforming growth factor β (TGF-β) signaling pathway and PI3K-Akt signaling pathway genes were enriched in the high-risk group, and the Wnt-β-catenin signaling pathway, DNA repair pathway and P53 pathway gene sets were enriched in the low-risk group. GSEA revealed genes related to TGF-β signaling and the PI3K-Akt signaling pathways were enriched in the high-risk group. CIBERSORT demonstrated that the low-risk group had greater infiltration of antitumor immune cells. The TMB and drug sensitivity results suggested that immunotherapy and chemotherapy are likely to be more effective in the low-risk group. Conclusions We established a new EMT pathway-related prognostic gene signature that can be used to effectively predict BC prognosis and treatment response.
Collapse
Affiliation(s)
- Xiaofeng Wan
- National Health Commission (NHC) Key Lab of Reproduction Regulation (Shanghai Institute for Biomedical and Pharmaceutical Technologies), Shanghai, China
| | - Jianmin Zhan
- National Health Commission (NHC) Key Lab of Reproduction Regulation (Shanghai Institute for Biomedical and Pharmaceutical Technologies), Shanghai, China
| | - Shuke Ye
- National Health Commission (NHC) Key Lab of Reproduction Regulation (Shanghai Institute for Biomedical and Pharmaceutical Technologies), Shanghai, China
| | - Chuanrong Chen
- Department of Oncology, Yijishan Hospital of Wannan Medical College, Wuhu, China
| | - Runsheng Li
- National Health Commission (NHC) Key Lab of Reproduction Regulation (Shanghai Institute for Biomedical and Pharmaceutical Technologies), Shanghai, China
| | - Ming Shen
- National Health Commission (NHC) Key Lab of Reproduction Regulation (Shanghai Institute for Biomedical and Pharmaceutical Technologies), Shanghai, China
| |
Collapse
|
23
|
Chen SY, Zhang YL, Li XR, Wang JR, Li KP, Wan S, Yang JW, Wang H, Cao JL, Wang CY, Fan XP, Fu SJ, Ding LY, Che TJ, Yang L. BIN1 inhibited tumor growth, metastasis and stemness by ALDH1/NOTCH pathway in bladder carcinoma. Hereditas 2025; 162:29. [PMID: 40016843 PMCID: PMC11866615 DOI: 10.1186/s41065-025-00384-w] [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: 09/25/2024] [Accepted: 02/01/2025] [Indexed: 03/01/2025] Open
Abstract
BACKGROUND Bladder cancer (BLCA) represents one of the most prevalent urological malignancies worldwide. Bridging integrator 1 (BIN1), a well-characterized tumor suppressor that interacts with and inhibits oncogenic Myc transcription factors, has demonstrated crucial roles in various cancer types. However, its specific functions and underlying molecular mechanisms in BLCA development and progression remain poorly understood. This study aims to elucidate the role of BIN1 in regulating BLCA cell proliferation, metastasis, and cancer stem cell properties. METHODS Using urinary proteomics analysis, we identified BIN1 as a significantly dysregulated protein in BLCA. The clinical significance of BIN1 was further validated through comprehensive analyses of public databases. BIN1 expression levels defined distinct molecular and immunological subtypes of BLCA. Through proteomic profiling of BIN1-overexpressing UMUC3 cells and corresponding controls, we identified ALDH1 as a key downstream effector in the BIN1-regulated ALDH1/NOTCH signaling axis. We employed multiple experimental approaches, including Western blot analysis, quantitative RT-PCR, immunofluorescence staining, wound healing assays, transwell migration assays, colony formation assays, tumor sphere formation assays, flow cytometry, CCK8 proliferation assays, and cell transfection experiments. RESULTS We observed significant downregulation of BIN1 in both BLCA tissues and cell lines compared to normal adjacent tissues and SV-HUC-1 cells, respectively. BIN1 overexpression inhibited cancer cell proliferation by promoting apoptosis and suppressed epithelial-mesenchymal transition (EMT), thereby reducing local invasion and distant metastasis. Additionally, BIN1 regulated cancer stem cell properties through modulation of ALDH1 expression, with NOTCH2 acting as a crucial downstream mediator of ALDH1 signaling. CONCLUSION Our findings demonstrate that BIN1 functions as a tumor suppressor in BLCA and suggest its potential utility as both a diagnostic biomarker and therapeutic target for BLCA treatment.
Collapse
Affiliation(s)
- Si-Yu Chen
- Department of Urology, The Second Hospital of Lanzhou University, Lanzhou, China
- Gansu Province Clinical Research Center for Urinary System Disease, Lanzhou, China
| | - Ya-Long Zhang
- Department of Urology, The Second Hospital of Lanzhou University, Lanzhou, China
- Gansu Province Clinical Research Center for Urinary System Disease, Lanzhou, China
| | - Xiao-Ran Li
- Department of Urology, The Second Hospital of Lanzhou University, Lanzhou, China
- Gansu Province Clinical Research Center for Urinary System Disease, Lanzhou, China
| | - Ji-Rong Wang
- Department of Urology, The Second Hospital of Lanzhou University, Lanzhou, China
- Gansu Province Clinical Research Center for Urinary System Disease, Lanzhou, China
| | - Kun-Peng Li
- Department of Urology, The Second Hospital of Lanzhou University, Lanzhou, China
- Gansu Province Clinical Research Center for Urinary System Disease, Lanzhou, China
| | - Shun Wan
- Department of Urology, The Second Hospital of Lanzhou University, Lanzhou, China
- Gansu Province Clinical Research Center for Urinary System Disease, Lanzhou, China
| | - Jian-Wei Yang
- Department of Urology, The Second Hospital of Lanzhou University, Lanzhou, China
- Gansu Province Clinical Research Center for Urinary System Disease, Lanzhou, China
| | - Hao Wang
- Department of Urology, The Second Hospital of Lanzhou University, Lanzhou, China
- Gansu Province Clinical Research Center for Urinary System Disease, Lanzhou, China
| | - Jin-Long Cao
- Department of Urology, The Second Hospital of Lanzhou University, Lanzhou, China
- Gansu Province Clinical Research Center for Urinary System Disease, Lanzhou, China
| | - Chen-Yang Wang
- Department of Urology, The Second Hospital of Lanzhou University, Lanzhou, China
- Gansu Province Clinical Research Center for Urinary System Disease, Lanzhou, China
| | - Xin-Peng Fan
- Department of Urology, The Second Hospital of Lanzhou University, Lanzhou, China
- Gansu Province Clinical Research Center for Urinary System Disease, Lanzhou, China
| | - Sheng-Jun Fu
- Department of Urology, The Second Hospital of Lanzhou University, Lanzhou, China
- Gansu Province Clinical Research Center for Urinary System Disease, Lanzhou, China
| | - Li-Yun Ding
- School of Physical Science and Technology, Lanzhou University, Lanzhou, China
| | - Tuan-Jie Che
- Baiyuan Company for Gene Technology, Lanzhou, China
| | - Li Yang
- Department of Urology, The Second Hospital of Lanzhou University, Lanzhou, China.
- Gansu Province Clinical Research Center for Urinary System Disease, Lanzhou, China.
| |
Collapse
|
24
|
El Masri R, Iannuzzo A, Kuentz P, Tacine R, Vincent M, Barbarot S, Morice-Picard F, Boralevi F, Oillarburu N, Mazereeuw-Hautier J, Duffourd Y, Faivre L, Sorlin A, Vabres P, Delon J. A postzygotic GNA13 variant upregulates the RHOA/ROCK pathway and alters melanocyte function in a mosaic skin hypopigmentation syndrome. Nat Commun 2025; 16:1751. [PMID: 39966435 PMCID: PMC11836271 DOI: 10.1038/s41467-025-56995-4] [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: 08/23/2023] [Accepted: 02/04/2025] [Indexed: 02/20/2025] Open
Abstract
The genetic bases of mosaic pigmentation disorders have increasingly been identified, but these conditions remain poorly characterised, and their pathophysiology is unclear. Here, we report in four unrelated patients that a recurrent postzygotic mutation in GNA13 is responsible for a recognizable syndrome with hypomelanosis of Ito associated with developmental anomalies. GNA13 encodes Gα13, a subunit of αβγ heterotrimeric G proteins coupled to specific transmembrane receptors known as G-protein coupled receptors. In-depth functional investigations revealed that this R200K mutation provides a gain of function to Gα13. Mechanistically, we show that this variant hyperactivates the RHOA/ROCK signalling pathway that consequently increases actin polymerisation and myosin light chains phosphorylation, and promotes melanocytes rounding. Our results also indicate that R200K Gα13 hyperactivates the YAP signalling pathway. All these changes appear to affect cell migration and adhesion but not the proliferation. Our results suggest that hypopigmentation can result from a defect in melanosome transfer to keratinocytes due to cell shape alterations. These findings highlight the interaction between heterotrimeric G proteins and the RHOA pathway, and their role in melanocyte function.
Collapse
Affiliation(s)
- Rana El Masri
- Université Paris Cité, CNRS, Inserm, Institut Cochin, Paris, France
- Department of Cell Physiology & Metabolism, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Alberto Iannuzzo
- Université Paris Cité, CNRS, Inserm, Institut Cochin, Paris, France
| | - Paul Kuentz
- UFR des Sciences de Santé, Inserm - Université de Bourgogne UMR1231 GAD "Génétique des Anomalies du Développement", FHU-TRANSLAD, Dijon, France
- Oncobiologie Génétique Bioinformatique, PCBio, Centre Hospitalier Universitaire de Besançon, Besançon, France
| | - Rachida Tacine
- Université Paris Cité, CNRS, Inserm, Institut Cochin, Paris, France
| | - Marie Vincent
- Unité de Génétique clinique, Service de génétique médicale, CHU de Nantes - Hôpital Mère-Enfant, Nantes, France
| | - Sébastien Barbarot
- Nantes Université, Department of Dermatology, CHU Nantes, INRAE, UMR 1280, PhAN, Nantes, France
| | - Fanny Morice-Picard
- MAGEC Reference Centre for Rare Genetic Skin Diseases, Paediatric Dermatology Unit, Department of Dermatology, CHU de Bordeaux - GH Pellegrin, Bordeaux, France
| | - Franck Boralevi
- MAGEC Reference Centre for Rare Genetic Skin Diseases, Paediatric Dermatology Unit, Department of Dermatology, CHU de Bordeaux - GH Pellegrin, Bordeaux, France
| | - Naia Oillarburu
- Service de dermatologie, CHU de Toulouse - Hôpital Larrey, Toulouse, France
| | | | - Yannis Duffourd
- UFR des Sciences de Santé, Inserm - Université de Bourgogne UMR1231 GAD "Génétique des Anomalies du Développement", FHU-TRANSLAD, Dijon, France
- CHU Dijon, Unité Fonctionnelle "Innovation diagnostique dans les maladies rares", FHU-TRANSLAD & Institut GIMI, Dijon, France
| | - Laurence Faivre
- UFR des Sciences de Santé, Inserm - Université de Bourgogne UMR1231 GAD "Génétique des Anomalies du Développement", FHU-TRANSLAD, Dijon, France
- CHU Dijon, Centre de Génétique et Centres de référence Anomalies du Développement et Déficience Intellectuelle, FHU-TRANSLAD & Institut GIMI, Dijon, France
| | - Arthur Sorlin
- UFR des Sciences de Santé, Inserm - Université de Bourgogne UMR1231 GAD "Génétique des Anomalies du Développement", FHU-TRANSLAD, Dijon, France
- MAGEC Reference Centre for Rare Genetic Skin Diseases and Paediatric Dermatology Unit, Department of Paediatrics, University Hospital Dijon-Bourgogne, FHU-TRANSLAD & Institut GIMI, Dijon, France
- National Center of Genetics (NCG), Laboratoire national de santé (LNS), 1 Rue Louis Rech, Dudelange, Luxembourg
| | - Pierre Vabres
- UFR des Sciences de Santé, Inserm - Université de Bourgogne UMR1231 GAD "Génétique des Anomalies du Développement", FHU-TRANSLAD, Dijon, France.
- MAGEC Reference Centre for Rare Genetic Skin Diseases and Paediatric Dermatology Unit, Department of Paediatrics, University Hospital Dijon-Bourgogne, FHU-TRANSLAD & Institut GIMI, Dijon, France.
- Rare Disease Collaborative Network (RDCN) Adult Mosaic Disorders Clinic, St John's Institute of Dermatology, Guy's and St Thomas' NHS Foundation Trust, London, United Kingdom.
| | - Jérôme Delon
- Université Paris Cité, CNRS, Inserm, Institut Cochin, Paris, France.
| |
Collapse
|
25
|
Meeusen B, Ambjørn SM, Veis J, Riley RC, Vit G, Brauer BL, Møller MH, Greiner EC, Chan CB, Weisser MB, Garvanska DH, Zhu H, Davey NE, Kettenbach AN, Ogris E, Nilsson J. A functional map of phosphoprotein phosphatase regulation identifies an evolutionary conserved reductase for the catalytic metal ions. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.12.637884. [PMID: 39990307 PMCID: PMC11844454 DOI: 10.1101/2025.02.12.637884] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 02/25/2025]
Abstract
Serine/Threonine phosphoprotein phosphatases (PPPs, PP1-PP7) are conserved metalloenzymes and central to intracellular signaling in eukaryotes, but the details of their regulation is poorly understood. To address this, we performed genome-wide CRISPR knockout and focused base editor screens in PPP perturbed conditions to establish a high-resolution functional map of PPP regulation that pinpoints novel regulatory mechanisms. Through this, we identify the orphan reductase CYB5R4 as an evolutionarily conserved activator of PP4 and PP6, but not the closely related PP2A. Heme binding is essential for CYB5R4 function and mechanistically involves the reduction of the metal ions in the active site. Importantly, CYB5R4-mediated activation of PP4 is critical for cell viability when cells are treated with DNA damage-inducing agents known to cause oxidative stress. The discovery of a dedicated PPP reductase points to shared regulatory principles with protein tyrosine phosphatases, where specific enzymes dictate activity by regulating the active site redox state. In sum, our work provides a resource for understanding PPP function and the regulation of intracellular signaling.
Collapse
Affiliation(s)
- Bob Meeusen
- Novo Nordisk Foundation Center for Protein Research, University of Copenhagen, Copenhagen, DK
| | - Sara M. Ambjørn
- Novo Nordisk Foundation Center for Protein Research, University of Copenhagen, Copenhagen, DK
| | - Jiri Veis
- Max Perutz Labs, Vienna Biocenter Campus (VBC), Dr.-Bohr-Gasse 9 / Vienna Biocenter 5, 1030, Vienna, Austria. Medical University of Vienna, Max Perutz Labs, Dr.-Bohr-Gasse 9 / Vienna Biocenter 5, 1030, Vienna, Austria
| | - Rachel C. Riley
- Department of Biochemistry and Cell Biology, Dartmouth Geisel School of Medicine, Hanover, NH, USA
| | - Gianmatteo Vit
- Novo Nordisk Foundation Center for Protein Research, University of Copenhagen, Copenhagen, DK
| | - Brooke L. Brauer
- Department of Biochemistry and Cell Biology, Dartmouth Geisel School of Medicine, Hanover, NH, USA
| | - Mads H. Møller
- Novo Nordisk Foundation Center for Protein Research, University of Copenhagen, Copenhagen, DK
| | - Elora C. Greiner
- Department of Biochemistry and Cell Biology, Dartmouth Geisel School of Medicine, Hanover, NH, USA
| | - Camilla B. Chan
- Novo Nordisk Foundation Center for Protein Research, University of Copenhagen, Copenhagen, DK
| | - Melanie B. Weisser
- Novo Nordisk Foundation Center for Protein Research, University of Copenhagen, Copenhagen, DK
| | - Dimitriya H. Garvanska
- Novo Nordisk Foundation Center for Protein Research, University of Copenhagen, Copenhagen, DK
| | - Hao Zhu
- University of Kansas Medical Center, Kansas City, KS, USA
| | | | - Arminja N. Kettenbach
- Department of Biochemistry and Cell Biology, Dartmouth Geisel School of Medicine, Hanover, NH, USA
- Dartmouth Cancer Center, Lebanon, NH, USA
| | - Egon Ogris
- Max Perutz Labs, Vienna Biocenter Campus (VBC), Dr.-Bohr-Gasse 9 / Vienna Biocenter 5, 1030, Vienna, Austria. Medical University of Vienna, Max Perutz Labs, Dr.-Bohr-Gasse 9 / Vienna Biocenter 5, 1030, Vienna, Austria
| | - Jakob Nilsson
- Novo Nordisk Foundation Center for Protein Research, University of Copenhagen, Copenhagen, DK
| |
Collapse
|
26
|
Naimo GD, Forestiero M, Giordano F, Leonetti AE, Gelsomino L, Panno ML, Andò S, Mauro L. Adiponectin Influences the Behavior of Stem Cells in Hormone-Resistant Breast Cancer. Cells 2025; 14:286. [PMID: 39996758 PMCID: PMC11853953 DOI: 10.3390/cells14040286] [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: 01/21/2025] [Revised: 02/11/2025] [Accepted: 02/13/2025] [Indexed: 02/26/2025] Open
Abstract
In the breast tumor microenvironment (TME), adipocytes exert a selective pressure on the behavior of breast cancer stem cells (BCSCs), which are involved in endocrine therapy resistance. In obesity, adipocytes secrete reduced levels of adiponectin, which promotes the growth and progression of ERα-positive breast cancer (BC). Here, we examined how low adiponectin levels affect the enrichment of the BCSC subpopulation and the mechanisms contributing to the maintenance of endocrine therapy resistance in BC. Flow cytometry, qRT-PCR, and Western blotting analysis were performed to assess stemness, the cell cycle, and apoptosis markers in MCF-7 wild-type (WT) and tamoxifen-resistant (TR) mammospheres. nLC-MS/MS was employed to profile and compare the proteome of BCSCs. Differentially expressed proteins were intersected with data from the MetacoreTM dataset. Our study demonstrated that adiponectin increased the percentage of CD44+/CD24-/ALDH1+ stem-like cells in TR MCF-7 mammospheres. Specifically, adiponectin contributed to the maintenance of BCSC bulk in TR MCF-7 cells through a slow cycling rate, supported by decreased levels of Cyclin D1 and Ki67 and increased p21 and p27 expression, and through escape from apoptosis, sustained by reduced ROS production and preserved maintenance of mitochondrial membrane potential. Our results provide new insights into the contribution of adiponectin to poor ERα-positive BC outcomes. Deeply understanding adiponectin's role in stemness may disclose novel therapeutic approaches to treat hormone-resistant obese BC patients.
Collapse
Affiliation(s)
- Giuseppina Daniela Naimo
- Department of Pharmacy Health and Nutritional Sciences, University of Calabria, 87036 Arcavacata di Rende, CS, Italy; (G.D.N.); (M.F.); (F.G.); (A.E.L.); (L.G.); (M.L.P.)
| | - Martina Forestiero
- Department of Pharmacy Health and Nutritional Sciences, University of Calabria, 87036 Arcavacata di Rende, CS, Italy; (G.D.N.); (M.F.); (F.G.); (A.E.L.); (L.G.); (M.L.P.)
| | - Francesca Giordano
- Department of Pharmacy Health and Nutritional Sciences, University of Calabria, 87036 Arcavacata di Rende, CS, Italy; (G.D.N.); (M.F.); (F.G.); (A.E.L.); (L.G.); (M.L.P.)
| | - Adele Elisabetta Leonetti
- Department of Pharmacy Health and Nutritional Sciences, University of Calabria, 87036 Arcavacata di Rende, CS, Italy; (G.D.N.); (M.F.); (F.G.); (A.E.L.); (L.G.); (M.L.P.)
| | - Luca Gelsomino
- Department of Pharmacy Health and Nutritional Sciences, University of Calabria, 87036 Arcavacata di Rende, CS, Italy; (G.D.N.); (M.F.); (F.G.); (A.E.L.); (L.G.); (M.L.P.)
| | - Maria Luisa Panno
- Department of Pharmacy Health and Nutritional Sciences, University of Calabria, 87036 Arcavacata di Rende, CS, Italy; (G.D.N.); (M.F.); (F.G.); (A.E.L.); (L.G.); (M.L.P.)
| | - Sebastiano Andò
- Department of Pharmacy Health and Nutritional Sciences, University of Calabria, 87036 Arcavacata di Rende, CS, Italy; (G.D.N.); (M.F.); (F.G.); (A.E.L.); (L.G.); (M.L.P.)
- Centro Sanitario, University of Calabria, 87036 Arcavacata di Rende, CS, Italy
| | - Loredana Mauro
- Department of Pharmacy Health and Nutritional Sciences, University of Calabria, 87036 Arcavacata di Rende, CS, Italy; (G.D.N.); (M.F.); (F.G.); (A.E.L.); (L.G.); (M.L.P.)
| |
Collapse
|
27
|
Lu J, Li C, Zhao T, Li F, Yao Z, Dong Y, Gong Z, Yan Y, Luo X, Wang H. Sodium Butyrate Alleviates Heat Stress-Induced Oxidative Stress and Skeletal Muscle Homeostasis Disruption by Promoting Autophagy in Mice. Nutrients 2025; 17:696. [PMID: 40005024 PMCID: PMC11858507 DOI: 10.3390/nu17040696] [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: 11/22/2024] [Revised: 01/19/2025] [Accepted: 02/13/2025] [Indexed: 02/27/2025] Open
Abstract
BACKGROUND The gradual rise in global temperatures can affect skeletal muscle development and intestinal microorganisms. However, the influence of microbial metabolites on skeletal muscle homeostasis under heat stress (HS) remains unclear. METHODS C57BL/6J mice were exposed to normal temperature or 40 °C conditions for 3 d, 7 d, or 14 d. The HS 7 d mice also were treated with sodium butyrate (NaB, 200 mg/kg, gavage). RESULTS Strikingly, the body weight, antioxidative ability (MDA, T-SOD, and GSH-Px), and average cross-sectional area decreased, but the blood glucose and core temperature increased under HS. However, the NaB treatment reversed these effects. Meanwhile, HS also increased the levels of TNF-α and CORT. Additionally, HS led to a reduction in the villus height and an increase in the crypt depth of the intestine. Microbial 16S rRNA sequencing analysis revealed that HS caused gut microbiota dysbiosis. NaB increased the expression of HSP70 under HS, to maintain skeletal muscle homeostasis. HS stimulated the expression of Pax7, which indicates that skeletal muscle homeostasis was disrupted. Meanwhile, the expressions of MyoG and MyoD were decreased under HS. The immunofluorescence results also show that HS triggered a shift from slow muscle fibers (MYH7) to fast muscle fibers (MYH1). However, NaB recovered the expressions of these muscle-related factors. HS inhibited autophagy initiation (mTOR, Beclin1, Atg5, Atg7, and Atg12), the formation (LC3 II/LC3 I) of autophagosomes, and the binding (p62 and LAMP1) of lysosomes to autophagosomes, which were activated by NaB. C2C12 cells were treated with H2O2 to simulate skeletal muscle oxidative stress, and treated with NaB in advance. Oxidative stress disrupted the homeostasis of the C2C12 cells, characterized by an increase in Pax 7 and decreases in MyoG and MyoD, but these changes were reversed by the NaB treatment. Meanwhile, NaB was unable to maintain the stable expression of Pax7 when autophagy was inhibited. CONCLUSIONS This suggests that NaB can regulate oxidative stress induced by HS through autophagy to maintain skeletal muscle homeostasis.
Collapse
Affiliation(s)
- Jiayin Lu
- College of Veterinary Medicine, Shanxi Agricultural University, No.1 Mingxian South Road, Taigu 030801, China; (C.L.); (T.Z.); (F.L.); (Z.Y.); (Y.D.); (Z.G.); (Y.Y.); (X.L.)
| | | | | | | | | | | | | | | | | | - Haidong Wang
- College of Veterinary Medicine, Shanxi Agricultural University, No.1 Mingxian South Road, Taigu 030801, China; (C.L.); (T.Z.); (F.L.); (Z.Y.); (Y.D.); (Z.G.); (Y.Y.); (X.L.)
| |
Collapse
|
28
|
Li J, Huang N, Zhang X, Wang H, Chen J, Wei Q. Functional analysis of yak alveolar type II epithelial cells at high and low altitudes based on single-cell sequencing. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2025; 1872:119889. [PMID: 39681250 DOI: 10.1016/j.bbamcr.2024.119889] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/13/2024] [Revised: 11/28/2024] [Accepted: 12/06/2024] [Indexed: 12/18/2024]
Abstract
The adaptation of lung cells to high-altitude environments represents a notable gap in our understanding of how animals cope with hypoxic conditions. Alveolar epithelial cells type II (AEC II) are crucial for lung development and repair. However, their, specific role in the adaptation of yaks to high-altitude environments remains unclear. In this study, we aimed to address this gap by investigating the differential responses of AEC II in yaks at high and low altitudes (4000 m and 2600 m, respectively). We used the 10 × scRNA-seq technology to construct a comprehensive cell atlas of yak lung tissue, and identified 15 distinct cell classes. AEC II in high-altitude yaks revealed increased immunomodulatory, adhesive, and metabolic activities, which are crucial for maintaining lung tissue stability and energy supply under hypoxic conditions. Furthermore, alveolar epithelial progenitor cells within AEC II can differentiate into both Alveolar epithelial cell type I (AEC I) and AEC II. SHIP1 and other factors are promoters of AEC I transdifferentiation, whereas SFTPC and others promote AEC II transdifferentiation. This study provides new insights into the evolutionary adaptation of lung cells in plateau animals by elucidating the molecular mechanisms underlying AEC II adaptation to high-altitude environments.
Collapse
Affiliation(s)
- Jingyi Li
- College of Eco-Environmental Engineering, Qinghai University, Xining, Qinghai, China
| | - Nating Huang
- College of Eco-Environmental Engineering, Qinghai University, Xining, Qinghai, China
| | - Xun Zhang
- College of Eco-Environmental Engineering, Qinghai University, Xining, Qinghai, China
| | - Huizhen Wang
- College of Eco-Environmental Engineering, Qinghai University, Xining, Qinghai, China
| | - Jiarui Chen
- College of Eco-Environmental Engineering, Qinghai University, Xining, Qinghai, China
| | - Qing Wei
- College of Eco-Environmental Engineering, Qinghai University, Xining, Qinghai, China; State Key Laboratory of Plateau Ecology and Agriculture, Qinghai University, Xining, Qinghai, China.
| |
Collapse
|
29
|
Hadjadji C, Devalloir Q, Gaillard C, van den Brink NW, Scheifler R. Evidence linking cadmium and/or lead exposure to immunomodulatory effects in mammals based upon an adverse outcome pathways approach, and research perspectives. CHEMOSPHERE 2025; 371:144056. [PMID: 39746483 DOI: 10.1016/j.chemosphere.2024.144056] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/23/2024] [Revised: 12/20/2024] [Accepted: 12/30/2024] [Indexed: 01/04/2025]
Abstract
For decades, studies have shown how exposure to non-essential trace metals such as lead (Pb) and cadmium (Cd) largely impact global wildlife. Ecoimmunotoxicology has emerged in the past two decades and focuses on the effects of pollutants on the immune system of free-ranging organisms. Adverse outcome pathways (AOPs) represent a conceptual approach to explore the mechanistic linkage between a molecular initiating event and adverse outcomes, potentially at all biological levels of organisation. The present paper proposes putative AOPs related to the effects of Cd, Pb, and the mixture Cd-Pb, on the immune system of mammals to address future questions in ecoimmunotoxicology. Molecular Initiating Events for both metals relate to entrance in cells through Ca2+ channels or bond to cell surfaces. Exposure to Cd, Pb and Cd-Pb share several similar Key Events (KEs), primarily an increase of oxidative stress (OS) in immune cells through production of reactive oxygen species. For both metals and the mixture, OS affects mitochondrial membranes, and induces apoptosis, ultimately decreasing immune cell number. Both metals affect innate immune system through nuclear factor kappa B (NF-κB) and mitogen-activated protein kinase (MAPK) inflammatory signalling pathways, leading to an upregulation of inflammatory markers and mediators. Adaptive immune system is also affected by the exposure to both metals though a decrease of CD4+/CD8+ ratio, a decrease of MHCII, an inactivation of TH1 and TH2 response, and an inhibition of the humoral response mediated by various Ig. Mixture effects of Cd-Pb are less documented resulting in a more speculative AOP, but potential synergic and antagonistic effects were identified. According to our AOPs, further research in ecoimmunotoxicology of metals in free-ranging mammals should focus on KEs related to NF-κB/MAPK inflammatory signalling pathways, changes in CD4+/CD8+ ratio and MHCII complexes, and on AOs related to auto-immune disorders and on the effective increase of infection rate, particularly in case of exposure to metal mixtures.
Collapse
Affiliation(s)
- Cloe Hadjadji
- Swiss Ornithological Institute, Seerose 1, CH-6204, Sempach, Switzerland; Laboratoire Chrono-Environnement, UMR 6249, CNRS/Université de Franche-Comté, 16 route de Gray, 25000, Besançon, France.
| | - Quentin Devalloir
- Laboratoire Chrono-Environnement, UMR 6249, CNRS/Université de Franche-Comté, 16 route de Gray, 25000, Besançon, France
| | - Colette Gaillard
- Laboratoire Chrono-Environnement, UMR 6249, CNRS/Université de Franche-Comté, 16 route de Gray, 25000, Besançon, France
| | - Nico W van den Brink
- Division of Toxicology, Wageningen University, Box 8000, 6700 EA, Wageningen, the Netherlands
| | - Renaud Scheifler
- Laboratoire Chrono-Environnement, UMR 6249, CNRS/Université de Franche-Comté, 16 route de Gray, 25000, Besançon, France
| |
Collapse
|
30
|
Park Y, Matsumoto S, Ogata K, Ma B, Kanada R, Isaka Y, Arichi N, Liang X, Maki R, Kozasa T, Okuno Y, Ohno H, Ishihama Y, Toyoshima F. Receptor-independent regulation of Gα13 by alpha-1-antitrypsin C-terminal peptides. J Biol Chem 2025; 301:108136. [PMID: 39730062 PMCID: PMC11815680 DOI: 10.1016/j.jbc.2024.108136] [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: 09/11/2024] [Revised: 12/17/2024] [Accepted: 12/19/2024] [Indexed: 12/29/2024] Open
Abstract
Alpha-1-antitrypsin (AAT), a circulating serine protease inhibitor, is an acute inflammatory response protein with anti-inflammatory functions. The C-terminal peptides of AAT are found in various tissues and have been proposed as putative bioactive peptides with multiple functions, but its mechanism of action remains unclear. We previously reported that a mouse AAT C-terminal peptide of 35 amino acids (mAAT-C1-35) penetrates plasma membrane and associates guanine nucleotide-binding protein subunit alpha 13 (Gα13). Here, we show that mAAT-C1-35 binds directly to the guanosine diphosphate (GDP)-bound form of Gα13 through the N-terminal region (mAAT-C1-17), thereby facilitating the interaction of Gα13・GDP with its effector proteins. The minimal sequence (mAAT-C3-16) and essential amino acid residue (Phe11) of mAAT-C1-17 were identified as being necessary for this effect. A molecular dynamics simulation for the Gα13・GDP-mAAT-C1-17 complex model showed that binding of mAAT-C1-17 to the region surrounded by switch regions of Gα13 stabilizes the flexible switch II and III regions, thereby maintaining their active conformation. In addition, mAAT-C1-35 activates the Gα13 signaling pathway in cells where Phe11 is required. Our study reveals the structure-based mechanism of action of AAT-C peptides in the regulation of Gα13 and demonstrates that AAT-C peptides represent a biological peptide capable of activating G protein signals in a manner that is independent of G-protein-coupled receptors.
Collapse
Affiliation(s)
- Yonghak Park
- Department of Biosystems Science, Institute for Life and Medical Sciences, Kyoto University, Kyoto, Japan; Department of Mammalian and Regulatory Networks, Graduate School of Biostudies, Kyoto University, Kyoto, Japan
| | - Shigeyuki Matsumoto
- Department of Biomedical Data Intelligence, Graduate School of Medicine, Kyoto University, Kyoto, Japan.
| | - Kosuke Ogata
- Department of Molecular Systems BioAnalysis, Graduate School of Pharmaceutical Sciences, Kyoto University, Kyoto, Japan
| | - Biao Ma
- HPC- and AI-driven Drug Development Platform Division, RIKEN Center for Computational Science, Kobe, Hyogo, Japan
| | - Ryo Kanada
- HPC- and AI-driven Drug Development Platform Division, RIKEN Center for Computational Science, Kobe, Hyogo, Japan
| | - Yuta Isaka
- HPC- and AI-driven Drug Development Platform Division, RIKEN Center for Computational Science, Kobe, Hyogo, Japan
| | - Norihito Arichi
- Department of Bioorganic Medicinal Chemistry, Graduate School of Pharmaceutical Sciences, Kyoto University, Kyoto, Japan
| | - Xiaowen Liang
- Department of Biosystems Science, Institute for Life and Medical Sciences, Kyoto University, Kyoto, Japan; Department of Mammalian and Regulatory Networks, Graduate School of Biostudies, Kyoto University, Kyoto, Japan
| | - Ritsuko Maki
- Department of Biosystems Science, Institute for Life and Medical Sciences, Kyoto University, Kyoto, Japan
| | - Tohru Kozasa
- Department of Biochemistry, Yokohama University of Pharmacy, Yokohama, Japan
| | - Yasushi Okuno
- Department of Biomedical Data Intelligence, Graduate School of Medicine, Kyoto University, Kyoto, Japan; HPC- and AI-driven Drug Development Platform Division, RIKEN Center for Computational Science, Kobe, Hyogo, Japan
| | - Hiroaki Ohno
- Department of Bioorganic Medicinal Chemistry, Graduate School of Pharmaceutical Sciences, Kyoto University, Kyoto, Japan
| | - Yasushi Ishihama
- Department of Molecular Systems BioAnalysis, Graduate School of Pharmaceutical Sciences, Kyoto University, Kyoto, Japan
| | - Fumiko Toyoshima
- Department of Biosystems Science, Institute for Life and Medical Sciences, Kyoto University, Kyoto, Japan; Department of Mammalian and Regulatory Networks, Graduate School of Biostudies, Kyoto University, Kyoto, Japan; Department of Homeostatic Medicine, Medical Research Laboratory, Institute of Integrated Research, Institute of Science Tokyo, Tokyo, Japan.
| |
Collapse
|
31
|
Cho Y, Song MK, Kim DI, Kim MS, Lee K. Adverse outcome pathway-based assessment of pulmonary toxicity from the in vivo mixture of biocides dinotefuran and cetylpyridinium chloride. Heliyon 2025; 11:e42134. [PMID: 39925360 PMCID: PMC11804698 DOI: 10.1016/j.heliyon.2025.e42134] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2024] [Revised: 12/24/2024] [Accepted: 01/20/2025] [Indexed: 02/11/2025] Open
Abstract
Despite the increasing use of biocides globally and their widespread application in various formulations, the understanding of the toxicity of biocide mixtures remains limited. We previously identified cetylpyridinium chloride and dinotefuran as a potential binary biocidal combination associated with pulmonary fibrosis, based on two intersecting adverse outcome pathways (AOPs) using the molecular initiating events (MIE) modeling method and in vitro testing. These compounds activate or inhibit toll-like receptor 4 (TLR4) and peroxisome proliferator-activated receptor-gamma (PPAR-γ), which are associated with pathways having the potential to cause pulmonary fibrosis. In this study, we aimed to validate these AOPs by assessing the toxicity of cetylpyridinium chloride and dinotefuran mixture. Sixty C57BL/6 male mice were exposed to either dinotefuran or cetylpyridinium chloride or a mixture of the two via intratracheal instillation (ITI) to examine the synergistic effects of MIE and key events (KEs) within putative AOPs. Various parameters, including clinical and histopathological indicators, changes in body weight and organ weight, inflammatory cell distribution, and inflammatory cytokine expression in the bronchoalveolar lavage fluid (BALF), were analyzed. Additionally, key indicators such as TLR4, NF-κB, TNF-α were investigated to validate the mechanistic aspects of putative AOPs associated with pulmonary fibrosis. We observed significant changes in body weight and neutrophil count, recognized indicators of inflammation, along with inflammatory cell infiltrates, in the group exposed to the mixture of the two biocides. Moreover, increased levels of markers associated with epithelial-mesenchymal transition (EMT) and fibrosis (TNF-α, Acta2, IL-1β, and MMP9), as well as elevated levels of TGF-β, a common downstream signaling factor of TLR4 and PPAR-γ, were identified. Collectively, our findings highlight the potential toxic effects of a mixture of these two biocides in an in vivo model and confirmed the effective function of the putative AOP.
Collapse
Affiliation(s)
- Yoon Cho
- Center for Respiratory Research, Division of Jeonbuk Advanced Bio Research, Korea Institute of Toxicology, 30, Baekhak 1-Gil, Jeongeup-si, Jeonbuk-do, 56212, Republic of Korea
| | - Mi-Kyung Song
- Center for Respiratory Research, Division of Jeonbuk Advanced Bio Research, Korea Institute of Toxicology, 30, Baekhak 1-Gil, Jeongeup-si, Jeonbuk-do, 56212, Republic of Korea
| | - Dong Im Kim
- Center for Respiratory Research, Division of Jeonbuk Advanced Bio Research, Korea Institute of Toxicology, 30, Baekhak 1-Gil, Jeongeup-si, Jeonbuk-do, 56212, Republic of Korea
| | - Min-Seok Kim
- Center for Respiratory Research, Division of Jeonbuk Advanced Bio Research, Korea Institute of Toxicology, 30, Baekhak 1-Gil, Jeongeup-si, Jeonbuk-do, 56212, Republic of Korea
- Department of Human and Environmental Toxicology, University of Science & Technology, Daejeon, 34113, Republic of Korea
| | - Kyuhong Lee
- Center for Respiratory Research, Division of Jeonbuk Advanced Bio Research, Korea Institute of Toxicology, 30, Baekhak 1-Gil, Jeongeup-si, Jeonbuk-do, 56212, Republic of Korea
- Department of Human and Environmental Toxicology, University of Science & Technology, Daejeon, 34113, Republic of Korea
| |
Collapse
|
32
|
Amissah HA, Antwi MH, Amissah TA, Combs SE, Shevtsov M. More than Just Protein Folding: The Epichaperome, Mastermind of the Cancer Cell. Cells 2025; 14:204. [PMID: 39936995 PMCID: PMC11817126 DOI: 10.3390/cells14030204] [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: 01/05/2025] [Revised: 01/26/2025] [Accepted: 01/28/2025] [Indexed: 02/13/2025] Open
Abstract
The epichaperome, a dynamic and integrated network of chaperone proteins, extends its roles beyond basic protein folding to protein stabilization and intracellular signal transduction to orchestrating a multitude of cellular processes critical for tumor survival. In this review, we explore the multifaceted roles of the epichaperome, delving into its diverse cellular locations, factors that modulate its formation and function, its liquid-liquid phase separation, and the key signaling and crosstalk pathways it regulates, including cellular metabolism and intracellular signal transduction. We further highlight techniques for isolating and identifying epichaperome networks, pitfalls, and opportunities. Further, we review the profound implications of the epichaperome for cancer treatment and therapy design, underscoring the need for strategic engineering that hinges on a comprehensive insight into the comprehensive structure and workings of the epichaperome across the heterogeneous cell subpopulations in the tumor milieu. By presenting a holistic view of the epichaperome's functions and mechanisms, we aim to underscore its potential as a key target for novel anti-cancer strategies, revealing that the epichaperome is not merely a piece of protein folding machinery but a mastermind that facilitates the malignant phenotype.
Collapse
Affiliation(s)
- Haneef Ahmed Amissah
- Institute of Life Sciences and Biomedicine, Department of Medical Biology and Biotechnology, School of Medicine and Life Sciences, Far Eastern Federal University, Vladivostok 690922, Russia;
- Diagnostics Laboratory Department, Trauma and Specialist Hospital, Winneba CE-122-2486, Central Region, Ghana
| | - Maxwell Hubert Antwi
- Department of Medical Laboratory Science, Faculty of Health and Allied Sciences, Koforidua Technical University, Koforidua EN-112-3991, Eastern Region, Ghana; (M.H.A.); (T.A.A.)
| | - Tawfeek Ahmed Amissah
- Department of Medical Laboratory Science, Faculty of Health and Allied Sciences, Koforidua Technical University, Koforidua EN-112-3991, Eastern Region, Ghana; (M.H.A.); (T.A.A.)
| | - Stephanie E. Combs
- Department of Radiation Oncology, Technische Universität München (TUM), Klinikum Rechts der Isar, 81675 Munich, Germany;
| | - Maxim Shevtsov
- Department of Radiation Oncology, Technische Universität München (TUM), Klinikum Rechts der Isar, 81675 Munich, Germany;
- Laboratory of Biomedical Nanotechnologies, Institute of Cytology of the Russian Academy of Sciences (RAS), Saint Petersburg 194064, Russia
- Personalized Medicine Centre, Almazov National Medical Research Centre, Saint Petersburg 197341, Russia
| |
Collapse
|
33
|
Muniyan S, Vengoji R, Nimmakayala RK, Seshacharyulu P, Perumalsamy B, Alsafwani ZW, Kakar SS, Smith LM, Shonka N, Teply BA, Lele SM, Ponnusamy MP, Batra SK. PAF1-mediated transcriptional reprogramming confers docetaxel resistance in advanced prostate cancer. Cancer Lett 2025; 609:217355. [PMID: 39603380 PMCID: PMC11912633 DOI: 10.1016/j.canlet.2024.217355] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Revised: 11/08/2024] [Accepted: 11/24/2024] [Indexed: 11/29/2024]
Abstract
Advanced prostate cancer (PCa) remains a significant clinical challenge, and docetaxel plays a significant role in disease management. Despite the efficacy of docetaxel as a first-line chemotherapy, resistance often develops. We developed three clinically relevant in vitro PCa cell models and transcriptomic analysis identified that the Paf1/RNA polymerase II complex component (PAF1)-associated pluripotent-transcription factor (TF), SOX2, plays a crucial role in docetaxel resistance. The cancer stem cell (CSC) transcriptional master regulator PAF1 is significantly higher in PCa cell lines, tumor tissues, and docetaxel resistant (DR) PCa cells than in age-matched control cells. To determine the molecular underlying and functional characteristics of PAF1 in resistance mechanisms, we performed coimmunoprecipitation, embryonic stem cell network proteins, in vitro tumor-initiating ability, and 3D multicellular organoid growth using PAF1 knockdown cells. Tet-inducible PAF1 depletion reduced the drug-efflux phenotype, tumor-initiating frequencies, and three-dimensional organoid growth of the docetaxel-resistant PCa cell lines. Functional studies also showed restoration of docetaxel sensitivity in a 3D tumorsphere model upon PAF1 depletion. PAF1 depletion was also associated with decreased pluripotent TFs and other CSC markers. This study provides a novel regulatory mechanism of docetaxel resistance in PCa through PAF1.
Collapse
Affiliation(s)
- Sakthivel Muniyan
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, 68198, USA.
| | - Raghupathy Vengoji
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Rama Krishna Nimmakayala
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Parthasarathy Seshacharyulu
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Balaji Perumalsamy
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Zahraa Wajih Alsafwani
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Sham S Kakar
- Department of Physiology, University of Louisville School of Medicine, Louisville, KY, 40202, USA
| | - Lynette M Smith
- Department of Biostatistics, College of Public Health, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Nicole Shonka
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, 68198, USA; Department of Internal Medicine, Division of Oncology and Hematology, University of Nebraska Medical Center, Omaha, NE, 68198, USA; Fred and Pamela Buffett Cancer Center, Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Benjamin A Teply
- Department of Internal Medicine, Division of Oncology and Hematology, University of Nebraska Medical Center, Omaha, NE, 68198, USA; Fred and Pamela Buffett Cancer Center, Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Subodh M Lele
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Moorthy P Ponnusamy
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, 68198, USA; Fred and Pamela Buffett Cancer Center, Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Surinder K Batra
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, 68198, USA; Fred and Pamela Buffett Cancer Center, Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, NE, 68198, USA.
| |
Collapse
|
34
|
Shetty SS, Ram Padam KS, Sharma M, Kudva A, Patel P, Radhakrishnan R. Novel transcripts of EMT driving the malignant transformation of oral submucous fibrosis. Sci Rep 2025; 15:3294. [PMID: 39865173 PMCID: PMC11770107 DOI: 10.1038/s41598-025-87790-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Accepted: 01/22/2025] [Indexed: 01/28/2025] Open
Abstract
Oral submucous fibrosis (OSF) is a chronic, progressive, and fibrotic condition of the oral mucosa that carries an elevated risk of malignant transformation. We aimed to identify and validate novel genes associated with the regulation of epithelial-to-mesenchymal transition (EMT) in OSF. Genes regulating EMT were identified through differential gene expression analysis, using a LogFC threshold of -1 and + 1 and a padj value < 0.05, based on data from GEO datasets and the TCGA-HNSC datasets. The curated EMT genes were correlated with functional cancer states and subjected to clustering to identify candidate genes. Integration of bioinformatics and proteomics led to the discovery of the EMT genes MMP9, SPARC, and ITGA5 as novel candidates. Comprehensive pathway and immunohistochemical analyses confirmed their roles in regulating EMT in OSF, oral squamous cell carcinoma (OSCC), and OSF-associated squamous cell carcinoma (OSFSCC). The significant roles of MMP9, SPARC, and ITGA5 in fibrosis and malignancy suggest a novel mechanism in which fibrosis-associated type 2 EMT undergoes transition to type 3 EMT, driving OSF towards malignancy.
Collapse
Affiliation(s)
- Smitha Sammith Shetty
- Department of Oral and Maxillofacial Pathology and Microbiology, Manipal College of Dental Sciences, Manipal Academy of Higher Education, Manipal, 576104, India
| | - Kanaka Sai Ram Padam
- Department of Cell and Molecular Biology, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal, Karnataka, India
| | - Mohit Sharma
- Department of Oral Pathology, SGT Dental College Hospital & Research Institute, Gurugram, 122505, Haryana, India
| | - Adarsh Kudva
- Department of Oral and Maxillofacial Surgery, Manipal College of Dental Sciences, Manipal Academy of Higher Education, Manipal, 576104, India
| | - Pratik Patel
- Sangee Oral Pathology Center, Haripura, Surat, 395003, Gujarat, India
| | - Raghu Radhakrishnan
- Department of Oral and Maxillofacial Pathology and Microbiology, Manipal College of Dental Sciences, Manipal Academy of Higher Education, Manipal, 576104, India.
- Unit of Oral and Maxillofacial Pathology, School of Clinical Dentistry, University of Sheffield, Sheffield, S102TA, UK.
- Unit of Oral and Maxillofacial Pathology, Oman Dental College, P.O Box 835, Muscat, Wattayah 116, Oman.
| |
Collapse
|
35
|
Yoon JH, Kim JK, Eun JW, Ashktorab H, Smoot DT, Nam SW, Park WS. NKX6.3 modulation of mitotic dynamics and genomic stability in gastric carcinogenesis. Cell Commun Signal 2025; 23:35. [PMID: 39833908 PMCID: PMC11748348 DOI: 10.1186/s12964-025-02030-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2024] [Accepted: 01/07/2025] [Indexed: 01/30/2025] Open
Abstract
BACKGROUND Gastric cancer remains a significant global health challenge, characterized by poor prognosis and high mortality rates. Mitotic integrity and genomic stability are crucial in maintaining cellular homeostasis and preventing tumorigenesis. The transcription factor NKX6.3 has emerged as a potential regulator of these processes in gastric epithelial cells, prompting an investigation into its role in gastric cancer development. METHODS We employed a combination of in vitro and in vivo techniques to elucidate the impact of NKX6.3 depletion on mitotic dynamics and genomic stability in gastric epithelial cells. Quantitative real-time PCR and Western blot analyses were conducted to assess the expression of mitosis-related genes and proteins. Flow cytometry was utilized to evaluate cell cycle distribution, while immunofluorescence microscopy enabled the visualization of mitotic abnormalities. Statistical analyses, including Student's t-test and ANOVA, were performed to determine the significance of our findings. RESULTS Our results demonstrate that NKX6.3 depletion leads to significant mitotic defects, characterized by increased chromosome misalignment and lagging chromosomes during anaphase. These abnormalities corresponded with elevated levels of genomic instability markers, indicating compromised genomic integrity. Furthermore, the loss of NKX6.3 resulted in altered expression of key regulatory proteins involved in mitosis and DNA repair pathways, suggesting a mechanistic link between NKX6.3 and the maintenance of genomic stability in gastric epithelial cells. Depletion of NKX6.3 resulted in accelerated cell cycle progression and the formation of abnormal mitotic figures, leading to genomic instability characterized by increased DNA content and structural abnormalities. In both in vitro and xenograft models, the depletion of NKX6.3 significantly upregulated AurkA and TPX2, which correlated with gains in DNA copy number. An inverse relationship was observed between NKX6.3 expression and the levels of AurkA and TPX2 in human gastric cancer tissues. CONCLUSIONS This study highlights the essential role of NKX6.3 in regulating mitotic integrity and genomic stability in gastric carcinogenesis. The findings suggest that targeting NKX6.3 may offer a novel therapeutic strategy for improving treatment outcomes in gastric cancer by restoring mitotic fidelity and genomic stability. TRIAL REGISTRATION This study was not registered.
Collapse
Affiliation(s)
- Jung Hwan Yoon
- Department of Pathology, Functional RNomics Research Center, College of Medicine, The Catholic University of Korea, 222 Banpo-daero, Seocho-gu, Seoul, 06591, South Korea
| | - Jeong-Kyu Kim
- Department of Life Science, Chung-Ang University, Seoul, South Korea
| | - Jung Woo Eun
- Department of Gastroenterology, Ajou University School of Medicine, Suwon, South Korea
| | - Hassan Ashktorab
- Department of Medicine, Howard University, District of Columbia, Washington, 20060, USA
| | - Duane T Smoot
- Department of Medicine, Meharry Medical Center, Nashville, TN, 37208, USA
| | - Suk Woo Nam
- Department of Pathology, Functional RNomics Research Center, College of Medicine, The Catholic University of Korea, 222 Banpo-daero, Seocho-gu, Seoul, 06591, South Korea
| | - Won Sang Park
- Department of Pathology, Functional RNomics Research Center, College of Medicine, The Catholic University of Korea, 222 Banpo-daero, Seocho-gu, Seoul, 06591, South Korea.
| |
Collapse
|
36
|
Sarma M, Chatterjee S. Machine Learning-Based Alzheimer's Disease Stage Diagnosis Utilizing Blood Gene Expression and Clinical Data: A Comparative Investigation. Diagnostics (Basel) 2025; 15:211. [PMID: 39857095 PMCID: PMC11765009 DOI: 10.3390/diagnostics15020211] [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: 12/01/2024] [Revised: 01/03/2025] [Accepted: 01/06/2025] [Indexed: 01/27/2025] Open
Abstract
Background/Objectives: This study presents a comparative analysis of the multistage diagnosis of Alzheimer's disease (AD), including mild cognitive impairment (MCI), utilizing two distinct types of biomarkers: blood gene expression and clinical biomarker samples. Both of these samples, obtained from participants in the Alzheimer's Disease Neuroimaging Initiative (ADNI), were independently analyzed utilizing machine learning (ML)-based multiclassifiers. This study applied novel machine learning-based data augmentation techniques to gene expression profile data that are high-dimensional, low-sample-size (HDLSS) and inherently highly imbalanced. The investigation obtained the highest multiclassification performance to date in the multistage diagnosis of Alzheimer's disease utilizing the blood gene expression profiles of Alzheimer's Disease Neuroimaging Initiative (ADNI) participants. Based on the performance results obtained, and other factors such as early prediction capabilities, this study compares the efficacies of the two types of biomarkers for multistage diagnosis. This study presents the sole investigation in which multiclassification-based AD stage diagnosis was conducted utilizing blood gene expression data. We obtained the best multiclassification result in both modalities of the ADNI data in terms of F1-score and were able to identify new genetic biomarkers. Methods: The combination of the XGBoost and SFBS (Sequential Floating Backward Selection) methods was used to select the features. We were able to select the 95 most effective gene probe sets out of 49,386. For the clinical study data, eight of the most effective biomarkers were selected using SFBS. A deep learning (DL) classifier was used to identify the stages-cognitive normal (CN), mild cognitive impairment (MCI), and Alzheimer's disease (AD)/dementia. DL, support vector machine (SVM), gradient boosting (GB), and random forest (RF) classifiers were used for the AD stage detection from gene expression profile data. Because of the high data imbalance in genomic data, borderline oversampling/data augmentation was applied in the model training and original samples for validation. Results: Utilizing clinical data, the highest ROC AUC scores attained were 0.989, 0.927, and 0.907 for the identification of the CN, MCI, and dementia stages, respectively. The highest F1 scores achieved were 0.971, 0.939, and 0.886. Employing gene expression data, we obtained ROC AUC scores of 0.763, 0.761, and 0.706 for the CN, MCI, and dementia stages, respectively, and F1 scores of 0.71, 0.77, and 0.53 for CN, MCI, and dementia, respectively. Conclusions: This represents the best outcome to date for AD stage diagnosis from ADNI blood gene expression profile data utilizing multiclassification techniques. The results indicated that our multiclassification model effectively manages the imbalanced data of a high-dimension, low-sample-size (HDLSS) nature to identify samples of the minority class. MAPK14, PLG, FZD2, FXYD6, and TEP1 are among the novel genes identified as being associated with AD risk.
Collapse
Affiliation(s)
- Manash Sarma
- Department of Computer Science and Engineering, Faculty of Engineering and Technology, Technology Campus (Peenya Campus), Ramaiah University of Applied Sciences, Bengaluru 560058, India
| | | |
Collapse
|
37
|
Panda VK, Mishra B, Mahapatra S, Swain B, Malhotra D, Saha S, Khanra S, Mishra P, Majhi S, Kumari K, Nath AN, Saha S, Jena S, Kundu GC. Molecular Insights on Signaling Cascades in Breast Cancer: A Comprehensive Review. Cancers (Basel) 2025; 17:234. [PMID: 39858015 PMCID: PMC11763662 DOI: 10.3390/cancers17020234] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2024] [Revised: 12/27/2024] [Accepted: 01/01/2025] [Indexed: 01/27/2025] Open
Abstract
The complex signaling network within the breast tumor microenvironment is crucial for its growth, metastasis, angiogenesis, therapy escape, stem cell maintenance, and immunomodulation. An array of secretory factors and their receptors activate downstream signaling cascades regulating breast cancer progression and metastasis. Among various signaling pathways, the EGFR, ER, Notch, and Hedgehog signaling pathways have recently been identified as crucial in terms of breast cancer proliferation, survival, differentiation, maintenance of CSCs, and therapy failure. These receptors mediate various downstream signaling pathways such as MAPK, including MEK/ERK signaling pathways that promote common pro-oncogenic signaling, whereas dysregulation of PI3K/Akt, Wnt/β-catenin, and JAK/STAT activates key oncogenic events such as drug resistance, CSC enrichment, and metabolic reprogramming. Additionally, these cascades orchestrate an intricate interplay between stromal cells, immune cells, and tumor cells. Metabolic reprogramming and adaptations contribute to aggressive breast cancer and are unresponsive to therapy. Herein, recent insights into the novel signaling pathways operating within the breast TME that aid in their advancement are emphasized and current developments in practices targeting the breast TME to enhance treatment efficacy are reviewed.
Collapse
Affiliation(s)
- Venketesh K. Panda
- School of Biotechnology, KIIT Deemed to Be University, Bhubaneswar 751024, India; (V.K.P.); (B.M.); (S.M.); (B.S.); (D.M.); (S.S.); (S.K.); (P.M.); (S.M.); (K.K.); (A.N.N.); (S.S.); (S.J.)
- School of Applied Sciences, KIIT Deemed to Be University, Bhubaneswar 751024, India
| | - Barnalee Mishra
- School of Biotechnology, KIIT Deemed to Be University, Bhubaneswar 751024, India; (V.K.P.); (B.M.); (S.M.); (B.S.); (D.M.); (S.S.); (S.K.); (P.M.); (S.M.); (K.K.); (A.N.N.); (S.S.); (S.J.)
| | - Samikshya Mahapatra
- School of Biotechnology, KIIT Deemed to Be University, Bhubaneswar 751024, India; (V.K.P.); (B.M.); (S.M.); (B.S.); (D.M.); (S.S.); (S.K.); (P.M.); (S.M.); (K.K.); (A.N.N.); (S.S.); (S.J.)
| | - Biswajit Swain
- School of Biotechnology, KIIT Deemed to Be University, Bhubaneswar 751024, India; (V.K.P.); (B.M.); (S.M.); (B.S.); (D.M.); (S.S.); (S.K.); (P.M.); (S.M.); (K.K.); (A.N.N.); (S.S.); (S.J.)
| | - Diksha Malhotra
- School of Biotechnology, KIIT Deemed to Be University, Bhubaneswar 751024, India; (V.K.P.); (B.M.); (S.M.); (B.S.); (D.M.); (S.S.); (S.K.); (P.M.); (S.M.); (K.K.); (A.N.N.); (S.S.); (S.J.)
| | - Suryendu Saha
- School of Biotechnology, KIIT Deemed to Be University, Bhubaneswar 751024, India; (V.K.P.); (B.M.); (S.M.); (B.S.); (D.M.); (S.S.); (S.K.); (P.M.); (S.M.); (K.K.); (A.N.N.); (S.S.); (S.J.)
| | - Sinjan Khanra
- School of Biotechnology, KIIT Deemed to Be University, Bhubaneswar 751024, India; (V.K.P.); (B.M.); (S.M.); (B.S.); (D.M.); (S.S.); (S.K.); (P.M.); (S.M.); (K.K.); (A.N.N.); (S.S.); (S.J.)
| | - Priyanka Mishra
- School of Biotechnology, KIIT Deemed to Be University, Bhubaneswar 751024, India; (V.K.P.); (B.M.); (S.M.); (B.S.); (D.M.); (S.S.); (S.K.); (P.M.); (S.M.); (K.K.); (A.N.N.); (S.S.); (S.J.)
| | - Sambhunath Majhi
- School of Biotechnology, KIIT Deemed to Be University, Bhubaneswar 751024, India; (V.K.P.); (B.M.); (S.M.); (B.S.); (D.M.); (S.S.); (S.K.); (P.M.); (S.M.); (K.K.); (A.N.N.); (S.S.); (S.J.)
| | - Kavita Kumari
- School of Biotechnology, KIIT Deemed to Be University, Bhubaneswar 751024, India; (V.K.P.); (B.M.); (S.M.); (B.S.); (D.M.); (S.S.); (S.K.); (P.M.); (S.M.); (K.K.); (A.N.N.); (S.S.); (S.J.)
| | - Angitha N. Nath
- School of Biotechnology, KIIT Deemed to Be University, Bhubaneswar 751024, India; (V.K.P.); (B.M.); (S.M.); (B.S.); (D.M.); (S.S.); (S.K.); (P.M.); (S.M.); (K.K.); (A.N.N.); (S.S.); (S.J.)
| | - Swarnali Saha
- School of Biotechnology, KIIT Deemed to Be University, Bhubaneswar 751024, India; (V.K.P.); (B.M.); (S.M.); (B.S.); (D.M.); (S.S.); (S.K.); (P.M.); (S.M.); (K.K.); (A.N.N.); (S.S.); (S.J.)
| | - Sarmistha Jena
- School of Biotechnology, KIIT Deemed to Be University, Bhubaneswar 751024, India; (V.K.P.); (B.M.); (S.M.); (B.S.); (D.M.); (S.S.); (S.K.); (P.M.); (S.M.); (K.K.); (A.N.N.); (S.S.); (S.J.)
| | - Gopal C. Kundu
- School of Biotechnology, KIIT Deemed to Be University, Bhubaneswar 751024, India; (V.K.P.); (B.M.); (S.M.); (B.S.); (D.M.); (S.S.); (S.K.); (P.M.); (S.M.); (K.K.); (A.N.N.); (S.S.); (S.J.)
- School of Applied Sciences, KIIT Deemed to Be University, Bhubaneswar 751024, India
- Kalinga Institute of Medical Sciences (KIMS), KIIT Deemed to Be University, Bhubaneswar 751024, India
| |
Collapse
|
38
|
Filippi A, Deculescu-Ioniță T, Hudiță A, Baldasici O, Gălățeanu B, Mocanu MM. Molecular Mechanisms of Dietary Compounds in Cancer Stem Cells from Solid Tumors: Insights into Colorectal, Breast, and Prostate Cancer. Int J Mol Sci 2025; 26:631. [PMID: 39859345 PMCID: PMC11766403 DOI: 10.3390/ijms26020631] [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: 11/21/2024] [Revised: 01/10/2025] [Accepted: 01/11/2025] [Indexed: 01/27/2025] Open
Abstract
Cancer stem cells (CSC) are known to be the main source of tumor relapse, metastasis, or multidrug resistance and the mechanisms to counteract or eradicate them and their activity remain elusive. There are different hypotheses that claim that the origin of CSC might be in regular stem cells (SC) and, due to accumulation of mutations, these normal cells become malignant, or the source of CSC might be in any malignant cell that, under certain environmental circumstances, acquires all the qualities to become CSC. Multiple studies indicate that lifestyle and diet might represent a source of wellbeing that can prevent and ameliorate the malignant phenotype of CSC. In this review, after a brief introduction to SC and CSC, we analyze the effects of phenolic and non-phenolic dietary compounds and we highlight the molecular mechanisms that are shown to link diets to CSC activation in colon, breast, and prostate cancer. We focus the analysis on specific markers such as sphere formation, CD surface markers, epithelial-mesenchymal transition (EMT), Oct4, Nanog, Sox2, and aldehyde dehydrogenase 1 (ALDH1) and on the major signaling pathways such as PI3K/Akt/mTOR, NF-κB, Notch, Hedgehog, and Wnt/β-catenin in CSC. In conclusion, a better understanding of how bioactive compounds in our diets influence the dynamics of CSC can raise valuable awareness towards reducing cancer risk.
Collapse
Affiliation(s)
- Alexandru Filippi
- Department of Biochemistry and Biophysics, “Carol Davila” University of Medicine and Pharmacy of Bucharest, 050474 Bucharest, Romania;
| | - Teodora Deculescu-Ioniță
- Department of Pharmacognosy, Phytochemistry and Phytotherapy, “Carol Davila” University of Medicine and Pharmacy of Bucharest, 050474 Bucharest, Romania;
| | - Ariana Hudiță
- Department of Biochemistry and Molecular Biology, University of Bucharest, 050095 Bucharest, Romania; (A.H.); (B.G.)
| | - Oana Baldasici
- Department of Genetics, Genomics and Experimental Pathology, The Oncology Institute “Prof. Dr. Ion Chiricuță”, 400015 Cluj-Napoca, Romania;
| | - Bianca Gălățeanu
- Department of Biochemistry and Molecular Biology, University of Bucharest, 050095 Bucharest, Romania; (A.H.); (B.G.)
| | - Maria-Magdalena Mocanu
- Department of Biochemistry and Biophysics, “Carol Davila” University of Medicine and Pharmacy of Bucharest, 050474 Bucharest, Romania;
| |
Collapse
|
39
|
Nosrati H, Heydari M. Titanium dioxide nanoparticles: a promising candidate for wound healing applications. BURNS & TRAUMA 2025; 13:tkae069. [PMID: 39759542 PMCID: PMC11697110 DOI: 10.1093/burnst/tkae069] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/28/2024] [Revised: 10/16/2024] [Indexed: 01/07/2025]
Abstract
Effective wound management and treatment are crucial in clinical practice, yet existing strategies often fall short in fully addressing the complexities of skin wound healing. Recent advancements in tissue engineering have introduced innovative approaches, particularly through the use of nanobiomaterials, to enhance the healing process. In this context, titanium dioxide nanoparticles (TiO2 NPs) have garnered attention due to their excellent biological properties, including antioxidant, anti-inflammatory, and antimicrobial properties. Furthermore, these nanoparticles can be modified to enhance their therapeutic benefits. Scaffolds and dressings containing TiO2 NPs have demonstrated promising outcomes in accelerating wound healing and enhancing tissue regeneration. This review paper covers the wound healing process, the biological properties of TiO2 NPs that make them suitable for promoting wound healing, methods for synthesizing TiO2 NPs, the use of scaffolds and dressings containing TiO2 NPs in wound healing, the application of modified TiO2 NPs in wound healing, and the potential toxicity of TiO2 NPs.
Collapse
Affiliation(s)
- Hamed Nosrati
- Department of Biology, Faculty of Science, Razi University, Kermanshah, Iran
| | - Morteza Heydari
- Research Group of Immune Cell Communication, Department of Immune Medicine, Universitätsklinikum Regensburg | UKR, Franz-Josef-Strauss-Allee 11, 93053, Regensburg, Germany
| |
Collapse
|
40
|
Shanavas S, Sen U, Banerjee R, Shenoy P S, Bose B. Effective Targeting of Colorectal Cancer Stem Cells by Inducing Differentiation Mediated by Low-Dose Vitamin C via β-Catenin Retention in the Cell Membrane. J Cell Biochem 2025; 126:e30686. [PMID: 39660395 DOI: 10.1002/jcb.30686] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2024] [Revised: 11/06/2024] [Accepted: 11/20/2024] [Indexed: 12/12/2024]
Abstract
Cancer stem cells (CSCs) are implicated as the underlying cause of tumor recurrence due to their refractoriness to conventional therapies. Targeting CSCs through novel approaches can hinder their survival and proliferation, potentially reducing the challenges associated with tumor relapse. Our previous study demonstrated that colorectal cancer stem cells (CR-CSCs) showed sensitivity to Vitamin C (Vit C), displaying a dose-responsive effect where low doses (2-10 µM) promoted cell proliferation while high doses induced cell death. In this study, we unraveled the mechanistic effects of low doses that, although induced proliferation, remarkably facilitated stemness reduction in HT-29 cell line-derived CR-CSCs. Our findings revealed that Vit C doses of 2 and 6 µM resulted in a reduction in stemness as evidenced by a reduced CD44+ cell population, representing CR-CSCs. The key finding was the remarkable increase in the expression of β-catenin protein following low-dose Vit C treatment, despite a reduction in stemness, accompanied by a mesenchymal to epithelial transition (MET). The sequestration of upregulated β-catenin via E-cadherin to the cell membrane was identified as a mechanism for reduced stemness, MET, and differentiation of CR-CSCs. Importantly, the epithelial phenotype induced by low-dose Vit C rendered CR-CSCs sensitive to conventional treatments, enhancing chemosensitivity to Cisplatin, Paclitaxel, and 5-Fluorouracil by 60%-90%. These findings suggest that low dose Vit C could serve as an adjuvant to conventional therapeutic strategies for targeting advanced colorectal cancer by sensitizing CR-CSCs to chemotherapy and potentially reducing tumor recurrence.
Collapse
Affiliation(s)
- Shanooja Shanavas
- Stem Cells and Regenerative Medicine Centre, Yenepoya Research Centre, Yenepoya (Deemed to be University), Mangalore, Karnataka, India
| | - Utsav Sen
- Stem Cells and Regenerative Medicine Centre, Yenepoya Research Centre, Yenepoya (Deemed to be University), Mangalore, Karnataka, India
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Rajkumar Banerjee
- Oils, Lipid Science & Technology Division, CSIR-Indian Institute of Chemical Technology, Hyderabad, India
| | - Sudheer Shenoy P
- Stem Cells and Regenerative Medicine Centre, Yenepoya Research Centre, Yenepoya (Deemed to be University), Mangalore, Karnataka, India
| | - Bipasha Bose
- Stem Cells and Regenerative Medicine Centre, Yenepoya Research Centre, Yenepoya (Deemed to be University), Mangalore, Karnataka, India
| |
Collapse
|
41
|
Cho HJ, Jung HJ. Cyclophilin A knockdown inhibits the proliferation and metastatic ability of AGS gastric cancer stem cells by downregulating CD147/STAT3/AKT/ERK and epithelial‑mesenchymal transition. Mol Med Rep 2025; 31:14. [PMID: 39513611 PMCID: PMC11551680 DOI: 10.3892/mmr.2024.13379] [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: 09/06/2024] [Accepted: 10/17/2024] [Indexed: 11/15/2024] Open
Abstract
Gastric cancer stem cells (GCSCs) contribute to the challenging aspects of gastric cancer, such as progression, metastasis, treatment resistance and recurrence. Inhibitors targeting cyclophilin A (CypA) have shown potential in curtailing GCSC growth. Building upon this, the current study delved deeper into understanding the functional role of CypA in controlling the proliferation and metastatic capabilities of GCSCs, employing CypA‑specific small interfering RNA. The results revealed that knockdown of CypA led to significant suppression of the growth and tumorsphere‑forming capacity of GCSCs derived from AGS cells. This effect was mediated by arresting the cell cycle at the G0/G1 and S phases, and promoting apoptosis. Furthermore, silencing of CypA exerted inhibitory effects on the migration and invasion of AGS GCSCs by modulating the process of epithelial‑mesenchymal transition. Notably, the observed antiproliferative and antimetastatic effects of CypA knockdown were associated with the downregulation of critical regulators of gastric cancer stemness, such as CD44, CD133, aldehyde dehydrogenase 1 family member A1, NANOG, OCT4 and SOX2. This regulation occurred through inactivation of the CD147/STAT3/AKT/ERK signaling pathway. Additionally, CypA knockdown effectively curbed in vivo tumor growth of AGS GCSCs in a chorioallantoic membrane assay using chick embryos. These findings underscore the critical role of CypA in promoting the proliferation and metastasis of GCSCs, highlighting its potential as an effective therapeutic target for eradicating GCSCs and improving gastric cancer treatment outcomes.
Collapse
Affiliation(s)
- Hee Jeong Cho
- Department of Life Science and Biochemical Engineering, Graduate School, Sun Moon University, Asan, Chungcheongnam 31460, Republic of Korea
| | - Hye Jin Jung
- Department of Life Science and Biochemical Engineering, Graduate School, Sun Moon University, Asan, Chungcheongnam 31460, Republic of Korea
- Department of Pharmaceutical Engineering and Biotechnology, Sun Moon University, Asan, Chungcheongnam 31460, Republic of Korea
| |
Collapse
|
42
|
Kim H, Jung SH, Jo S, Han JW, Yoon M, Lee JH. Anti‑angiogenic effect of Bryopsis plumosa‑derived peptide via aquaporin 3 in non‑small cell lung cancer. Int J Oncol 2025; 66:5. [PMID: 39611488 PMCID: PMC11637497 DOI: 10.3892/ijo.2024.5711] [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: 06/10/2024] [Accepted: 10/30/2024] [Indexed: 11/30/2024] Open
Abstract
Developing novel anti‑angiogenic agents with minimal toxicity is notably challenging for cancer therapeutics. The discovery and development of peptides, whether derived from natural sources or synthesized, has potential for developing anti‑angiogenic agents characterized by their ability to penetrate cancer cells, high specificity and low toxicity. The present study identified a Bryopsis plumose‑derived anticancer and anti‑angiogenesis marine‑derived peptide 06 (MP06). A 22‑amino acid peptide was synthesized and conjugated with fluorescein isothiocyanate (FITC‑MP06) for intracellular localization in H1299 non‑small cell lung cancer cells. Regulatory effects of this peptide on the viability, migration and self‑renewal of lung cancer cells was assessed. Furthermore, anti‑angiogenic effect of MP06 was investigated by monitoring vascular tube formation in human umbilical vein endothelial cells and a zebrafish model. Aquaporin (AQP)3, a membrane channel in various tissues, is involved in regulating stemness, epithelial‑mesenchymal transition (EMT) and angiogenesis. MP06 downregulated AQP3 expression. Consistently, AQP3 knockdown by RNA silencing downregulated its gene expression, leading to a decrease in stemness, EMT and angiogenesis properties in H1299 cells. MP06 could thus serve as a novel therapeutic target with anticancer and angiogenesis properties for non‑small cell lung cancer.
Collapse
Affiliation(s)
- Heabin Kim
- Department of Bio-material Research, National Marine Biodiversity Institute of Korea, Seocheon 33662, Republic of Korea
| | - Seung-Hyun Jung
- Department of Bio-material Research, National Marine Biodiversity Institute of Korea, Seocheon 33662, Republic of Korea
| | - Seonmi Jo
- Department of Biological Application & Technology, National Marine Biodiversity Institute of Korea, Seocheon 33662, Republic of Korea
| | - Jong Won Han
- Department of Ecology & Conservation, National Marine Biodiversity Institute of Korea, Seocheon 33662, Republic of Korea
| | - Moongeun Yoon
- Department of Bio-material Research, National Marine Biodiversity Institute of Korea, Seocheon 33662, Republic of Korea
| | - Jei Ha Lee
- Department of Bio-material Research, National Marine Biodiversity Institute of Korea, Seocheon 33662, Republic of Korea
| |
Collapse
|
43
|
Zu F, Chen C, Geng Q, Li H, Chan B, Luo G, Wu M, Ilmer M, Renz BW, Bentum-Ennin L, Gu H, Sheng W. Smad2 Cooperating with TGIF2 Contributes to EMT and Cancer Stem Cells Properties in Pancreatic Cancer via Co-Targeting SOX2. Int J Biol Sci 2025; 21:524-543. [PMID: 39781447 PMCID: PMC11705628 DOI: 10.7150/ijbs.102381] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2024] [Accepted: 11/18/2024] [Indexed: 01/12/2025] Open
Abstract
The underlying mechanisms between cancer stem cells (CSC) and epithelial-mesenchymal transition (EMT) in pancreatic cancer (PC) remain unclear. In this study, we identified TGIF2 as a target gene of CSC using sncRNA and machine learning. TGIF2 is closely related to the expression of SOX2, EGFR, and E-cadherin, indicating poor prognosis. Mechanistically, TGIF2 promoted the EMT phenotype and CSC properties following the activation of SOX2, Slug, CD44, and ERGF/MAPK signaling, which were rescued by SOX2 silencing. TGIF2 silencing contributes to the opposite phenotype via SOX2. Notably, Smad2 cooperates with TGIF2 to co-regulate the SOX2 promoter, which in turn promotes EMT and CSC signaling by transactivating Slug and EGFR, respectively. The transactivation of EGFR/MAPK signaling by SOX2 promotes TGIF2 nuclear translocation, forming a positive feedback loop in vitro. Moreover, the interaction of TGIF2 and SOX2 with EGFR inhibitors promoted subcutaneous tumors and liver metastasis in vivo. Thus, the TGIF2/SOX2 axis contributes to CSC, EMT, and chemoresistance, providing a promising target for PC therapy.
Collapse
Affiliation(s)
- Fuqiang Zu
- Department of General Surgery, the Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, China
| | - ChuanPing Chen
- Department of Pharmacy, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230022, China
| | - Qilong Geng
- Department of Clinical Medicine, Anhui Medical University, Hefei, 230022, China
| | - Haoyu Li
- Department of Clinical Medicine, Anhui Medical University, Hefei, 230022, China
| | - Boyuan Chan
- Department of Clinical Medicine, Anhui Medical University, Hefei, 230022, China
| | - Guopei Luo
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
| | - Mengcheng Wu
- Department of General Surgery, the First Affiliated Hospital of Anhui Medical University, Hefei, 230022, China
| | - Matthias Ilmer
- German Cancer Consortium (DKTK), Partner Site Munich and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Bernhard W Renz
- German Cancer Consortium (DKTK), Partner Site Munich and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Lutterodt Bentum-Ennin
- Department of Immunology, School of Basic Medical Sciences, Anhui Medical University, Hefei 230032, China
| | - Hao Gu
- Department of Immunology, School of Basic Medical Sciences, Anhui Medical University, Hefei 230032, China
| | - Weiwei Sheng
- Department of General Surgery, the First Affiliated Hospital of Anhui Medical University, Hefei, 230022, China
| |
Collapse
|
44
|
Park G, Park S, Oh S, Choi N, Choi J. Effects of culture temperature (37°C, 39°C) and oxygen concentration (20%, 2%) on proliferation and differentiation of C2C12 cells. JOURNAL OF ANIMAL SCIENCE AND TECHNOLOGY 2025; 67:224-235. [PMID: 39974777 PMCID: PMC11833204 DOI: 10.5187/jast.2023.e130] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/18/2023] [Revised: 11/08/2023] [Accepted: 11/14/2023] [Indexed: 02/21/2025]
Abstract
Cells, culture media, and so on are important elements of cultured meat production technology. Also, the environment (humidity, temperature, air composition, dissolved oxygen tension, etc.) for in vivo muscle production are important. Among cell culture conditions, culture temperature and oxygen concentration are important physical factors that can affect cells. The objective of this study was to determine effects of culture temperature and oxygen concentration on proliferation and differentiation of muscle cells. This study was conducted using C2C12 cells of rat myoblasts widely used in muscle physiology. The temperature was chosen to induce some thermal stress at 39°C, and the oxygen concentration was selected at 2% to mimic the oxygen levels present in muscle cells in vivo. Culture conditions consisted of CON (37°C/20% O2), T1 (37°C/2% O2), T2 (39°C/20% O2) and T3 (39°C/2% O2). In terms of cell proliferation, temperature conditions had a significant impact (p < 0.05), and a temperature of 39°C was found to reduce the cell count. Oxygen conditions had a significant impact on 3-(4,5-dimethylthiazol-2-yl)-5-(3-carboxymethoxyphenyl)-2-(4-sulfophenyl)-2H-tetrazolium (MTS) absorbance, and temperature conditions were found to have a greater influence on cell proliferation compared to oxygen condition and interaction condition. The temperature conditions were found to have a significant impact (p < 0.05) on the expression of proteins related to myogenesis compared to oxygen conditions. The significant increase (p < 0.05) in the protein expression levels of Myh, Myod1, Myog, and Mb in T2 compared to CON, and in T3 compared to T1, suggests that a temperature of 39°C enhances the expression of myogenic differentiation proteins. These results indicate that temperature conditions have a significant impact (p < 0.05) on cell proliferation and differentiation, more so than oxygen conditions and interaction conditions. And a temperature of 39°C was found to inhibit cell proliferation, but in the case of differentiation, it was observed to be promoted due to the upregulation of myogenic differentiation proteins.
Collapse
Affiliation(s)
- Gyutae Park
- Department of Animal Science, Chungbuk
National University, Cheongju 28644, Korea
| | - Sanghun Park
- Department of Animal Science, Chungbuk
National University, Cheongju 28644, Korea
| | - Sehyuk Oh
- Department of Animal Science, Chungbuk
National University, Cheongju 28644, Korea
| | - Nayoung Choi
- Department of Animal Science, Chungbuk
National University, Cheongju 28644, Korea
| | - Jungseok Choi
- Department of Animal Science, Chungbuk
National University, Cheongju 28644, Korea
| |
Collapse
|
45
|
Prišlin Šimac M, Naletilić Š, Kostanić V, Kunić V, Zorec TM, Poljak M, Vlaj D, Kogoj R, Turk N, Brnić D. Canid alphaherpesvirus 1 infection alters the gene expression and secretome profile of canine adipose-derived mesenchymal stem cells in vitro. Virol J 2024; 21:336. [PMID: 39731173 PMCID: PMC11673362 DOI: 10.1186/s12985-024-02603-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2024] [Accepted: 12/09/2024] [Indexed: 12/29/2024] Open
Abstract
BACKGROUND Canine adipose-derived mesenchymal stem cells (cAD-MSCs) demonstrate promising tissue repair and regeneration capabilities. However, the procurement and preservation of these cells or their secreted factors for therapeutic applications pose a risk of viral contamination, and the consequences for cAD-MSCs remain unexplored. Consequently, this research sought to assess the impact of canid alphaherpesvirus 1 (CHV) on the functional attributes of cAD-MSCs, including gene expression profiles and secretome composition. METHODS To this end, abdominal adipose tissue from 12 healthy dogs was harvested to isolate cAD-MSCs. These samples were tested for CHV contamination before introducing a wild-type CHV strain via serial passages. Following CHV infection, real-time reverse transcription-polymerase chain reaction array and liquid chromatography with tandem mass spectrometry assessments enabled analyses of gene expression and secretome's proteomic profile, respectively. RESULTS This study showed that the initial cAD-MSC populations were devoid of CHV. cAD-MSCs showed susceptibility to infection with wild-type CHV, leading to notable modifications in gene expression and secretome profile. The observed genomic variations in gene expression indicate potential impacts on the stemness, migration, and other functional properties of cAD-MSCs, highlighting the need for further studies to evaluate their functional capacity post-infection. Moreover, gene expression and secretome analyses suggest a shift in stem cell differentiation toward an adipogenic phenotype. CONCLUSION To the best of our knowledge, this is the first study of the effects of virus infection on gene expression and secretome composition in cAD-MSCs. The outcomes of our study underscore the imperative of routine viral screening prior to the therapeutic use of cAD-MSCs. Moreover, these findings provide novel insights into the pathogenic mechanisms of CHV and pave the way for future canine stem cell and virus research.
Collapse
Affiliation(s)
| | - Šimun Naletilić
- Department for Pathological Morphology, Croatian Veterinary Institute, Zagreb, Croatia
| | | | - Valentina Kunić
- Virology Department, Croatian Veterinary Institute, Zagreb, Croatia
| | - Tomaž Mark Zorec
- Institute of Microbiology and Immunology, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| | - Mario Poljak
- Institute of Microbiology and Immunology, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| | - Doroteja Vlaj
- Institute of Microbiology and Immunology, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| | - Rok Kogoj
- Institute of Microbiology and Immunology, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| | - Nenad Turk
- Department of Microbiology and Infectious Diseases With Clinic, Faculty of Veterinary Medicine, University of Zagreb, Zagreb, Croatia
| | - Dragan Brnić
- Virology Department, Croatian Veterinary Institute, Zagreb, Croatia.
| |
Collapse
|
46
|
Stanco D, Lipsa D, Bogni A, Bremer-Hoffmann S, Clerbaux LA. An Adverse Outcome Pathway for food nanomaterial-induced intestinal barrier disruption. FRONTIERS IN TOXICOLOGY 2024; 6:1474397. [PMID: 39776762 PMCID: PMC11703861 DOI: 10.3389/ftox.2024.1474397] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Accepted: 11/19/2024] [Indexed: 01/11/2025] Open
Abstract
Introduction The ingestion of nanomaterials (NMs) may impair the intestinal barrier, but the underlying mechanisms remain evasive, and evidence has not been systematically gathered or produced. A mechanistic-based approach would be instrumental in assessing whether relevant NMs disrupt the intestinal barrier, thereby supporting the NM risk assessment in the food sector. Methods In this study, we developed an adverse outcome pathway (AOP) based on biological plausibility and by leveraging information from an existing NM-relevant AOP that leads to hepatic outcomes. We then extracted the current evidence from the literature for a targeted selection of NMs with high relevance to the food sector, namely, ZnO, CuO, FeO, SiO2, and Ag NMs and nanocellulose. Results We propose a new AOP (AOP 530) that starts with endocytic lysosomal uptake, leading to lysosomal disruption inducing mitochondrial dysfunction. Mitochondrial impairments can lead to cell injury/death and disrupt the intestinal barrier. The evidence collected supports that these food-related NMs can be taken up by intestinal cells and indicates that intestinal barrier disruption may occur due to Ag, CuO, and SiO2 NMs, while only few studies support this outcome for FeO and ZnO. Lysosomal disruption and mitochondrial dysfunction are rarely evaluated. For nanocellulose, none of the studies report toxicity-related events. Conclusion The collection of existing scientific evidence supporting our AOP linking NM uptake to intestinal barrier impairments allowed us to highlight current evidence gaps and data inconsistencies. These inconsistencies could be associated with the variety of stressors, biological systems, and key event (KE)-related assays used in different studies. This underscores the need for further harmonized methodologies and the production of mechanistic evidence for the safety regulatory assessment of NMs in the food sector.
Collapse
Affiliation(s)
- Deborah Stanco
- European Commission, Joint Research Center (JRC), Ispra, Italy
| | | | - Alessia Bogni
- European Commission, Joint Research Center (JRC), Ispra, Italy
| | | | - Laure-Alix Clerbaux
- Institute of Experimental and Clinical Research, UCLouvain, Brussels, Belgium
| |
Collapse
|
47
|
Li W, Gao H, Liu J. Identified VCAM1 as prognostic gene in gastric cancer by co-expression network analysis. Discov Oncol 2024; 15:771. [PMID: 39692880 PMCID: PMC11655750 DOI: 10.1007/s12672-024-01603-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Accepted: 11/19/2024] [Indexed: 12/19/2024] Open
Abstract
The diffuse gastric cancer (DGC) is a malignant tumor distinct from intestinal gastric cancer (IGC). This study aims to identify genetic variances and potential diagnostic and therapeutic approaches for diverse types of gastric cancer utilizing an extensive dataset. Data from RNA sequencing and clinical pathological details were acquired from The Cancer Genome Atlas (TCGA) database and the Gene Expression Omnibus (GEO) dataset. Co-expression gene modules were constructed via Weighted Gene Co-Expression Network Analysis (WGCNA), followed by deciphering gene functions and protein-protein interaction networks within significantly associated modules. In total, analysis was conducted on 56,753 genes from 247 individuals with gastric cancer. Particularly, 621 genes from the green module exhibited strong associations with the Lauren type of gastric cancer. The prominent genes in the green module showed enrichment in processes such as signal transduction, immune response, and the positive regulation of GTPase activity. Noteworthy among these, VCAM1 was identified as the central gene linked to patients' prognosis. Moreover, 72 gastric cancer specimens were collected from The First Affiliated Hospital of University of Science and Technology of China. Immunohistochemical analysis demonstrated a significantly higher expression of VCAM1 in DGC compared to IGC (p = 0.019). Furthermore, it was confirmed that VCAM1 expression serves as a prognostic indicator for patients with DGC (p = 0.002), a correlation not observed in IGC (p = 0.760). In conclusion, this study identifies VCAM1 as a promising diagnostic and prognostic factor, suggesting novel avenues for diagnostic and therapeutic approaches in gastric cancer.
Collapse
Affiliation(s)
- Wenjuan Li
- Department of Oncology, Division of Life Sciences and Medicine, The First Affiliated Hospital of University of Science and Technology of China, University of Science and Technology of China, Hefei, Anhui, China
- Breast Cancer Center, Division of Life Sciences and Medicine,The First Affiliated Hospital of University of Science and Technology of China, University of Science and Technology of China, NO. 107, West 2nd Ring Road, Hefei, Anhui, China
- Department of Oncology, Anhui Provincial Cancer Hospital, Hefei, Anhui, China
| | - Hong Gao
- Breast Cancer Center, Division of Life Sciences and Medicine,The First Affiliated Hospital of University of Science and Technology of China, University of Science and Technology of China, NO. 107, West 2nd Ring Road, Hefei, Anhui, China
| | - Jianjun Liu
- Breast Cancer Center, Division of Life Sciences and Medicine,The First Affiliated Hospital of University of Science and Technology of China, University of Science and Technology of China, NO. 107, West 2nd Ring Road, Hefei, Anhui, China.
- Department of Oncology, Anhui Provincial Cancer Hospital, Hefei, Anhui, China.
| |
Collapse
|
48
|
Donzelli G, Gehring R, Murugadoss S, Roos T, Schaffert A, Linzalone N. A critical review on the toxicological and epidemiological evidence integration for assessing human health risks to environmental chemical exposures. REVIEWS ON ENVIRONMENTAL HEALTH 2024:reveh-2024-0072. [PMID: 39679553 DOI: 10.1515/reveh-2024-0072] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/09/2024] [Accepted: 11/27/2024] [Indexed: 12/17/2024]
Abstract
Toxicology and epidemiology are the two traditional public health scientific disciplines which can contribute to investigate harmful health effects of exposure to toxic substances. Several frameworks for integrating different lines of evidence were proposed since 2011, evolving based of the emergence of new methodologies and approaches. Through the comparison of various theoretical frameworks for evidence integration, we examined similarities, differences, strengths, and weaknesses to provide insights into potential directions for future research. We identified several key challenges of the integration approach to be applied to risk assessment. More specifically, collaboration within a multidisciplinary team of scientists, toxicologists, epidemiologists, and risk assessors, is strongly recommended to be aligned with key regulatory objectives and promote a harmonized approach. Moreover, literature search transparency and systematicity have to be ensured by following validated guidelines, developing parallel protocols for collecting epidemiological and toxicological evidence from various sources, including human, animal, and new approach methodologies (NAMs). Also, the adoption of tailored quality assessment tools is essential to grade the certainty in evidence. Lastly, we recommend the use of the Adverse Outcome Pathway framework to provide a structured understanding of toxicity mechanisms and allow the integration of human, animal, and NAMs data within a single framework.
Collapse
Affiliation(s)
- Gabriele Donzelli
- Institute of Clinical Physiology of the National Research Council (CNR-IFC), Pisa, Italy
| | - Ronette Gehring
- Department of Population Health Sciences, Institute for Risk Assessment Sciences (IRAS), Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands
| | - Sivakumar Murugadoss
- Scientific Direction of Chemical and Physical Health Risks, SCIENSANO, Brussels, Belgium
| | - Tom Roos
- Department of Population Health Sciences, Institute for Risk Assessment Sciences (IRAS), Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands
| | - Alexandra Schaffert
- Institute of Medical Biochemistry, Medical University Innsbruck, Innsbruck, Austria
| | - Nunzia Linzalone
- Institute of Clinical Physiology of the National Research Council (CNR-IFC), Pisa, Italy
| |
Collapse
|
49
|
Thangasparan S, Kamisah Y, Ugusman A, Mohamad Anuar NN, Ibrahim N‘I. Unravelling the Mechanisms of Oxidised Low-Density Lipoprotein in Cardiovascular Health: Current Evidence from In Vitro and In Vivo Studies. Int J Mol Sci 2024; 25:13292. [PMID: 39769058 PMCID: PMC11676878 DOI: 10.3390/ijms252413292] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2024] [Revised: 12/03/2024] [Accepted: 12/04/2024] [Indexed: 01/11/2025] Open
Abstract
Cardiovascular diseases (CVD) are the number one cause of death worldwide, with atherosclerosis, which is the formation of fatty plaques in the arteries, being the most common underlying cause. The activation of inflammatory events and endothelium dysfunction are crucial for the development and pathophysiology of atherosclerosis. Elevated circulating levels of low-density lipoprotein (LDL) have been associated with severity of atherosclerosis. LDL can undergo oxidative modifications, resulting in oxidised LDL (oxLDL). OxLDL has been found to have antigenic potential and contribute significantly to atherosclerosis-associated inflammation by activating innate and adaptive immunity. Various inflammatory stimuli such as interleukin-6 (IL-6), tumour necrosis factor-alpha (TNF-α) and intercellular adhesion molecule 1 (ICAM-1) play major roles in atherosclerosis. To date, studies have provided valuable insights into the role of oxLDL in the development of atherosclerosis. However, there remains a gap in understanding the specific pathways involved in this process. This review aims to provide and discuss the mechanisms by which oxLDL modulates signalling pathways that cause cardiovascular diseases by providing in vitro and in vivo experimental evidence. Its critical role in triggering and sustaining endothelial dysfunction highlights its potential as a therapeutic target. Advancing the understanding of its atherogenic role and associated signalling pathways could pave the way for novel targeted therapeutic strategies to combat atherosclerosis more effectively.
Collapse
Affiliation(s)
- Sahsikala Thangasparan
- Department of Pharmacology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Jalan Yaacob Latif, Bandar Tun Razak, Cheras 56000, Kuala Lumpur, Malaysia; (S.T.); (Y.K.)
| | - Yusof Kamisah
- Department of Pharmacology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Jalan Yaacob Latif, Bandar Tun Razak, Cheras 56000, Kuala Lumpur, Malaysia; (S.T.); (Y.K.)
- Cardiovascular and Pulmonary Research Group, Universiti Kebangsaan Malaysia, Bangi 43600, Selangor, Malaysia; (A.U.); (N.N.M.A.)
| | - Azizah Ugusman
- Cardiovascular and Pulmonary Research Group, Universiti Kebangsaan Malaysia, Bangi 43600, Selangor, Malaysia; (A.U.); (N.N.M.A.)
- Department of Physiology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Jalan Yaacob Latif, Bandar Tun Razak, Cheras 56000, Kuala Lumpur, Malaysia
| | - Nur Najmi Mohamad Anuar
- Cardiovascular and Pulmonary Research Group, Universiti Kebangsaan Malaysia, Bangi 43600, Selangor, Malaysia; (A.U.); (N.N.M.A.)
- Programme of Biomedical Science, Center for Toxicology & Health Risk Studies (CORE), Faculty of Health Sciences, Universiti Kebangsaan Malaysia, Jalan Raja Muda Abdul Aziz, Kuala Lumpur 50300, Malaysia
| | - Nurul ‘Izzah Ibrahim
- Department of Pharmacology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Jalan Yaacob Latif, Bandar Tun Razak, Cheras 56000, Kuala Lumpur, Malaysia; (S.T.); (Y.K.)
- Cardiovascular and Pulmonary Research Group, Universiti Kebangsaan Malaysia, Bangi 43600, Selangor, Malaysia; (A.U.); (N.N.M.A.)
| |
Collapse
|
50
|
Liaghat M, Ferdousmakan S, Mortazavi SH, Yahyazadeh S, Irani A, Banihashemi S, Seyedi Asl FS, Akbari A, Farzam F, Aziziyan F, Bakhtiyari M, Arghavani MJ, Zalpoor H, Nabi-Afjadi M. The impact of epithelial-mesenchymal transition (EMT) induced by metabolic processes and intracellular signaling pathways on chemo-resistance, metastasis, and recurrence in solid tumors. Cell Commun Signal 2024; 22:575. [PMID: 39623377 PMCID: PMC11610171 DOI: 10.1186/s12964-024-01957-4] [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: 02/13/2024] [Accepted: 11/22/2024] [Indexed: 12/06/2024] Open
Abstract
The intricate cellular process, known as the epithelial-mesenchymal transition (EMT), significantly influences solid tumors development. Changes in cell shape, metabolism, and gene expression linked to EMT facilitate tumor cell invasion, metastasis, drug resistance, and recurrence. So, a better understanding of the intricate processes underlying EMT and its role in tumor growth may lead to the development of novel therapeutic approaches for the treatment of solid tumors. This review article focuses on the signals that promote EMT and metabolism, the intracellular signaling pathways leading to EMT, and the network of interactions between EMT and cancer cell metabolism. Furthermore, the functions of EMT in treatment resistance, recurrence, and metastasis of solid cancers are covered. Lastly, treatment approaches that focus on intracellular signaling networks and metabolic alterations brought on by EMT will be discussed.
Collapse
Affiliation(s)
- Mahsa Liaghat
- Department of Medical Laboratory Sciences, Faculty of Medical Sciences, Kazerun Branch, Islamic Azad University, Kazerun, Iran
- Network of Immunity in Infection, Malignancy & Autoimmunity (NIIMA), Universal Scientific Education & Research Network (USERN), Tehran, Iran
| | - Saeid Ferdousmakan
- Department of Pharmacy Practice, Nargund College of Pharmacy, Bangalore, 560085, India
| | | | - Sheida Yahyazadeh
- Department of Immunology, Faculty of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Asrin Irani
- Department of Biology, Faculty of Basic Sciences, University of Guilan, Rasht, Iran
| | - Sara Banihashemi
- Department of Bioscience, School of Science and Technology, Nottingham Trent University, Nottingham, UK
| | | | - Abdullatif Akbari
- Network of Immunity in Infection, Malignancy & Autoimmunity (NIIMA), Universal Scientific Education & Research Network (USERN), Tehran, Iran
- Shiraz Neuroscience Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Farnoosh Farzam
- Department of Biochemistry, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran
| | - Fatemeh Aziziyan
- Network of Immunity in Infection, Malignancy & Autoimmunity (NIIMA), Universal Scientific Education & Research Network (USERN), Tehran, Iran
- Department of Biochemistry, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran
| | - Maryam Bakhtiyari
- Network of Immunity in Infection, Malignancy & Autoimmunity (NIIMA), Universal Scientific Education & Research Network (USERN), Tehran, Iran
- Department of Medical Laboratory Sciences, Faculty of Allied Medicine, Qazvin University of Medical Sciences, Qazvin, Iran
| | - Mohammad Javad Arghavani
- Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Fasa University of Medical Sciences, Fasa, Iran
| | - Hamidreza Zalpoor
- Network of Immunity in Infection, Malignancy & Autoimmunity (NIIMA), Universal Scientific Education & Research Network (USERN), Tehran, Iran.
- Shiraz Neuroscience Research Center, Shiraz University of Medical Sciences, Shiraz, Iran.
| | - Mohsen Nabi-Afjadi
- Department of Biochemistry, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran.
| |
Collapse
|