1
|
Kocak OF, Yaman ME, Eroglu A. An integrated analytical approach for biomarker discovery in esophageal cancer: Combining trace element and oxidative stress profiling with machine learning. J Trace Elem Med Biol 2025; 89:127678. [DOI: 10.1016/j.jtemb.2025.127678] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 06/17/2025]
|
2
|
Bakrania A, Mo Y, Zheng G, Bhat M. RNA nanomedicine in liver diseases. Hepatology 2025; 81:1847-1877. [PMID: 37725757 PMCID: PMC12077345 DOI: 10.1097/hep.0000000000000606] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Accepted: 09/08/2023] [Indexed: 09/21/2023]
Abstract
The remarkable impact of RNA nanomedicine during the COVID-19 pandemic has demonstrated the expansive therapeutic potential of this field in diverse disease contexts. In recent years, RNA nanomedicine targeting the liver has been paradigm-shifting in the management of metabolic diseases such as hyperoxaluria and amyloidosis. RNA nanomedicine has significant potential in the management of liver diseases, where optimal management would benefit from targeted delivery, doses titrated to liver metabolism, and personalized therapy based on the specific site of interest. In this review, we discuss in-depth the different types of RNA and nanocarriers used for liver targeting along with their specific applications in metabolic dysfunction-associated steatotic liver disease, liver fibrosis, and liver cancers. We further highlight the strategies for cell-specific delivery and future perspectives in this field of research with the emergence of small activating RNA, circular RNA, and RNA base editing approaches.
Collapse
Affiliation(s)
- Anita Bakrania
- Department of Medicine, Toronto General Hospital Research Institute, Toronto, Ontario, Canada
- Department of Medicine, Ajmera Transplant Program, University Health Network, Toronto, Ontario, Canada
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| | - Yulin Mo
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
- Institute of Medical Sciences, University of Toronto, Toronto, Ontario, Canada
| | - Gang Zheng
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
- Institute of Medical Sciences, University of Toronto, Toronto, Ontario, Canada
- Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada
| | - Mamatha Bhat
- Department of Medicine, Toronto General Hospital Research Institute, Toronto, Ontario, Canada
- Department of Medicine, Ajmera Transplant Program, University Health Network, Toronto, Ontario, Canada
- Department of Medicine, Division of Gastroenterology, University Health Network and University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
3
|
Koç ÖK. Azo dye-based on-site assay paper kit for rapid and selective naked-eye and smartphone-assisted detection of deferoxamine drug. Talanta 2025; 295:128333. [PMID: 40382865 DOI: 10.1016/j.talanta.2025.128333] [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: 01/30/2025] [Revised: 05/01/2025] [Accepted: 05/13/2025] [Indexed: 05/20/2025]
Abstract
Ferric ion {Fe(III)} is an integral part of many structural and functional components in the body and plays critical roles in physiological and pathological processes. High Fe(III) concentrations damage many systems and organs in the body. The most common method used to prevent excessive accumulation of Fe(III) ions is chelation therapy and the most commonly used drug is deferoxamine (DFO). In this study, an azo dye-based test-paper probe was developed for the colorimetric determination of DFO. The bi-dental azo-dye, named DAZS (diazosalicylaldehyde), was synthesized with a 58 % yield as due to the coupling reaction of 4,4'-diaminobibenzyl and salicylaldehyde. The surface of cellulose-based chromatographic papers immersed in DAZS solution was coated with DAZS and peraped DAZS-coated paper (DAZS-paper) was used as a sensor kit. The structure of DAZS, DAZS-paper, and its interaction mechanism with Fe(III) and DFO were elucidated by FTIR, 1H NMR, 13C NMR, STEM, EDX, and AFM techniques. One arm of DAZS, a symmetrical molecule, interacts with the cellulose-based chromatographic paper, while the other arm remains exposed. This exposed arm interacts with the Fe(III), interrupting the intramolecular charge transfer (ICT) mechanism present in the DAZS molecule, and the color of the DAZS-paper becomes lighter. This interaction, which forms the Fe(III) sensing strategy, is disrupted in the presence of DFO. As a result of its strong chelating feature, DFO forms a chelate complex with Fe(III), enabling the separation of Fe(III) from DAZS. With the separation of the Fe(III), the ICT mechanism is activated again in the free part of DAZS, providing the coloration of DAZS-paper. Although the observed coloration is directly proportional to the concentration of DFO, color analysis was performed with a smartphone via the "colorimeter" application. By analyzing the test paper and solution medium with a smartphone, the detection limits (LOD) of DFO were found to be 95.0 nmol L-1 and 82.0 nmol L-1, respectively. In the selectivity study conducted with various Fe(III) chelating agents, DFO was determined with high selectivity. It was found that common metal cations and anions did not significantly affect the developed probe. The developed method, which was verified by a chromatographic reference method, can also be easily applied to the commercial drug Desferal and fetal bovine serum containing DFO.
Collapse
Affiliation(s)
- Ömer Kaan Koç
- Department of Chemistry, Faculty of Engineering, İstanbul University-Cerrahpaşa, Avcılar, 34320, İstanbul, Türkiye.
| |
Collapse
|
4
|
Di Lorenzo R, Marzetti E, Coelho-Junior HJ, Calvani R, Pesce V, Landi F, Leeuwenburgh C, Picca A. Iron Metabolism and Muscle Aging: Where Ferritinophagy Meets Mitochondrial Quality Control. Cells 2025; 14:672. [PMID: 40358196 PMCID: PMC12072144 DOI: 10.3390/cells14090672] [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: 04/04/2025] [Revised: 04/30/2025] [Accepted: 05/02/2025] [Indexed: 05/15/2025] Open
Abstract
In older adults with reduced physical performance, an increase in the labile iron pool within skeletal muscle is observed. This accumulation is associated with an altered expression of mitochondrial quality control (MQC) markers and increased mitochondrial DNA damage, supporting the hypothesis that impaired MQC contributes to muscle dysfunction during aging. The autophagy-lysosome system plays a critical role in MQC by tagging and engulfing proteins and organelles for degradation in lysosomes. The endolysosomal system is also instrumental in transferrin recycling, which, in turn, regulates cellular iron uptake. In the neuromuscular system, the autophagy-lysosome system supports the structural integrity of neuromuscular junctions, and its dysfunction contributes to muscle atrophy. While MQC was thought to protect against iron-induced cell death, the discovery of ferroptosis, a form of iron-dependent cell death, has highlighted a complex interplay between MQC and iron-inflicted damage. Ferritinophagy, the autophagic degradation of ferritin, if overactivated, can induce ferroptosis. Alternatively, aging may impair ferritinophagy, leading to ferritin accumulation and the release of toxic labile iron under stress, exacerbating oxidative damage and cellular senescence. Physical activity supports muscle health also by preserving mitochondrial quantity and quality and enhancing bioenergetics. However, therapeutic strategies for preventing or reversing physical function decline in aging are still lacking due to the insufficient understanding of the underlying mechanisms. Unveiling how disruptions in iron homeostasis impact muscle quality in older adults may allow for the development of therapeutic strategies targeting iron handling to alleviate age-associated muscle decline.
Collapse
Affiliation(s)
- Rosa Di Lorenzo
- Department of Biosciences, Biotechnologies, and Environment, Università degli Studi di Bari Aldo Moro, Via Edoardo Orabona 4, 70125 Bari, Italy; (R.D.L.); (V.P.)
| | - Emanuele Marzetti
- Department of Geriatrics, Orthopedics and Rheumatology, Università Cattolica del Sacro Cuore, L.go F. Vito 1, 00168 Rome, Italy; (R.C.); (F.L.)
- Fondazione Policlinico Universitario “Agostino Gemelli” IRCCS, L.go A. Gemelli 8, 00168 Rome, Italy; (H.J.C.-J.); (A.P.)
| | - Helio José Coelho-Junior
- Fondazione Policlinico Universitario “Agostino Gemelli” IRCCS, L.go A. Gemelli 8, 00168 Rome, Italy; (H.J.C.-J.); (A.P.)
| | - Riccardo Calvani
- Department of Geriatrics, Orthopedics and Rheumatology, Università Cattolica del Sacro Cuore, L.go F. Vito 1, 00168 Rome, Italy; (R.C.); (F.L.)
- Fondazione Policlinico Universitario “Agostino Gemelli” IRCCS, L.go A. Gemelli 8, 00168 Rome, Italy; (H.J.C.-J.); (A.P.)
| | - Vito Pesce
- Department of Biosciences, Biotechnologies, and Environment, Università degli Studi di Bari Aldo Moro, Via Edoardo Orabona 4, 70125 Bari, Italy; (R.D.L.); (V.P.)
| | - Francesco Landi
- Department of Geriatrics, Orthopedics and Rheumatology, Università Cattolica del Sacro Cuore, L.go F. Vito 1, 00168 Rome, Italy; (R.C.); (F.L.)
- Fondazione Policlinico Universitario “Agostino Gemelli” IRCCS, L.go A. Gemelli 8, 00168 Rome, Italy; (H.J.C.-J.); (A.P.)
| | - Christiaan Leeuwenburgh
- Department of Physiology and Aging, University of Florida, 2004 Mowry Road, Gainesville, FL 32611, USA
| | - Anna Picca
- Fondazione Policlinico Universitario “Agostino Gemelli” IRCCS, L.go A. Gemelli 8, 00168 Rome, Italy; (H.J.C.-J.); (A.P.)
- Department of Medicine and Surgery, LUM University, Str. Statale 100, 70010 Casamassima, Italy
| |
Collapse
|
5
|
Seke M, Stankovic A, Zivkovic M. Capacity of fullerenols to modulate neurodegeneration induced by ferroptosis: Focus on multiple sclerosis. Mult Scler Relat Disord 2025; 97:106378. [PMID: 40088719 DOI: 10.1016/j.msard.2025.106378] [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: 12/27/2023] [Revised: 01/10/2025] [Accepted: 03/05/2025] [Indexed: 03/17/2025]
Abstract
Multiple sclerosis is an inflammatory disease of the central nervous system (CNS), characterized by oligodendrocyte loss and demyelination of axons leading to neurodegeneration and severe neurological disability. Despite the existing drugs that have immunomodulatory effects an adequate therapy that slow down or stop neuronal death has not yet been found. Oxidative stress accompanied by excessive release of iron into the extracellular space, mitochondrial damage and lipid peroxidation are important factors in the controlled cell death named ferroptosis, latterly recognized in MS. As the fullerenols exhibit potent antioxidant activity, recent results imply that they could have protective effects by suppressing ferroptosis. Based on the current knowledge we addressed the main mechanisms of the protective effects of fullerenols in the CNS in relation to ferroptosis. Inhibition of inflammation, iron overload and lipid peroxidation through the signal transduction mechanism of Nuclear Factor Erythroid 2-Related Factor 2 (NRF2), chelation of heavy metals and free radical scavenging using fullerenols are proposed as benefitial strategy preventing MS progression. Current review connects ferroptosis molecular targets and important factors of MS progression, with biomedical properties and mechanisms of fullerenols' actions, to propose new treatment strategies that could be addaptobale in other neurodegenerative diseases.
Collapse
Affiliation(s)
- Mariana Seke
- Laboratory for Radiobiology and Molecular Genetics, ˮVinčaˮ Institute of Nuclear Sciences -National Institute of The Republic of Serbia, University of Belgrade, Mike Petrovica Alasa 12-14, Belgrade 11 000, Serbia
| | - Aleksandra Stankovic
- Laboratory for Radiobiology and Molecular Genetics, ˮVinčaˮ Institute of Nuclear Sciences -National Institute of The Republic of Serbia, University of Belgrade, Mike Petrovica Alasa 12-14, Belgrade 11 000, Serbia
| | - Maja Zivkovic
- Laboratory for Radiobiology and Molecular Genetics, ˮVinčaˮ Institute of Nuclear Sciences -National Institute of The Republic of Serbia, University of Belgrade, Mike Petrovica Alasa 12-14, Belgrade 11 000, Serbia.
| |
Collapse
|
6
|
Al-Omairi M, Alamir SG, Salman BI, El Deeb S, Alrashdi YBA, Al-Harrasi A, Ibrahim AE. Investigating Trace and Macro-element Composition of Herbal and Nutraceutical Dietary Supplements Marketed in Oman: Insights into Safety and Labeling. Biol Trace Elem Res 2025; 203:2911-2923. [PMID: 39143445 DOI: 10.1007/s12011-024-04343-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/09/2024] [Accepted: 08/07/2024] [Indexed: 08/16/2024]
Abstract
With the global increase in the use of dietary supplements to provide nutrients in one's regular diet, these supplements' potential health risks and benefits have become a topic of significant interest. Interestingly, as dietary supplements, the Food and Drug Administration (FDA) in the United States (USA), Europe, and most countries do not require manufacturers and distributors to obtain approval or provide safety assessments before marketing those products. This research explores for the first time 16 heavy, trace and macro-elemental contents, namely, As, Ni, Cd, Pb, Cu, Co, Mn, Cr, Zn, V, Fe, Al, K, Na, Mg, and Ca, within 24 nutraceutical and herbal supplements marketed in Oman. The research is focusing on ensuring their compositions, concentrations, and freedom of toxic elements. ICP-OES was utilized, preceded by a microwave digestion technique to digest the samples in concentrated HNO3 and HCl (3:1, v/v). The method was validated within linear ranges of 0.03-5.00 ppm and 1.0-200.0 ppm for micro- and macro-elements, respectively, with %recoveries ranging from 90 to 104%. The limits of detection ranged from 0.01 to 0.09 and 0.14 to 0.30 ppm, while the limits of quantification ranged from 0.03 to 0.28 and 0.46 to 0.91 ppm for micro- and macro-elements, respectively. The detected levels were compared to online databases for risk assessment. Although As and Cd were not detected in all samples, Pb was found in nine samples, with some exceeding regulated limits of exposure. About 80% of the samples contained Al, of which two samples were susceptible to serious health risks of exceeding exposure limits in their compiled doses. The locally harvested Omani herbal supplements revealed significant amounts of Zn, Mg, Mn, and Cu. The results highlighted the potential risks associated with both dosage compliance and labeling discrepancies.
Collapse
Affiliation(s)
- Mohamed Al-Omairi
- Natural and Medical Sciences Research Center, University of Nizwa, Birkat Al Mauz, Nizwa, 616, Oman
| | - Samy G Alamir
- Natural and Medical Sciences Research Center, University of Nizwa, Birkat Al Mauz, Nizwa, 616, Oman
- Pharmaceutical Analytical Chemistry Department, Faculty of Pharmacy, Ain Shams University, Abassia, Cairo, 11566, Egypt
| | - Baher I Salman
- Pharmaceutical Analytical Chemistry Department, Faculty of Pharmacy, Al-Azhar University, Assiut Branch, Assiut, 71524, Egypt
| | - Sami El Deeb
- Institute of Medicinal and Pharmaceutical Chemistry, Technische Universitaet Braunschweig, 38106, Brunswick, Germany
| | | | - Ahmed Al-Harrasi
- Natural and Medical Sciences Research Center, University of Nizwa, Birkat Al Mauz, Nizwa, 616, Oman.
| | - Adel Ehab Ibrahim
- Natural and Medical Sciences Research Center, University of Nizwa, Birkat Al Mauz, Nizwa, 616, Oman.
| |
Collapse
|
7
|
Ahn SH, Halgren K, Grzesiak G, MacRenaris KW, Sue A, Xie H, Demireva E, O'Halloran TV, Petroff MG. Autoimmune regulator deficiency causes sterile epididymitis and impacts male fertility through disruption of inorganic physiology. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2025:vkaf054. [PMID: 40267393 DOI: 10.1093/jimmun/vkaf054] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Accepted: 02/12/2025] [Indexed: 04/25/2025]
Abstract
Autoimmune regulator (AIRE), a transcriptional regulator expressed by medullary thymic epithelial cells, is required for shaping the self-antigen tolerant T cell receptor repertoire. In humans, AIRE mutations caues autoimmune polyglandular syndrome type 1. Among other symptoms, men with autoimmune polyglandular syndrome type 1 commonly experience testicular insufficiency and infertility, but the mechanisms causing infertility are unknown. Using an Aire-deficient mouse model, we demonstrate that male subfertility is caused by sterile epididymitis characterized by immune cell infiltration and extensive fibrosis. In addition, we reveal that the presence of autoreactive immune cells and inflammation in epididymides of Aire-deficient mice are required for iron deposition in the interstitium, which is brought on by macrophages. We further demonstrate that male subfertility is associated with a decrease in metals zinc, copper, and selenium, which serve as cofactors in several antioxidant enzymes. We also show an increase in DNA damage of epididymal sperm of Aire-/- animals as a key contributing factor to subfertility. The absence of Aire results in autoimmune attack of the epididymis leading to fibrosis, iron deposition, and copper, zinc, and selenium imbalance, ultimately resulting in sperm DNA damage and subfertility. These results highlight the requirement of Aire to promote immune tolerance to the epididymis, and that its disruption causes an imbalance of inorganic elements with resulting consequence on male fertility.
Collapse
Affiliation(s)
- Soo Hyun Ahn
- Department of Pathobiology Diagnostic Investigation, College of Veterinary Medicine, Michigan State University, East Lansing, MI, United States
| | - Katrina Halgren
- Department of Pathobiology Diagnostic Investigation, College of Veterinary Medicine, Michigan State University, East Lansing, MI, United States
- Pathobiology & Diagnostic Investigation, Lyman Briggs College, Michigan State University, East Lansing, MI, United States
| | - Geoffrey Grzesiak
- Department of Pathobiology Diagnostic Investigation, College of Veterinary Medicine, Michigan State University, East Lansing, MI, United States
| | - Keith W MacRenaris
- Elemental Health Institute and Department of Microbiology, Genetics, and Immunology, Michigan State University, East Lansing, MI, United States
- Department of Chemistry, Michigan State University, East Lansing, MI, United States
- Department of Microbiology, Genetics, and Immunology, Michigan State University, East Lansing, MI, United States
| | - Aaron Sue
- Elemental Health Institute and Department of Microbiology, Genetics, and Immunology, Michigan State University, East Lansing, MI, United States
- Department of Chemistry, Michigan State University, East Lansing, MI, United States
- Department of Microbiology, Genetics, and Immunology, Michigan State University, East Lansing, MI, United States
| | - Huirong Xie
- Transgenic and Genome Editing Facility, Institute for Quantitative Health Science and Engineering, Research Technology Support Facility, Michigan State University, East Lansing, MI, United States
| | - Elena Demireva
- Transgenic and Genome Editing Facility, Institute for Quantitative Health Science and Engineering, Research Technology Support Facility, Michigan State University, East Lansing, MI, United States
| | - Thomas V O'Halloran
- Elemental Health Institute and Department of Microbiology, Genetics, and Immunology, Michigan State University, East Lansing, MI, United States
- Department of Chemistry, Michigan State University, East Lansing, MI, United States
- Department of Microbiology, Genetics, and Immunology, Michigan State University, East Lansing, MI, United States
| | - Margaret G Petroff
- Department of Pathobiology Diagnostic Investigation, College of Veterinary Medicine, Michigan State University, East Lansing, MI, United States
- Department of Microbiology, Genetics, and Immunology, Michigan State University, East Lansing, MI, United States
| |
Collapse
|
8
|
Wang N, Zhou D, Xu K, Kou D, Chen C, Li C, Ge J, Chen L, Zeng J, Gao M. Iron Homeostasis-Regulated Adaptive Metabolism of PEGylated Ultrasmall Iron Oxide Nanoparticles. ACS NANO 2025; 19:13381-13398. [PMID: 40135968 DOI: 10.1021/acsnano.5c01399] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/27/2025]
Abstract
Iron oxide nanoparticles have become increasingly significant in the biomedical field due to their exceptional magnetic properties and biocompatibility. However, understanding their in vivo metabolism and transformation is crucial due to the potential biological effects they may induce. This study investigates the metabolic pathways of PEGylated ultrasmall iron oxide nanoparticles (PUSIONPs) in vivo, particularly under varying iron statuses and dosages. Employing a comprehensive analytical approach─including magnetic resonance imaging, elemental analysis, histological assessments, hematological analysis, and Western blot analysis─the biodistribution and transformation of PUSIONPs were mapped. The findings reveal significant differences in the metabolic fate of PUSIONPs between iron-sufficient and iron-deficient conditions, underscoring the pivotal role of iron homeostasis in regulating PUSIONPs biodegradation. In iron-deficient states, degradation and transformation were markedly accelerated, with the released iron rapidly incorporated into hemoglobin. Additionally, the liver and spleen exhibited different PUSIONPs metabolism rates due to their distinct physiological roles: the spleen, primarily responsible for iron recycling, facilitated faster degradation, while the liver, serving as an iron storage organ, showed slower degradation. Under iron deficiency, most degradation products were directed toward hemoglobin synthesis, whereas under normal conditions, the liver gradually metabolized the degradation products, and the spleen retained higher iron levels. Moreover, PUSIONPs degradation demonstrated dose dependency, with higher doses slowing degradation and reducing the utilization rate by the iron-deficient body. Comprehensive safety evaluations confirmed that PUSIONPs exhibit excellent biocompatibility across all doses, with no significant safety concerns. Compared to the clinically used intravenous iron supplement iron sucrose, PUSIONPs also demonstrated superior bioavailability and more effective iron supplementation. These findings provide critical insights into the interaction between iron oxide nanoparticles and iron metabolism, offering a foundation for future research and the broader application of PUSIONPs in biomedical contexts.
Collapse
Affiliation(s)
- Ning Wang
- Center for Molecular Imaging and Nuclear Medicine, State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiological Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou 215123, China
| | - Dandan Zhou
- Center for Molecular Imaging and Nuclear Medicine, State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiological Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou 215123, China
| | - Keyang Xu
- Center for Molecular Imaging and Nuclear Medicine, State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiological Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou 215123, China
| | - Dandan Kou
- Center for Molecular Imaging and Nuclear Medicine, State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiological Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou 215123, China
| | - Can Chen
- Center for Molecular Imaging and Nuclear Medicine, State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiological Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou 215123, China
| | - Cang Li
- Center for Molecular Imaging and Nuclear Medicine, State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiological Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou 215123, China
| | - Jianxian Ge
- Center for Molecular Imaging and Nuclear Medicine, State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiological Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou 215123, China
| | - Lei Chen
- Center for Molecular Imaging and Nuclear Medicine, State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiological Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou 215123, China
| | - Jianfeng Zeng
- Center for Molecular Imaging and Nuclear Medicine, State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiological Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou 215123, China
| | - Mingyuan Gao
- Center for Molecular Imaging and Nuclear Medicine, State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiological Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou 215123, China
- School of life Sciences, Soochow University, Suzhou 215123, China
- The Second Affiliated Hospital of Soochow University, Suzhou 215123, China
| |
Collapse
|
9
|
Yumrukuz Şenel M, Şahin R, Çolak M, Çoban H, Erel F, Sarıoğlu N. Are the serum iron parameters related to the severity of obstructive sleep apnea syndrome? Sleep Breath 2025; 29:132. [PMID: 40095103 DOI: 10.1007/s11325-025-03301-7] [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: 08/30/2024] [Revised: 02/21/2025] [Accepted: 03/10/2025] [Indexed: 03/19/2025]
Abstract
PURPOSE To evaluate the correlation and relationship between iron parameters including serum iron level, iron-binding capacity, ferritin, transferrin saturation, and the severity of OSAS in the patients who underwent polysomnography. METHODS We retrospectively reviewed 209 patients and divided the patients into two groups; AHI ≥ 30 and AHI < 30. The groups were compared using the Mann-Whitney U and the chi-square test. In addition, Spearman's correlation analysis was performed to analyze the correlation between AHI and iron parameters. RESULTS The mean age of the patients was 47.9 ± 13.7 (19-89) years. Of the 209 patients, 40.7% (n = 85) were female and 59.3% (n = 124) were male. Iron and transferrin saturation was significantly lower in the patients with AHI ≥ 30 compared to the patients with AHI < 30. In female patients, there wasn't any correlation between AHI and iron, ferritin, transferrin saturation, and iron-binding capacity. But, there was a significant negative correlation between the AHI and iron (r = -0.292, p = 0.001) and transferrin saturation (r = -0.349, p < 0.001) in male patients. Also, the AHI was significantly positively correlated with iron binding capacity (r = 0.307, p = 0.001) in male patients. CONCLUSION Our results showed that iron levels were lower in severe OSAS. Suggesting that iron levels decrease as a result of oxidative stress and inflammation seen in OSAS, iron parameters may be a good biomarker in OSAS patients.
Collapse
Affiliation(s)
- Merve Yumrukuz Şenel
- Department of Chest Diseases, Faculty of Medicine, Balıkesir University, Balıkesir, Türkiye.
| | - Rabia Şahin
- Department of Chest Diseases, Faculty of Medicine, Balıkesir University, Balıkesir, Türkiye
| | - Mustafa Çolak
- Department of Chest Diseases, Faculty of Medicine, Balıkesir University, Balıkesir, Türkiye
| | - Hikmet Çoban
- Department of Chest Diseases, Faculty of Medicine, Balıkesir University, Balıkesir, Türkiye
| | - Fuat Erel
- Department of Chest Diseases, Faculty of Medicine, Balıkesir University, Balıkesir, Türkiye
| | - Nurhan Sarıoğlu
- Department of Chest Diseases, Faculty of Medicine, Balıkesir University, Balıkesir, Türkiye
| |
Collapse
|
10
|
Hofmann BJ, Aljohani ET, Cicovacki N, Lee I, Warren DT, Sobolewski A, Stringer T, Lord RM. Ferrocenyl β-Diketonate Compounds: Extended Ring Systems for Improved Anticancer Activity. Chembiochem 2025; 26:e202400759. [PMID: 39446339 DOI: 10.1002/cbic.202400759] [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: 09/16/2024] [Revised: 10/19/2024] [Indexed: 11/16/2024]
Abstract
A library of ferrocenyl β-diketonate compounds with varying degrees of aromatic functionality have been synthesized and fully characterized. This includes cyclic voltammetry and the analysis of four new structures by single crystal X-ray diffraction. The compounds cytotoxic potential has been determined by MTT screening against pancreatic carcinoma (MIA PaCa-2), ovarian adenocarcinoma (A2780), breast adenocarcinomas (MDA-MB-231 and MCF-7) and normal epithelial retinal (ARPE-19). The compounds show a general trend, where increasing the number of aromatic rings in the molecule yields an increase in cytotoxicity and follows the trend anthracenyl>naphthyl>phenyl>methyl. The compounds are particularly sensitive to the triple negative cancer cell line MDA-MB-231, and the potential modes of action have been studied by production of reactive oxygen species using fluorescence microscopy and cell morphology using Scanning Electron Microscopy. All assays highlight the ferrocenyl β-diketonate with an anthracenyl substituent to be the lead compound in this library. The decomposition of this compound was also observed within cells, yielding a cytotoxic fluorescent molecule, which has been visualized by confocal microscopy.
Collapse
Affiliation(s)
- Benjamin J Hofmann
- School of Chemistry, Pharmacy and Pharmacology, University of East Anglia, Norwich, Norfolk, NR4 7TJ, UK
- Department of Chemistry, University of Warwick, Coventry, CV4 7SH, UK
| | - Enas T Aljohani
- School of Chemistry, Pharmacy and Pharmacology, University of East Anglia, Norwich, Norfolk, NR4 7TJ, UK
| | - Natalia Cicovacki
- School of Chemistry, Pharmacy and Pharmacology, University of East Anglia, Norwich, Norfolk, NR4 7TJ, UK
| | - Ivan Lee
- School of Chemistry, Pharmacy and Pharmacology, University of East Anglia, Norwich, Norfolk, NR4 7TJ, UK
| | - Derek T Warren
- School of Chemistry, Pharmacy and Pharmacology, University of East Anglia, Norwich, Norfolk, NR4 7TJ, UK
| | - Anastasia Sobolewski
- School of Chemistry, Pharmacy and Pharmacology, University of East Anglia, Norwich, Norfolk, NR4 7TJ, UK
| | - Tameryn Stringer
- School of Chemistry, Pharmacy and Pharmacology, University of East Anglia, Norwich, Norfolk, NR4 7TJ, UK
- School of Chemistry, University of Waikato, Hamilton, 3240, New Zealand
| | - Rianne M Lord
- School of Chemistry, Pharmacy and Pharmacology, University of East Anglia, Norwich, Norfolk, NR4 7TJ, UK
- Department of Chemistry, University of Warwick, Coventry, CV4 7SH, UK
| |
Collapse
|
11
|
Gil J, Kim D, Choi S, Bae ON. Cadmium-induced iron dysregulation contributes to functional impairment in brain endothelial cells via the ferroptosis pathway. Toxicol Appl Pharmacol 2025; 495:117233. [PMID: 39842614 DOI: 10.1016/j.taap.2025.117233] [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: 10/04/2024] [Revised: 01/12/2025] [Accepted: 01/15/2025] [Indexed: 01/24/2025]
Abstract
Cadmium (Cd2+) is a heavy metal that is a major hazardous environmental contaminant, ubiquitously present in the environment. Cd2+ exposure has been closely associated with an increased prevalence and severity of neurological and cardiovascular diseases (CVD). The blood-brain barrier (BBB) plays a crucial role in protecting the brain from external environmental factors. Mitochondria play an important role in maintaining the barrier function of brain endothelial cells by regulating energy metabolism and redox homeostasis. In this study, we aimed to assess the cytotoxic effects of Cd2+ on the integrity and function of brain endothelial cells. After 24 h of exposure, Cd2+ reduced cell survival, tight junction protein expression, and trans-endothelial electrical resistance (TEER) in bEnd.3 cells suggest a potential BBB integrity disruption by Cd2+ exposure. To clarify the underlying mechanism, we further investigated the role of mitochondria in iron overload-mediated cell death following Cd2+ exposure. Cd2+ induced a substantial reduction in mitochondrial basal respiration and ATP production in brain endothelial cells, suggesting mitochondrial dysfunction. In addition, Cd2+ exposure led to impaired autophagy, elevated iron levels, and increased lipid peroxidation, indicating the initiation of ferroptosis, a form of cell death triggered by iron. In summary, our research suggests that Cd2+ exposure can disrupt BBB function by causing mitochondrial dysfunction and disrupting iron homeostasis.
Collapse
Affiliation(s)
- Junkyung Gil
- College of Pharmacy, Institute of Pharmaceutical Science and Technology, Hanyang University ERICA Campus, Ansan, South Korea.
| | - Donghyun Kim
- College of Pharmacy, Institute of Pharmaceutical Science and Technology, Hanyang University ERICA Campus, Ansan, South Korea.
| | - Sungbin Choi
- College of Pharmacy, Institute of Pharmaceutical Science and Technology, Hanyang University ERICA Campus, Ansan, South Korea.
| | - Ok-Nam Bae
- College of Pharmacy, Institute of Pharmaceutical Science and Technology, Hanyang University ERICA Campus, Ansan, South Korea.
| |
Collapse
|
12
|
Leandri R, Buonocore S, Power K. Transferrin Receptor 2 in Canine Testicular Tumors: An Emerging Key Role in Seminomas. Animals (Basel) 2025; 15:264. [PMID: 39858264 PMCID: PMC11758335 DOI: 10.3390/ani15020264] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2024] [Revised: 01/14/2025] [Accepted: 01/16/2025] [Indexed: 01/27/2025] Open
Abstract
Transferrin Receptor 2 (TfR2) is a homolog of Transferrin Receptor 1 (TfR1), involved in regulating intra and extracellular iron levels. Altered iron pathways have been associated with cancer onset and progression; however, their role in canine tumors remains poorly explored. This study investigated TfR2 immunohistochemical expression in non-neoplastic canine testis for the first time and in the most common types of canine testicular tumors: intratubular seminomas (ITSEMs), diffuse seminomas (DSEMs), Leydig cell tumors (LCTs), and Sertoli cell tumors (SCTs). Immunohistochemical analysis revealed a differential pattern of TfR2 expression according to tumor type, with high expression observed in ITSEMs and DSEMs, occasional expression in LCTs, and absence in SCTs. These results suggest that TfR2 may play a relevant role in canine seminoma development. Furthermore, the specific expression of TfR2 in seminomas highlights its potential as a therapeutic target, where its role in iron regulation and possible compensatory mechanisms warrant further investigation.
Collapse
Affiliation(s)
| | | | - Karen Power
- Department of Biology, University of Naples Federico II, 80126 Naples, Italy; (R.L.); (S.B.)
| |
Collapse
|
13
|
Ahn SH, Halgren K, Grzesiak G, MacRenaris KW, Sue A, Xie H, Demireva E, O'Halloran TV, Petroff MG. Autoimmune regulator deficiency causes sterile epididymitis and impacts male fertility through disruption of inorganic physiology. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.11.632558. [PMID: 39868100 PMCID: PMC11761525 DOI: 10.1101/2025.01.11.632558] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/28/2025]
Abstract
Autoimmune regulator (AIRE), a transcription factor expressed by medullary thymic epithelial cells, is required for shaping the self-antigen tolerant T cell receptor repertoire. Humans with mutations in AIRE suffer from Autoimmune Polyglandular Syndrome Type 1 (APS-1). Among many symptoms, men with APS-1 commonly experience testicular insufficiency and infertility, but the mechanisms causing infertility are unknown. Using an Aire -deficient mouse model, we demonstrate that male subfertility is caused by sterile epididymitis characterized by immune cell infiltration and extensive fibrosis. In addition, we reveal that the presence of autoreactive immune cells and inflammation in epididymides of Aire- deficient mice are required for iron (Fe) deposition in the interstitium, which is brought on by macrophages. We further demonstrate that male subfertility is associated with a decrease in metals zinc (Zn), copper (Cu), and selenium (Se) which serve as cofactors in several antioxidant enzymes. We also show increase in DNA damage of epididymal sperm of Aire -/- animals as a key contributing factor to subfertility. The absence of Aire results in autoimmune attack of the epididymis leading to fibrosis, Fe deposition, and Cu, Zn and Se imbalance, ultimately resulting in sperm DNA damage and subfertility. These results highlight the requirement of Aire to promote immune tolerance throughout the epididymis, disruption of which causes an imbalance of inorganic elements with resulting consequence on male fertility. Key points Breakdown of epididymal self-tolerance promotes disruption of inorganic elements. Autoimmunity causes interstitial fibrosis resulting in sperm DNA damage and subfertility. Elevated interstitial iron and macrophages contribute to fibrosis.
Collapse
|
14
|
Ju JJ, Hang LH. Neuroinflammation and iron metabolism after intracerebral hemorrhage: a glial cell perspective. Front Neurol 2025; 15:1510039. [PMID: 39882361 PMCID: PMC11774705 DOI: 10.3389/fneur.2024.1510039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2024] [Accepted: 12/30/2024] [Indexed: 01/31/2025] Open
Abstract
Intracerebral hemorrhage (ICH) is the most common subtype of hemorrhagic stroke causing significant morbidity and mortality. Previously clinical treatments for ICH have largely been based on a single pathophysiological perspective, and there remains a lack of curative interventions. Following the rupture of cerebral blood vessels, blood metabolites activate resident immune cells such as microglia and astrocytes, and infiltrate peripheral immune cells, leading to the release of a series of inflammatory mediators. Degradation of hemoglobin produces large amounts of iron ions, leading to an imbalance of iron homeostasis and the production of large quantities of harmful hydroxyl radicals. Neuroinflammation and dysregulation of brain iron metabolism are both important pathophysiological changes in ICH, and both can exacerbate secondary brain injury. There is an inseparable relationship between brain iron metabolism disorder and activated glial cells after ICH. Glial cells participate in brain iron metabolism through various mechanisms; meanwhile, iron accumulation exacerbates neuroinflammation by activating inflammatory signaling pathways modulating the functions of inflammatory cells, and so on. This review aims to explore neuroinflammation from the perspective of iron metabolism, linking the complex pathophysiological changes, delving into the exploration of treatment approaches for ICH, and offering insights that could enhance clinical management strategies.
Collapse
Affiliation(s)
- Jia-Jun Ju
- Gusu School, Nanjing Medical University, The First People’s Hospital of Kunshan, Kunshan, China
| | - Li-Hua Hang
- Gusu School, Nanjing Medical University, The First People’s Hospital of Kunshan, Kunshan, China
- Kunshan Cancer Pain Prevention and Treatment Key Laboratory, Kunshan, China
| |
Collapse
|
15
|
Diao H, Chen Y, Liu F. Iron-Catalyzed Cross-Dehydrogenative Coupling. Molecules 2025; 30:250. [PMID: 39860120 PMCID: PMC11767816 DOI: 10.3390/molecules30020250] [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: 10/17/2024] [Revised: 12/01/2024] [Accepted: 12/31/2024] [Indexed: 01/27/2025] Open
Abstract
This review highlights significant advances in iron-catalyzed cross-dehydrogenative coupling (CDC), a method pivotal for forming carbon-carbon (C-C) bonds directly from C-H bonds. This technique uses iron-a naturally abundant, inexpensive, and environmentally benign transition metal-as a catalyst to facilitate the coupling of two unfunctionalized C-H bonds. This method stands out for avoiding pre-functionalized substrates, reducing both waste and cost in organic synthesis. The discussion includes a variety of CDC methodologies involving combinations of C(sp3)-H with C(sp3)-H, C(sp3)-H with C(sp2)-H, and C(sp3)-H with C(sp)-H bonds. These methods have been successfully applied in synthesizing complex molecules and pharmaceuticals, highlighting the versatility and efficiency of iron catalysis.
Collapse
Affiliation(s)
- Haiyan Diao
- Department of Chemistry, Fudan University, Shanghai 200438, China
| | - Yujia Chen
- School of Chemistry and Chemical Engineering, Lanzhou Jiaotong University, Lanzhou 730070, China
| | - Feng Liu
- Department of Chemistry, Fudan University, Shanghai 200438, China
| |
Collapse
|
16
|
Freindorf M, Kraka E. A Closer Look at the FeS Heme Bonds in Azotobacter vinelandii Bacterioferritin: QM/MM and Local Mode Analysis. J Comput Chem 2025; 46:e70012. [PMID: 39749917 PMCID: PMC11697534 DOI: 10.1002/jcc.70012] [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: 09/29/2024] [Revised: 12/04/2024] [Accepted: 12/05/2024] [Indexed: 01/04/2025]
Abstract
Using the QM/MM methodology and a local mode analysis, we investigated a character and a strength of FeS bonds of heme groups in oxidized and reduced forms of Bacterioferritin from Azotobacter vinelandii. The strength of the FeS bonds was correlated with a bond length, an energy density at a bond critical point, and a charge difference of the F and S atoms. Changing the oxidation state from ferrous to ferric generally makes the FeS bonds weaker, longer, more covalent, and more polar. We also investigated the SFeS bond bending and found that the stronger FeS bond, generally makes the SFeS bond bending stiffer, which could play a key role in the balance between ferric and ferrous oxidation states and related biological activities.
Collapse
Affiliation(s)
- Marek Freindorf
- Chemistry DepartmentSouthern Methodist UniversityDallasTexasUSA
| | - Elfi Kraka
- Chemistry DepartmentSouthern Methodist UniversityDallasTexasUSA
| |
Collapse
|
17
|
Limón I, Bedmar J, Fernández-Hernán JP, Multigner M, Torres B, Rams J, Cifuentes SC. A Review of Additive Manufacturing of Biodegradable Fe and Zn Alloys for Medical Implants Using Laser Powder Bed Fusion (LPBF). MATERIALS (BASEL, SWITZERLAND) 2024; 17:6220. [PMID: 39769819 PMCID: PMC11677746 DOI: 10.3390/ma17246220] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/30/2024] [Revised: 12/14/2024] [Accepted: 12/16/2024] [Indexed: 01/11/2025]
Abstract
This review explores the advancements in additive manufacturing (AM) of biodegradable iron (Fe) and zinc (Zn) alloys, focusing on their potential for medical implants, particularly in vascular and bone applications. Fe alloys are noted for their superior mechanical properties and biocompatibility but exhibit a slow corrosion rate, limiting their biodegradability. Strategies such as alloying with manganese (Mn) and optimizing microstructure via laser powder bed fusion (LPBF) have been employed to increase Fe's corrosion rate and mechanical performance. Zn alloys, characterized by moderate biodegradation rates and biocompatible corrosion products, address the limitations of Fe, though their mechanical properties require improvement through alloying and microstructural refinement. LPBF has enabled the fabrication of dense and porous structures for both materials, with energy density optimization playing a critical role in achieving defect-free parts. Fe alloys exhibit higher strength and hardness, while Zn alloys offer better corrosion control and biocompatibility. In vitro and in vivo studies demonstrate promising outcomes for both materials, with Fe alloys excelling in load-bearing applications and Zn alloys in controlled degradation and vascular applications. Despite these advancements, challenges such as localized corrosion, cytotoxicity, and long-term performance require further investigation to fully harness the potential of AM-fabricated Fe and Zn biodegradable implants.
Collapse
Affiliation(s)
- Irene Limón
- Department of Applied Mathematics, Materials Science and Engineering and Electronic Technology, Escuela Superior de Ciencias Experimentales y Tecnología (ESCET), Universidad Rey Juan Carlos, C/Tulipán s/n, 28933 Móstoles, Spain; (I.L.); (J.B.); (J.P.F.-H.); (M.M.); (B.T.); (J.R.)
| | - Javier Bedmar
- Department of Applied Mathematics, Materials Science and Engineering and Electronic Technology, Escuela Superior de Ciencias Experimentales y Tecnología (ESCET), Universidad Rey Juan Carlos, C/Tulipán s/n, 28933 Móstoles, Spain; (I.L.); (J.B.); (J.P.F.-H.); (M.M.); (B.T.); (J.R.)
| | - Juan Pablo Fernández-Hernán
- Department of Applied Mathematics, Materials Science and Engineering and Electronic Technology, Escuela Superior de Ciencias Experimentales y Tecnología (ESCET), Universidad Rey Juan Carlos, C/Tulipán s/n, 28933 Móstoles, Spain; (I.L.); (J.B.); (J.P.F.-H.); (M.M.); (B.T.); (J.R.)
| | - Marta Multigner
- Department of Applied Mathematics, Materials Science and Engineering and Electronic Technology, Escuela Superior de Ciencias Experimentales y Tecnología (ESCET), Universidad Rey Juan Carlos, C/Tulipán s/n, 28933 Móstoles, Spain; (I.L.); (J.B.); (J.P.F.-H.); (M.M.); (B.T.); (J.R.)
| | - Belén Torres
- Department of Applied Mathematics, Materials Science and Engineering and Electronic Technology, Escuela Superior de Ciencias Experimentales y Tecnología (ESCET), Universidad Rey Juan Carlos, C/Tulipán s/n, 28933 Móstoles, Spain; (I.L.); (J.B.); (J.P.F.-H.); (M.M.); (B.T.); (J.R.)
- Institute of Research on Technologies for Sustainability (ITPS), Universidad Rey Juan Carlos, C/Tulipán s/n, 28933 Móstoles, Spain
| | - Joaquín Rams
- Department of Applied Mathematics, Materials Science and Engineering and Electronic Technology, Escuela Superior de Ciencias Experimentales y Tecnología (ESCET), Universidad Rey Juan Carlos, C/Tulipán s/n, 28933 Móstoles, Spain; (I.L.); (J.B.); (J.P.F.-H.); (M.M.); (B.T.); (J.R.)
- Institute of Research on Technologies for Sustainability (ITPS), Universidad Rey Juan Carlos, C/Tulipán s/n, 28933 Móstoles, Spain
| | - Sandra C. Cifuentes
- Department of Applied Mathematics, Materials Science and Engineering and Electronic Technology, Escuela Superior de Ciencias Experimentales y Tecnología (ESCET), Universidad Rey Juan Carlos, C/Tulipán s/n, 28933 Móstoles, Spain; (I.L.); (J.B.); (J.P.F.-H.); (M.M.); (B.T.); (J.R.)
| |
Collapse
|
18
|
Busquets MA, Rifai F. Vitrectomy With tPA for Submacular Hemorrhage Following Domestic Abuse. Ophthalmic Surg Lasers Imaging Retina 2024; 55:734-736. [PMID: 39037354 DOI: 10.3928/23258160-20240705-01] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/23/2024]
Abstract
A 35-year-old woman presented with visual acuity of 20/400 due to submacular hemorrhage 24 hours after confirmed domestic abuse with blunt trauma to the head. Surgical intervention with pars plana vitrectomy, subretinal tissue plasminogen activator (tPA) injection, and fluid-air exchange yielded prompt resolution of the pathology, regaining functional vision of 20/40 and an ability to return to work within one week. [Ophthalmic Surg Lasers Imaging Retina 2024;55:734-736.].
Collapse
|
19
|
Vechalapu SK, Kumar R, Sachan SK, Shaikh K, Mahapatra AD, Draksharapu A, Allimuthu D. Copper and Manganese Complexes of Pyridinecarboxaldimine Induce Oxidative Cell Death in Cancer Cells. ACS APPLIED BIO MATERIALS 2024; 7:6696-6705. [PMID: 39240687 DOI: 10.1021/acsabm.4c00854] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/08/2024]
Abstract
Leveraging the versatile redox behavior of transition metal complexes with heterocyclic ligands offers significant potential for discovering new anticancer therapeutics. This study presents a systematic investigation of a pyridinecarboxaldimine ligand (PyIm) with late 3d-transition metals inhibiting cancer cell proliferation and the mechanism of action. Synthesis and thorough characterization of authentic metal complexes of redox-active late 3d-transition metals enabled the validation of antiproliferative activity in liver cancer cells. Notably, (PyIm)2Mn(II) (1) and (PyIm)2Cu(II) (5) complexes exhibited a good inhibitory profile against liver cancer cells (EC50: 4.0 μM for 1 and 1.7 μM for 5) with excellent selectivity over normal kidney cells (Selectivity index, SI = 17 for 5). Subsequently, evaluation of these complexes in cancers cell lines from four different sites of origin (liver, breast, blood, and bone) demonstrated a predominant selectivity to liver and a moderate selectivity to breast cancer and leukemia cells over the normal kidney cells. The mechanism of action studies highlighted no expected DNA damage in cells, rather, the enhancement of extracellular and intracellular reactive oxygen species (ROS) resulting in mitochondrial damage leading to oxidative cell death in cancer cells. Notably, these complexes potentiated the antiproliferative effect of commercially used cancer therapeutics (cisplatin, oxaliplatin, doxorubicin, and dasatinib) in liver cancer cells. These findings position redox-active metal complexes for further evaluation as promising candidates for developing anticancer therapeutics and combination therapies.
Collapse
Affiliation(s)
- Sai Kumari Vechalapu
- Department of Chemistry, Indian Institute of Technology Kanpur, Kanpur 208016, India
| | - Rakesh Kumar
- Department of Chemistry, Indian Institute of Technology Kanpur, Kanpur 208016, India
| | - Sharad Kumar Sachan
- Department of Chemistry, Indian Institute of Technology Kanpur, Kanpur 208016, India
| | - Kanchan Shaikh
- Department of Chemistry, Indian Institute of Technology Kanpur, Kanpur 208016, India
| | | | - Apparao Draksharapu
- Department of Chemistry, Indian Institute of Technology Kanpur, Kanpur 208016, India
| | - Dharmaraja Allimuthu
- Department of Chemistry, Indian Institute of Technology Kanpur, Kanpur 208016, India
| |
Collapse
|
20
|
Taveepanich S, Chayajarus K, Jittimanee J, Phusri N, Thongdee P, Sawatdee K, Kamsri P, Punkvang A, Suttisintong K, Pungpo P, Suwannaloet W, Thongrung R, Pangjit K. Iron chelating, antioxidant, and anti-inflammatory properties of brazilin from Caesalpinia sappan Linn. Heliyon 2024; 10:e38213. [PMID: 39397930 PMCID: PMC11470788 DOI: 10.1016/j.heliyon.2024.e38213] [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: 06/14/2024] [Revised: 09/01/2024] [Accepted: 09/19/2024] [Indexed: 10/15/2024] Open
Abstract
Background Iron overload and inflammation are severe conditions that can lead to various chronic diseases. However, the current iron chelator drugs have their limitations. The phytochemical compounds from herbals, such as brazilin, the major active compound in Caesalpinia sappan Linn., have significant therapeutic potential in various chronic diseases. Our study was designed to examine the effect of brazilin on iron chelating properties, antioxidant activity in hepatocytes, and anti-inflammatory potential in macrophages. Methods This study focused on the isolation, purification, and evaluation of brazilin, the principal bioactive constituent found in C. sappan wood. Brazilin was extracted via methanol maceration followed by column chromatography purification. The purified compound was characterized using high-performance liquid chromatography (HPLC), nuclear magnetic resonance (NMR) spectroscopy, and mass spectrometry (MS). The antioxidant potential of brazilin was assessed by in vitro assays, including 2,2-diphenyl-1-picrylhydrazyl (DPPH), 2,2'-azinobis-(3-ethylbenzthiazolin-6-sulfonic acid (ABTS), and ferric-reducing antioxidant power (FRAP). Furthermore, its cellular antioxidant activity was evaluated using hydrogen peroxide-induced oxidative stress in the hepatocellular carcinoma cell line (Huh-7). The iron-chelating capacity of brazilin was determined spectrophotometrically, and Job's plot method was used to elucidated the stoichiometry of the iron-brazilin complex formation. The anti-inflammatory properties of brazilin were investigated in lipopolysaccharide (LPS)-stimulated macrophages (RAW 264.7). Nitric oxide (NO) inhibition was quantified using the Griess reagent, while the expression levels of pro-inflammatory cytokines, interleukin-6 (IL-6) and tumor necrosis factor-alpha (TNF-α), were evaluated by RT-qPCR. Results The results demonstrated that brazilin exhibited potent antioxidant activity in vitro and hepatocytes in a concentration-dependent manner. It also showed anti-inflammatory activity, in which NO production was significantly reduced and IL-6 and TNF-α expression in LPS-induced macrophages were repressed. Furthermore, it can bind ferric and ferrous ions. Brazilin acts as a bidentate iron chelator that forms a complex with iron in a 2:1 ratio, and two water molecules are used as additional chelators in this complex. Conclusions Our findings have significant implications. Brazilin can potentially alleviate the harmful effects of iron-induced oxidative stress and inflammatory disorders.
Collapse
Affiliation(s)
- Somjintana Taveepanich
- Department of Chemistry and Center of Excellence for Innovation in Chemistry, Faculty of Science, Ubon Ratchathani University, Ubon Ratchathani, 34190, Thailand
| | - Kampanart Chayajarus
- Department of Chemistry and Center of Excellence for Innovation in Chemistry, Faculty of Science, Ubon Ratchathani University, Ubon Ratchathani, 34190, Thailand
| | - Jutharat Jittimanee
- College of Medicine and Public Health, Ubon Ratchathani University, Ubon Ratchathani, 34190, Thailand
| | - Naruedon Phusri
- Department of Chemistry, Faculty of Science, Ubon Ratchathani University, Ubon Ratchathani, 34190, Thailand
| | - Paptawan Thongdee
- Department of Chemistry, Faculty of Science, Ubon Ratchathani University, Ubon Ratchathani, 34190, Thailand
| | - Khemmisara Sawatdee
- Department of Chemistry, Faculty of Science, Ubon Ratchathani University, Ubon Ratchathani, 34190, Thailand
| | - Pharit Kamsri
- Division of Chemistry, Faculty of Science, Nakhon Phanom University, Nakhon Phanom, 48000, Thailand
| | - Auradee Punkvang
- Division of Chemistry, Faculty of Science, Nakhon Phanom University, Nakhon Phanom, 48000, Thailand
| | - Khomson Suttisintong
- National Nanotechnology Center, NSTDA, 111 Thailand Science Park, Klong Luang, Pathum Thani, 12120, Thailand
| | - Pornpan Pungpo
- Department of Chemistry and Center of Excellence for Innovation in Chemistry, Faculty of Science, Ubon Ratchathani University, Ubon Ratchathani, 34190, Thailand
| | - Wanwisa Suwannaloet
- College of Medicine and Public Health, Ubon Ratchathani University, Ubon Ratchathani, 34190, Thailand
| | - Ruttiya Thongrung
- College of Medicine and Public Health, Ubon Ratchathani University, Ubon Ratchathani, 34190, Thailand
| | - Kanjana Pangjit
- College of Medicine and Public Health, Ubon Ratchathani University, Ubon Ratchathani, 34190, Thailand
| |
Collapse
|
21
|
Ghazaiean M, Aliasgharian A, Karami H, Ghasemi MM, Darvishi‐Khezri H. Antioxidative effects of N-acetylcysteine in patients with β-thalassemia: A quick review on clinical trials. Health Sci Rep 2024; 7:e70096. [PMID: 39381531 PMCID: PMC11458667 DOI: 10.1002/hsr2.70096] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Revised: 08/15/2024] [Accepted: 09/05/2024] [Indexed: 10/10/2024] Open
Abstract
Background and Aims Several studies have highlighted the potent antioxidant properties of N-acetyl cysteine (NAC). This review aimed to assess the impact of NAC on oxidative stress biomarkers in patients with β-thalassemia. Methods The review included articles published before 2024 that investigated the effects of NAC on oxidative stress in individuals with β-thalassemia. A comprehensive search was conducted across various databases, including Scopus, PubMed, Web of Science, Trip, and CENTRAL. Only English-language clinical trials were considered for inclusion in this review. Besides, the number needed to treat (NNT) was calculated based on the included studies. Results Ninety-nine articles were retrieved from electronic databases, and after a thorough review, eight articles were selected for comprehensive text analysis. The highest dose of NAC administered was 10 mg/kg/day (equivalent to 600 mg/day) over a period of 3-6 months. All the studies assessing the impact of NAC on oxidative stress indicators in β-thalassemia patients demonstrated positive effects during the 3-month follow-up period. Most estimated NNTs fell into 1-5, suggesting significant clinical therapeutic value in this context. Conclusion The current potency of NAC alone appears to be effective in ameliorating oxidative stress in patients with β-thalassemia major. While a 3-month duration seems adequate to demonstrate the antioxidant properties of NAC in this population, larger and well-designed clinical trials are warranted. Current clinical evidence possesses a high risk of bias.
Collapse
Affiliation(s)
- Mobin Ghazaiean
- Student Research Committee, Faculty of MedicineMazandaran University of Medical SciencesSariIran
- Gut and Liver Research Center, Non‐Communicable Disease InstituteMazandaran University of Medical SciencesSariIran
| | - Aily Aliasgharian
- Thalassemia Research Center (TRC), Hemoglobinopathy InstituteMazandaran University of Medical SciencesSariIran
| | - Hossein Karami
- Thalassemia Research Center (TRC), Hemoglobinopathy InstituteMazandaran University of Medical SciencesSariIran
| | - Mohammad Mohsen Ghasemi
- Student Research Committee, Faculty of MedicineMazandaran University of Medical SciencesSariIran
| | - Hadi Darvishi‐Khezri
- Thalassemia Research Center (TRC), Hemoglobinopathy InstituteMazandaran University of Medical SciencesSariIran
| |
Collapse
|
22
|
Sun JY, Zhou ZR, Wang YQ, Zhu DY, Ma DR. OsHRZ1 negatively regulates rice resistant to Magnaporthe oryzae infection by targeting OsVOZ2. Transgenic Res 2024; 33:489-501. [PMID: 39333318 DOI: 10.1007/s11248-024-00415-8] [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: 07/03/2024] [Accepted: 09/23/2024] [Indexed: 09/29/2024]
Abstract
Rice blast disease caused by Magnaporthe oryzae significantly reduces yield production. Blast resistance is closely associated with iron (Fe) status, but the mechanistic basis linking iron status to immune function in rice remains largely unknown. Here, iron-binding haemerythrin RING ubiquitin ligases OsHRZ1 was confirmed to play key roles in iron-mediated rice blast resistance. The expression of OsHRZ1 was suppressed by M. oryzae inoculation and high iron treatment. Both mutants of OsHRZ1 enhanced rice resistance to M. oryzae. OsPR1a was up-regulated in OsHRZ1 mutants. Yeast two-hybrid, bimolecular fluorescence complementation, and Co-IP assay results indicated that OsHRZ1 interacts with Vascular Plant One Zinc Finger 2 (OsVOZ2) in the nucleus. Additionally, the vitro ubiquitination assay indicated that OsHRZ1 can ubiquitinate OsVOZ2 and mediate the degradation of OsVOZ2. The mutants of OsVOZ2 showed reduced resistance to M. oryzae and down-regulated the expression of OsPR1a. Yeast one-hybrid, EMSA, and dual-luciferase reporter assay results indicated that OsVOZ2 directly binds to the promoter of OsPR1a, activating its expression. In summary, OsHRZ1 plays an important role in rice disease resistance by mediated degradation of OsVOZ2 thus shaping PR gene expression dynamics in rice cells. This highlights an important link between iron signaling and rice pathogen defenses.
Collapse
Affiliation(s)
- Jia-Ying Sun
- Rice Research Institute, Shenyang Agricultural University, Shenyang, China
| | - Zeng-Ran Zhou
- College of Plant Protection, Shenyang Agricultural University, Shenyang, China
| | - Yu-Qi Wang
- College of Plant Protection, Shenyang Agricultural University, Shenyang, China
| | - Dong-Yu Zhu
- College of Plant Protection, Shenyang Agricultural University, Shenyang, China
| | | |
Collapse
|
23
|
Asejeje FO, Asejeje GI, Ogunro OB, Adedara AO, Abolaji AO. Protective role of hesperetin in Drosophila melanogaster model of ferrous sulphate-induced toxicity. BMC Pharmacol Toxicol 2024; 25:70. [PMID: 39334451 PMCID: PMC11438368 DOI: 10.1186/s40360-024-00792-0] [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: 05/24/2024] [Accepted: 09/17/2024] [Indexed: 09/30/2024] Open
Abstract
The toxicological hazard of iron-containing products is a public health concern that inspires research in identifying and developing readily available, inexpensive antidotes. Natural products, like plant-sourced antioxidants, can be of great value in this regard. Hesperetin a flavonoid abundantly present in citrus fruits is known to possess a diverse pharmacological and antioxidant attribute. The present study investigated the alleviation of detrimental effects of ferrous sulphate (FeSO4) by hesperetin in Drosophila melanogaster. Flies were exposed to FeSO4 (10 µM) alone or supplemented with hesperetin (50 or 100 µM) via diet for 7 consecutive days. Antioxidant enzyme activities, non-enzymatic antioxidant levels, acetylcholinesterase activity and oxidative stress markers were then measured. Hesperetin supplementation significantly (p < 0.05) attenuated FeSO4-induced oxidative stress by enhancement of enzymic antioxidants (catalase and glutathione-S-transferases) activities, preservation of non-enzymic antioxidants (total thiols and non-protein thiols), and reduction of other markers of oxidative stress (hydrogen peroxide, protein carbonyl and lipid peroxidation) in D. melanogaster. In addition, hesperetin supplementation decreased nitric oxide levels and enhanced acetylcholinesterase activity. Furthermore, hesperetin supplementation improved FeSO4-induced locomotor deficit, while there was no significant difference in cell viability (mitochondrial metabolic rate) in the treatment groups. This study suggests that hesperetin might be a promising functional agent in preventing iron toxicity and similar metal-induced impairments.
Collapse
Affiliation(s)
- Folake Olubukola Asejeje
- Department of Chemical Sciences, Faculty of Natural Sciences, Ajayi Crowther University, Oyo, Oyo State, Nigeria
| | - Gbolahan Iyiola Asejeje
- Drosophila Research and Training Centre, A2 Ajao Dental Street, Salami Somade Estate, Off Iyaniwura Bus Stop, Basorun, Ibadan, Oyo State, Nigeria.
- Department of Chemistry, University of Ibadan, Ibadan, Oyo State, Nigeria.
| | - Olalekan Bukunmi Ogunro
- Department of Biological Sciences, KolaDaisi University, Ibadan, Oyo State, 2001213, Nigeria
| | - Adeola O Adedara
- Drosophila Research and Training Centre, A2 Ajao Dental Street, Salami Somade Estate, Off Iyaniwura Bus Stop, Basorun, Ibadan, Oyo State, Nigeria
| | - Amos Olalekan Abolaji
- Molecular Drug Metabolism and Toxicology Unit, Department of Biochemistry, University of Ibadan, Ibadan, Oyo State, Nigeria.
- Drosophila Laboratory, Drug Metabolism and Toxicology Unit, Department of Biochemistry, College of Medicine, University of Ibadan, Ibadan, Oyo State, Nigeria.
| |
Collapse
|
24
|
Fatima S, Zhou H, Chen Y, Liu Q. Role of ferroptosis in the pathogenesis of heart disease. Front Physiol 2024; 15:1450656. [PMID: 39318361 PMCID: PMC11420141 DOI: 10.3389/fphys.2024.1450656] [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: 06/17/2024] [Accepted: 08/30/2024] [Indexed: 09/26/2024] Open
Abstract
Ferroptosis is a new form of regulated necrosis characterized by iron-dependent lipid peroxidation, leading to irreparable lipid damage, membrane permeabilization, and necrotic cell death. Ferroptosis has recently been implicated in the pathogenesis of multiple forms of heart disease such as myocardial infarction, cardiac hypertrophy, heart failure, and various cardiomyopathies. Important progress has also been made regarding how ferroptosis is regulated in vitro and in vivo as well as its role in cardiac homeostasis and disease pathogenesis. In this review, we discuss molecular mechanisms that regulates ferroptosis in the heart, including pathways leading to iron overload and lipid peroxidation as well as the roles of key organelles in this process. We also discuss recent findings pertaining to the new pathogenic role of ferroptosis in various forms of heart disease as well as genetic and pharmacologic strategies targeting ferroptosis in the heart.
Collapse
Affiliation(s)
| | | | | | - Qinghang Liu
- Department of Physiology and Biophysics, University of Washington, Seattle, WA, United States
| |
Collapse
|
25
|
Kao AT, Cabanlong CV, Padilla K, Xue X. Unveiling ferroptosis as a promising therapeutic avenue for colorectal cancer and colitis treatment. Acta Pharm Sin B 2024; 14:3785-3801. [PMID: 39309484 PMCID: PMC11413686 DOI: 10.1016/j.apsb.2024.05.025] [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: 12/28/2023] [Revised: 03/31/2024] [Accepted: 04/30/2024] [Indexed: 09/25/2024] Open
Abstract
Ferroptosis is a novel type of regulated cell death (RCD) involving iron accumulation and lipid peroxidation. Since its discovery in 2012, various studies have shown that ferroptosis is associated with the pathogenesis of various diseases. Ferroptotic cell death has also been linked to intestinal dysfunction but can act as either a positive or negative regulator of intestinal disease, depending on the cell type and disease context. The continued investigation of mechanisms underlying ferroptosis provides a wealth of potential for developing novel treatments. Considering the growing prevalence of intestinal diseases, particularly colorectal cancer (CRC) and inflammatory bowel disease (IBD), this review article focuses on potential therapeutics targeting the ferroptotic pathway in relation to CRC and IBD.
Collapse
Affiliation(s)
| | | | - Kendra Padilla
- Department of Biochemistry and Molecular Biology, University of New Mexico, Albuquerque, NM 87131, USA
| | - Xiang Xue
- Department of Biochemistry and Molecular Biology, University of New Mexico, Albuquerque, NM 87131, USA
| |
Collapse
|
26
|
Zhang DD. Ironing out the details of ferroptosis. Nat Cell Biol 2024; 26:1386-1393. [PMID: 38429476 DOI: 10.1038/s41556-024-01361-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Accepted: 01/22/2024] [Indexed: 03/03/2024]
Abstract
Ferroptosis, spurred by excess labile iron and lipid peroxidation, is implicated in various diseases. Advances have been made in comprehending the lipid-peroxidation side of ferroptosis, but the exact role of iron in driving ferroptosis remains unknown. Although iron overload is characterized in multiple disease states, the potential role of ferroptosis within them remains undefined. This overview focuses on the 'ferro' side of ferroptosis, highlighting iron dysregulation in human diseases and potential therapeutic strategies targeting iron regulation and metabolism.
Collapse
Affiliation(s)
- Donna D Zhang
- Center for Inflammation Science and Systems Medicine, The Herbert Wertheim UF Scripps Institute for Biomedical Innovation and Technology, Jupiter, FL, USA.
| |
Collapse
|
27
|
Farrell R, Pascuzzi N, Chen YL, Kim M, Torres M, Gollahon L, Chen KHE. Prolactin Drives Iron Release from Macrophages and Uptake in Mammary Cancer Cells through CD44. Int J Mol Sci 2024; 25:8941. [PMID: 39201626 PMCID: PMC11354873 DOI: 10.3390/ijms25168941] [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: 07/27/2024] [Revised: 08/13/2024] [Accepted: 08/15/2024] [Indexed: 09/02/2024] Open
Abstract
Iron is an essential element for human health. In humans, dysregulated iron homeostasis can result in a variety of disorders and the development of cancers. Enhanced uptake, redistribution, and retention of iron in cancer cells have been suggested as an "iron addiction" pattern in cancer cells. This increased iron in cancer cells positively correlates with rapid tumor growth and the epithelial-to-mesenchymal transition, which forms the basis for tumor metastasis. However, the source of iron and the mechanisms cancer cells adopt to actively acquire iron is not well understood. In the present study, we report, for the first time, that the peptide hormone, prolactin, exhibits a novel function in regulating iron distribution, on top of its well-known pro-lactating role. When stimulated by prolactin, breast cancer cells increase CD44, a surface receptor mediating the endocytosis of hyaluronate-bound iron, resulting in the accumulation of iron in cancer cells. In contrast, macrophages, when treated by prolactin, express more ferroportin, the only iron exporter in cells, giving rise to net iron output. Interestingly, when co-culturing macrophages with pre-stained labile iron pools and cancer cells without any iron staining, in an iron free condition, we demonstrate direct iron flow from macrophages to cancer cells. As macrophages are one of the major iron-storage cells and it is known that macrophages infiltrate tumors and facilitate their progression, our work therefore presents a novel regulatory role of prolactin to drive iron flow, which provides new information on fine-tuning immune responses in tumor microenvironment and could potentially benefit the development of novel therapeutics.
Collapse
Affiliation(s)
- Reagan Farrell
- Department of Biological Sciences, Texas Tech University, Lubbock, TX 79409, USA; (R.F.); (N.P.); (M.T.); (L.G.)
| | - Nicholas Pascuzzi
- Department of Biological Sciences, Texas Tech University, Lubbock, TX 79409, USA; (R.F.); (N.P.); (M.T.); (L.G.)
| | - Yi-Ling Chen
- Department of Electronic Engineering, National Kaohsiung University of Science and Technology, Kaohsiung 80778, Taiwan
| | - Mary Kim
- Department of Biological Sciences, Texas Tech University, Lubbock, TX 79409, USA; (R.F.); (N.P.); (M.T.); (L.G.)
| | - Miguel Torres
- Department of Biological Sciences, Texas Tech University, Lubbock, TX 79409, USA; (R.F.); (N.P.); (M.T.); (L.G.)
| | - Lauren Gollahon
- Department of Biological Sciences, Texas Tech University, Lubbock, TX 79409, USA; (R.F.); (N.P.); (M.T.); (L.G.)
| | - Kuan-Hui Ethan Chen
- Department of Biological Sciences, Texas Tech University, Lubbock, TX 79409, USA; (R.F.); (N.P.); (M.T.); (L.G.)
| |
Collapse
|
28
|
Pustimbara A, Li C, Ogura SI. Hemin enhances the 5-aminolevulinic acid-photodynamic therapy effect through the changes of cellular iron homeostasis. Photodiagnosis Photodyn Ther 2024; 48:104253. [PMID: 38901716 DOI: 10.1016/j.pdpdt.2024.104253] [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: 05/17/2024] [Revised: 06/13/2024] [Accepted: 06/17/2024] [Indexed: 06/22/2024]
Abstract
BACKGROUND Photodynamic therapy (PDT) has been utilized as a promising alternative cancer treatment due to its minimum invasiveness over the years. Exogenous 5-aminolevulinic acid (ALA) triggers protoporphyrin IX (PpIX) accumulation, which happens in cancer cells. However, certain types of cancer exhibit reduced effectiveness in the PpIX accumulation mechanism. This study aimed to determine the effect of ALA-PDT combination with hemin on gastric carcinoma TMK-1 cells. METHODS This study utilized TMK-1 gastric cancer cell line to evaluate PpIX, ROS, and Fe2+ accumulation following the administration of ALA, hemin, and a combination of ALA and hemin PDT. We also evaluate the mRNA expressions related to iron homeostasis and treatment impacts on cell viability. RESULTS The co-addition of ALA and hemin PDT for 4 h of treatment resulted in a significant decrease in cell viability by up to 18 %. While ALA-PDT enhanced PpIX metabolism, the addition of hemin influenced both the production of reactive oxygen species (ROS) and cellular iron homeostasis by inducing Fe2+ accumulation and affecting mRNA levels of IRP, Tfr1, Ferritin, NFS1, and SDHB. CONCLUSION These findings suggest that the addition of ALA and hemin enhances phototoxicity in TMK-1 cells. The combination of ALA and hemin with PDT induces cell death, evidenced by increased cytotoxicity properties such as PpIX and ROS, along with significant changes in TMK-1 gastric cancer iron homeostasis. Therefore, the combination of ALA and hemin could be one of the alternatives in photodynamic therapy for cancer in the future.
Collapse
Affiliation(s)
- Anantya Pustimbara
- Tokyo Institute of Technology, School of Life Science and Technology, 4259 Midori-ku, Yokohama, 2268501, Japan.
| | - Chenhan Li
- Tokyo Institute of Technology, School of Life Science and Technology, 4259 Midori-ku, Yokohama, 2268501, Japan.
| | - Shun-Ichiro Ogura
- Tokyo Institute of Technology, School of Life Science and Technology, 4259 Midori-ku, Yokohama, 2268501, Japan.
| |
Collapse
|
29
|
Zhang Y, Jiang Y, Dong X, Luo S, Jiao G, Weng K, Bao Q, Zhang Y, Vongsangnak W, Chen G, Xu Q. Follicular fluid-derived exosomal HMOX1 promotes granulosa cell ferroptosis involved in follicular atresia in geese (Anser cygnoides). Poult Sci 2024; 103:103912. [PMID: 38943808 PMCID: PMC11261456 DOI: 10.1016/j.psj.2024.103912] [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: 03/13/2024] [Revised: 05/04/2024] [Accepted: 05/23/2024] [Indexed: 07/01/2024] Open
Abstract
The proliferation and death of granulosa cells (GCs) in poultry play a decisive role in follicular fate and egg production. The follicular fluid (FF) contains a variety of nutrients and genetic substances to ensure the communication between follicular cells. Exosomes, as a new intercellular communication, could carry and transport the proteins, RNA, and lipids to react on GCs, which had been found in FF of various domestic animals. Whether exosomes of FF in poultry play a similar role is unclear. In this study, geese, a poultry with low egg production, were chosen, and the effect of FF exosomes on the proliferation and death of GCs was investigated. Firstly, there were not only a large number of healthy small yellow follicles (HSYFs) but also some atresia small yellow follicles (ASYFs) in the egg-laying stage. Also, the GC layers of ASYFs became loose interconnections, inward detachment, and diminished survival rate than that of HSYFs. Besides, compared to HSYFs, the contents of E2, P4, and the mRNA expression levels of ferroptosis-related genes GPX4, FPN1, and FTH1 were significantly decreased, while COX2, NCOA4, VDAC3 mRNA were significantly increased, and the structure of mitochondrial cristae disappeared and the outer membrane broke in the GC layers of ASYFs. Moreover, the ROS, MDA, and oxidation levels in the GC layers of ASYFs were significantly higher than those of HSYFs. All these hinted that ferroptosis might result in a large number of GCs death and involvement in follicle atresia. Secondly, FF exosomes were isolated from HSYFs and ASYFs, respectively, and identified by TEM, NTA, and detection of exosome marker proteins. Also, we found the exosomes were phagocytic by GCs by tracking CM-Dil. Moreover, the addition of ASYF-FF exosomes significantly elevated the MDA content, Fe2+ levels, and the mitochondrial membrane potential (MMP) in GCs, thus significantly inhibiting the proliferation of GCs, which was restored by the ferroptosis inhibitor ferrostatin-1. Thirdly, the proteomic sequencing was performed between FF-derived exosomes of HSYFs and ASYFs. We obtained 1615 differentially expressed proteins, which were mainly enriched in the protein transport and ferroptosis pathways. Among them, HMOX1 was enriched in the ferroptosis pathway based on differential protein-protein interaction network analysis. Finally, the role of HMOX1 in regulating ferroptosis in GCs was further explored. The highly expressed HMOX1 was observed in the exosomes of ASYF-FF than that in HSYF-FF. Overexpression of HMOX1 increased ATG5, LC3II, and NCOA4 expression and reduced the expression of FTH1, GPX4, PCBP2, FPN1 in the ferroptosis pathway, also promoted intracellular Fe2+ accumulation and MDA surge, which drove ferroptosis in GCs. The effects of HMOX1 on ferroptosis could be blocked by its inhibitor Znpp. Taken together, the important protein HMOX1 was identified in FF, which could be delivered to GCs via exosomes, triggering ferroptosis and thus determining the fate of follicles.
Collapse
Affiliation(s)
- Yu Zhang
- Key Laboratory for Evaluation and Utilization of Poultry Genetic Resources of Ministry of Agriculture and Rural Affairs, Yangzhou University, Yangzhou, Jiangsu 225009, China; Joint International Research Laboratory of Agriculture & Agri-Product Safety of Ministry of Education, Yangzhou University, Yangzhou 225009, Jiangsu, China
| | - Youluan Jiang
- Key Laboratory for Evaluation and Utilization of Poultry Genetic Resources of Ministry of Agriculture and Rural Affairs, Yangzhou University, Yangzhou, Jiangsu 225009, China
| | - Xiaoqian Dong
- Key Laboratory for Evaluation and Utilization of Poultry Genetic Resources of Ministry of Agriculture and Rural Affairs, Yangzhou University, Yangzhou, Jiangsu 225009, China
| | - Shuwen Luo
- Key Laboratory for Evaluation and Utilization of Poultry Genetic Resources of Ministry of Agriculture and Rural Affairs, Yangzhou University, Yangzhou, Jiangsu 225009, China
| | - Guoyu Jiao
- Key Laboratory for Evaluation and Utilization of Poultry Genetic Resources of Ministry of Agriculture and Rural Affairs, Yangzhou University, Yangzhou, Jiangsu 225009, China
| | - Kaiqi Weng
- Key Laboratory for Evaluation and Utilization of Poultry Genetic Resources of Ministry of Agriculture and Rural Affairs, Yangzhou University, Yangzhou, Jiangsu 225009, China
| | - Qiang Bao
- Key Laboratory for Evaluation and Utilization of Poultry Genetic Resources of Ministry of Agriculture and Rural Affairs, Yangzhou University, Yangzhou, Jiangsu 225009, China
| | - Yang Zhang
- Key Laboratory for Evaluation and Utilization of Poultry Genetic Resources of Ministry of Agriculture and Rural Affairs, Yangzhou University, Yangzhou, Jiangsu 225009, China; Joint International Research Laboratory of Agriculture & Agri-Product Safety of Ministry of Education, Yangzhou University, Yangzhou 225009, Jiangsu, China
| | - Wanwipa Vongsangnak
- Department of Zoology, Faculty of Science, Kasetsart University, Bangkok 10900, Thailand
| | - Guohong Chen
- Key Laboratory for Evaluation and Utilization of Poultry Genetic Resources of Ministry of Agriculture and Rural Affairs, Yangzhou University, Yangzhou, Jiangsu 225009, China; Joint International Research Laboratory of Agriculture & Agri-Product Safety of Ministry of Education, Yangzhou University, Yangzhou 225009, Jiangsu, China
| | - Qi Xu
- Key Laboratory for Evaluation and Utilization of Poultry Genetic Resources of Ministry of Agriculture and Rural Affairs, Yangzhou University, Yangzhou, Jiangsu 225009, China; Joint International Research Laboratory of Agriculture & Agri-Product Safety of Ministry of Education, Yangzhou University, Yangzhou 225009, Jiangsu, China.
| |
Collapse
|
30
|
Dugan C, Peeling P, Burden R, Richards T. Efficacy of iron supplementation on physical capacity in non-anaemic iron-deficient individuals: protocol for an individual patient data meta-analysis. Syst Rev 2024; 13:182. [PMID: 39010146 PMCID: PMC11247796 DOI: 10.1186/s13643-024-02559-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/06/2023] [Accepted: 05/13/2024] [Indexed: 07/17/2024] Open
Abstract
BACKGROUND A deficiency in iron stores is associated with various adverse health complications, which, if left untreated, can progress to states of anaemia, whereby there is significant detriment to an individual's work capacity and quality of life due to compromised erythropoiesis. The most common methods employed to treat an iron deficiency include oral iron supplementation and, in persistent and/or unresponsive cases, intravenous iron therapy. The efficacy of these treatments, particularly in states of iron deficiency without anaemia, is equivocal. Indeed, both randomised control trials and aggregate data meta-analyses have produced conflicting evidence. Therefore, this study aims to assess the efficacy of both oral and intravenous iron supplementation on physical capacity, quality of life, and fatigue scores in iron-deficient non-anaemic individuals using individual patient data (IPD) meta-analysis techniques. METHODS All potential studies, irrespective of design, will be sourced through systematic searches on the following databases: Cochrane Central Register of Controlled Trials, MEDLINE Ovid, Embase Ovid, Web of Science: Science Citation Index Expanded, Web of Science: Conference Proceedings Citation Index-Science, ClinicalTrials.gov, and World Health Organization (WHO) International Clinical Trials Registry Platform. Individual patient data from all available trials will be included and subsequently analysed in a two-stage approach. Predetermined subgroup and sensitivity analyses will be employed to further explain results. DISCUSSION The significance of this IPD meta-analysis is one of consolidating a clear consensus to better inform iron-deficient individuals of the physiological response associated with iron supplementation. The IPD approach, to the best of our knowledge, is novel for this research topic. As such, the findings will significantly contribute to the current body of evidence. SYSTEMATIC REVIEW REGISTRATION PROSPERO CRD42020191739.
Collapse
Affiliation(s)
- Cory Dugan
- School of Human Sciences, University of Western Australia, Perth, Australia.
| | - Peter Peeling
- School of Human Sciences, University of Western Australia, Perth, Australia
| | - Richard Burden
- National Centre for Sport and Exercise Medicine, School of Sport, Exercise and Health Sciences, Loughborough University, Loughborough, UK
| | - Toby Richards
- Division of Surgery, University of Western Australia, Perth, Australia
| |
Collapse
|
31
|
Chu J, Wang K, Lu L, Zhao H, Hu J, Xiao W, Wu Q. Advances of Iron and Ferroptosis in Diabetic Kidney Disease. Kidney Int Rep 2024; 9:1972-1985. [PMID: 39081773 PMCID: PMC11284386 DOI: 10.1016/j.ekir.2024.04.012] [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: 01/21/2024] [Revised: 03/29/2024] [Accepted: 04/01/2024] [Indexed: 08/02/2024] Open
Abstract
Diabetes mellitus presents a significant threat to human health because it disrupts energy metabolism and gives rise to various complications, including diabetic kidney disease (DKD). Metabolic adaptations occurring in the kidney in response to diabetes contribute to the pathogenesis of DKD. Iron metabolism and ferroptosis, a recently defined form of cell death resulting from iron-dependent excessive accumulation of lipid peroxides, have emerged as crucial players in the progression of DKD. In this comprehensive review, we highlight the profound impact of adaptive and maladaptive responses regulating iron metabolism on the progression of kidney damage in diabetes. We summarize the current understanding of iron homeostasis and ferroptosis in DKD. Finally, we propose that precise manipulation of iron metabolism and ferroptosis may serve as potential strategies for kidney management in diabetes.
Collapse
Affiliation(s)
- Jiayi Chu
- Department of Radiology, Center of Regenerative and Aging Medicine, the Fourth Affiliated Hospital of School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Zhejiang, China
| | - Kewu Wang
- Department of Radiology, Center of Regenerative and Aging Medicine, the Fourth Affiliated Hospital of School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Zhejiang, China
| | - Lulu Lu
- Department of Nutrition and Toxicology, Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines of Zhejiang Province, School of Public Health, Hangzhou Normal University, Hangzhou, China
| | - Hui Zhao
- Department of Radiology, Center of Regenerative and Aging Medicine, the Fourth Affiliated Hospital of School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Zhejiang, China
| | - Jibo Hu
- Department of Radiology, Center of Regenerative and Aging Medicine, the Fourth Affiliated Hospital of School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Zhejiang, China
| | - Wenbo Xiao
- Department of Radiology, the First Affiliated Hospital, College of Medicine, Zhejiang University, Zhejiang, China
| | - Qian Wu
- Department of Radiology, Center of Regenerative and Aging Medicine, the Fourth Affiliated Hospital of School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Zhejiang, China
| |
Collapse
|
32
|
Frydrych A, Frankowski M, Jurowski K. The toxicological analysis and assessment of essential elements (Cu, Fe, Mn, Zn) in Food for Special Medical Purposes (FSMP) dedicated to oncological patients available in Polish pharmacies. Food Chem Toxicol 2024; 189:114768. [PMID: 38810942 DOI: 10.1016/j.fct.2024.114768] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Revised: 05/23/2024] [Accepted: 05/25/2024] [Indexed: 05/31/2024]
Abstract
Foods for Special Medical Purposes (FSMP) for oncology patients, available in pharmacies, play a crucial role in providing nutrition and supplementation. However, the scientific literature lacks comprehensive research on the safety of essential trace elements in these products. This study aimed to assess Cu, Fe, Mn and Zn levels in commonly prescribed FSMPs (n = 23) from Polish pharmacies. Using ICP-MS after microwave-induced digestion (using concentrated nitric acid and hydrogen peroxide), we evaluated element levels. Our research used three approaches: the raw score for Cu, Fe, Mn, and Zn; single intake per serving; and the daily ration, compared with the reference values of the European Food Safety Authority. Discrepancies were found between the actual and declared product compositions, influenced by the route of administration and the recommended intake. Despite variations, all products were considered safe for oncological patients based on current evidence. However, it is recommended to have clear guidelines for FSMPs in cancer care. This pioneering study evaluates the safety and quality of prescription FSMPs for cancer patients from toxicological and nutritional perspectives, highlighting the need for standardised protocols in pharmacy-dispensed FSMPs.
Collapse
Affiliation(s)
- Adrian Frydrych
- Laboratory of Innovative Toxicological Research and Analyzes, Institute of Medical Sciences, Medical College, Rzeszów University, Al. mjr. W. Kopisto 2a, 35-959, Rzeszów, Poland
| | - Marcin Frankowski
- Department of Analytical and Environmental Chemistry, Faculty of Chemistry, Adam Mickiewicz University in Poznań, Uniwersytetu Poznańskiego 8, 61-614, Poznań, Poland
| | - Kamil Jurowski
- Laboratory of Innovative Toxicological Research and Analyzes, Institute of Medical Sciences, Medical College, Rzeszów University, Al. mjr. W. Kopisto 2a, 35-959, Rzeszów, Poland; Department of Regulatory and Forensic Toxicology, Institute of Medical Expertises in Łódź, ul. Aleksandrowska 67/93, 91-205, Łódź, Poland.
| |
Collapse
|
33
|
Wang J, Huang L, Chen L, Chen S, Liu S. Efficacy of anti-VEGF intravitreal injection in traumatic submacular hemorrhage: a retrospective study. Int Ophthalmol 2024; 44:259. [PMID: 38909337 DOI: 10.1007/s10792-024-03168-9] [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: 12/25/2023] [Accepted: 06/15/2024] [Indexed: 06/24/2024]
Abstract
OBJECTIVE In this study we investigated the efficacy of short-term intravitreal injections of anti-vascular endothelial growth factors (anti-VEGF) in treating traumatic submacular hemorrhage. METHODS A total of 115 patients were diagnosed with submacular hemorrhage between 2018 and 2022 at Shenzhen Eye Hospital. In a retrospective analysis, we examined 13 of these patients who presented with submacular hemorrhage and choroidal rupture due to ocular trauma. Eight patients were treated with intravitreal anti-VEGF injection and 5 with oral drugs. We systematically analyzed changes in their ocular conditions pre and post-treatment. The evaluations encompassed best-corrected visual acuity (BCVA), optical coherence tomography, fundus fluorescein angiography, and retinal imaging. RESULTS The 13 patients diagnosed with submacular hemorrhage comprised of 10 males and 3 female, with their age ranging between 27 and 64 years, with an average age of 38.1 years (standard deviation [SD]: 11.27). A statistically significant reduction in central foveal thickness (CFT) was observed following intravitreal injections of anti-VEGF drugs (P = 0.03). In control group, the CFT was reduced without statistical significance (P = 0.10). The BCVA of the patients in treatment group improved significantly from 1.15 (SD: 0.62. Range: 0.4-2) to 0.63 (SD: 0.59. Range: 0.1-1.6), indicating an average increase of 4.13 lines (SD: 3.36. Range: 0-9) as measured by the visual acuity test using an eye chart (P = 0.01). The difference between baseline visual acuity and final visual acuity was not statistically significant in control group (P = 0.51). CONCLUSION Short-term administration of anti-VEGF drugs exhibited significant efficacy in reducing submacular hemorrhage following ocular trauma and enhancing visual acuity.
Collapse
Affiliation(s)
- Jiaming Wang
- Shenzhen Eye Hospital, Jinan University, Shenzhen Eye Institute, 18th Zetian Road, Futian District, Shenzhen, 518040, China
| | - Liuhui Huang
- Centre for Eye Research Australia, Royal Victorian Eye and Ear Hospital, Melbourne, VIC, 3002, Australia
- Ophthalmology, Department of Surgery, University of Melbourne, Melbourne, VIC, 3010, Australia
- Department of Ophthalmology, Tenth People's Hospital Affiliated with Shanghai Tongji University School of Medicine, Shanghai, 200072, China
| | - Lifei Chen
- Shenzhen Eye Hospital, Jinan University, Shenzhen Eye Institute, 18th Zetian Road, Futian District, Shenzhen, 518040, China
| | - Sheng Chen
- Shenzhen Eye Hospital, Jinan University, Shenzhen Eye Institute, 18th Zetian Road, Futian District, Shenzhen, 518040, China.
| | - Shenwen Liu
- Shenzhen Eye Hospital, Jinan University, Shenzhen Eye Institute, 18th Zetian Road, Futian District, Shenzhen, 518040, China.
| |
Collapse
|
34
|
Vechalapu SK, Kumar R, Chatterjee N, Gupta S, Khanna S, Thimmappa PY, Senthil S, Eerlapally R, Joshi MB, Misra SK, Draksharapu A, Allimuthu D. Redox modulator iron complexes trigger intrinsic apoptosis pathway in cancer cells. iScience 2024; 27:109899. [PMID: 38799569 PMCID: PMC11126827 DOI: 10.1016/j.isci.2024.109899] [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: 01/22/2024] [Revised: 03/21/2024] [Accepted: 05/01/2024] [Indexed: 05/29/2024] Open
Abstract
The emergence of multidrug resistance in cancer cells necessitates the development of new therapeutic modalities. One way cancer cells orchestrate energy metabolism and redox homeostasis is through overloaded iron pools directed by iron regulatory proteins, including transferrin. Here, we demonstrate that targeting redox homeostasis using nitrogen-based heterocyclic iron chelators and their iron complexes efficiently prevents the proliferation of liver cancer cells (EC50: 340 nM for IITK4003) and liver cancer 3D spheroids. These iron complexes generate highly reactive Fe(IV)=O species and accumulate lipid peroxides to promote oxidative stress in cells that impair mitochondrial function. Subsequent leakage of mitochondrial cytochrome c activates the caspase cascade to trigger the intrinsic apoptosis pathway in cancer cells. This strategy could be applied to leverage the inherent iron overload in cancer cells to selectively promote intrinsic cellular apoptosis for the development of unique iron-complex-based anticancer therapeutics.
Collapse
Affiliation(s)
- Sai Kumari Vechalapu
- Department of Chemistry, Indian Institute of Technology Kanpur, Uttar Pradesh 208016, India
| | - Rakesh Kumar
- Department of Chemistry, Indian Institute of Technology Kanpur, Uttar Pradesh 208016, India
| | - Niranjan Chatterjee
- Department of Biological Sciences and Bioengineering, Indian Institute of Technology Kanpur, Uttar Pradesh 208016, India
| | - Sikha Gupta
- Department of Chemistry, Indian Institute of Technology Kanpur, Uttar Pradesh 208016, India
| | - Shweta Khanna
- Department of Chemistry, Indian Institute of Technology Kanpur, Uttar Pradesh 208016, India
| | - Pooja Yedehalli Thimmappa
- Department of Ageing Research, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal 576104, India
| | - Sathyapriya Senthil
- Department of Chemistry, Indian Institute of Technology Kanpur, Uttar Pradesh 208016, India
| | - Raju Eerlapally
- Department of Chemistry, Indian Institute of Technology Kanpur, Uttar Pradesh 208016, India
| | - Manjunath B. Joshi
- Department of Ageing Research, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal 576104, India
| | - Santosh K. Misra
- Department of Biological Sciences and Bioengineering, Indian Institute of Technology Kanpur, Uttar Pradesh 208016, India
| | - Apparao Draksharapu
- Department of Chemistry, Indian Institute of Technology Kanpur, Uttar Pradesh 208016, India
| | - Dharmaraja Allimuthu
- Department of Chemistry, Indian Institute of Technology Kanpur, Uttar Pradesh 208016, India
| |
Collapse
|
35
|
Wang H, Liu X, Yan X, Du Y, Pu F, Ren J, Qu X. An ATPase-Mimicking MXene nanozyme pharmacologically breaks the ironclad defense system for ferroptosis cancer therapy. Biomaterials 2024; 307:122523. [PMID: 38432004 DOI: 10.1016/j.biomaterials.2024.122523] [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: 10/20/2023] [Revised: 02/04/2024] [Accepted: 02/25/2024] [Indexed: 03/05/2024]
Abstract
Anticancer nanomedicines used for ferroptosis therapy generally rely on the direct delivery of Fenton catalysts to drive lipid peroxidation in cancer cells. However, the therapeutic efficacy is limited by the ferroptosis resistance caused by the intracellular anti-ferroptotic signals. Herein, we report the intrinsic ATPase-mimicking activity of a vanadium carbide MXene nanozyme (PVCMs) to pharmacologically modulate the nuclear factor erythroid 2-related factor 2 (Nrf2) program, which is the master anti-ferroptotic mediator in the ironclad defense system in triple-negative breast cancer (TNBC) cells. The PVCMs perform high ATPase-like activity that can effectively and selectively catalyze the dephosphorylation of ATP to generate ADP. Through a cascade mechanism initiated by falling energy status, PVCMs can powerfully hinder the Nrf2 program to selectively drive ferroptosis in TNBC cells in response to PVCMs-induced glutathione depletion. This study provides a paradigm for the use of pharmacologically active nanozymes to moderate specific cellular signals and elicit desirable pharmacological activities for therapeutic applications.
Collapse
Affiliation(s)
- Huan Wang
- State Key Laboratory of Rare Earth Resources Utilization and Laboratory of Chemical Biology, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, PR China
| | - Xinchen Liu
- Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, School of Stomatology, Jilin University, Changchun 130021, PR China
| | - Xiangyu Yan
- State Key Laboratory of Powder Metallurgy, Central South University, Changsha, Hunan 410083, PR China
| | - Yong Du
- State Key Laboratory of Powder Metallurgy, Central South University, Changsha, Hunan 410083, PR China
| | - Fang Pu
- State Key Laboratory of Rare Earth Resources Utilization and Laboratory of Chemical Biology, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, PR China
| | - Jinsong Ren
- State Key Laboratory of Rare Earth Resources Utilization and Laboratory of Chemical Biology, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, PR China.
| | - Xiaogang Qu
- State Key Laboratory of Rare Earth Resources Utilization and Laboratory of Chemical Biology, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, PR China.
| |
Collapse
|
36
|
Zhao J, Ma W, Wang S, Zhang K, Xiong Q, Li Y, Yu H, Du H. Differentiation of intestinal stem cells toward goblet cells under systemic iron overload stress are associated with inhibition of Notch signaling pathway and ferroptosis. Redox Biol 2024; 72:103160. [PMID: 38631120 PMCID: PMC11040173 DOI: 10.1016/j.redox.2024.103160] [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: 03/14/2024] [Revised: 04/09/2024] [Accepted: 04/13/2024] [Indexed: 04/19/2024] Open
Abstract
Iron overload can lead to oxidative stress and intestinal damage and happens frequently during blood transfusions and iron supplementation. However, how iron overload influences intestinal mucosa remains unknown. Here, the aim of current study was to investigate the effects of iron overload on the proliferation and differentiation of intestinal stem cells (ISCs). An iron overload mouse model was established by intraperitoneal injection of 120 mg/kg body weight iron dextran once a fortnight for a duration of 12 weeks, and an iron overload enteroid model was produced by treatment with 3 mM or 10 mM of ferric ammonium citrate for 24 h. We found that iron overload caused damage to intestinal morphology with a 64 % reduction in villus height/crypt depth ratio, and microvilli injury in the duodenum. Iron overload mediated epithelial function by inhibiting the expression of nutrient transporters and enhancing the expression of secretory factors in the duodenum. Meanwhile, iron overload inhibited the proliferation of ISCs and regulated their differentiation into secretory mature cells, such as goblet cells, through inhibiting Notch signaling pathway both in mice and enteroid. Furthermore, iron overload caused oxidative stress and ferroptosis in intestinal epithelial cells. In addition, ferroptosis could also inhibit Notch signaling pathway, and affected the proliferation and differentiation of ISCs. These findings reveal the regulatory role of iron overload on the proliferation and differentiation of ISCs, providing a new insight into the internal mechanism of iron overload affecting intestinal health, and offering important theoretical basis for the scientific application of iron nutrition regulation.
Collapse
Affiliation(s)
- Jing Zhao
- Key Laboratory of Molecular Animal Nutrition, Ministry of Education, College of Animal Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Wan Ma
- Key Laboratory of Molecular Animal Nutrition, Ministry of Education, College of Animal Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Sisi Wang
- Key Laboratory of Molecular Animal Nutrition, Ministry of Education, College of Animal Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Kang Zhang
- Key Laboratory of Molecular Animal Nutrition, Ministry of Education, College of Animal Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Qingqing Xiong
- Key Laboratory of Molecular Animal Nutrition, Ministry of Education, College of Animal Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Yunqin Li
- Analysis Center of Agrobiology and Environmental Science, Zhejiang University, Hangzhou, 310058, China
| | - Hong Yu
- Key Laboratory of Precise Diagnosis and Treatment of Abdominal Infection of Zhejiang Province, Department of General Surgery, Sir Run-Run Shaw Hospital, Zhejiang University, Hangzhou, 310016, China
| | - Huahua Du
- Key Laboratory of Molecular Animal Nutrition, Ministry of Education, College of Animal Sciences, Zhejiang University, Hangzhou, 310058, China; Key Laboratory of Precise Diagnosis and Treatment of Abdominal Infection of Zhejiang Province, Department of General Surgery, Sir Run-Run Shaw Hospital, Zhejiang University, Hangzhou, 310016, China.
| |
Collapse
|
37
|
Nie J, Ai J, Hong W, Bai Z, Wang B, Yang J, Zhang Z, Mo F, Yang J, Sun Q, Wei X. Cisplatin-induced oxPAPC release enhances MDSCs infiltration into LL2 tumour tissues through MCP-1/CCL2 and LTB4/LTB4R pathways. Cell Prolif 2024; 57:e13570. [PMID: 37905494 PMCID: PMC10984104 DOI: 10.1111/cpr.13570] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Revised: 10/09/2023] [Accepted: 10/15/2023] [Indexed: 11/02/2023] Open
Abstract
Lung cancer is the leading global cause of cancer-related death, however, resistance to chemotherapy drugs remains a huge barrier to effective treatment. The elevated recruitment of myeloid derived suppressor cells (MDSCs) to tumour after chemotherapy has been linked to resistance of chemotherapy drugs. Nevertheless, the specific mechanism remains unclear. oxPAPC is a bioactive principal component of minimally modified low-density lipoproteins and regulates inflammatory response. In this work, we found that cisplatin, oxaliplatin and ADM all increased oxPAPC release in tumour. Treating macrophages with oxPAPC in vitro stimulated the secretion of MCP-1 and LTB4, which strongly induced monocytes and neutrophils chemotaxis, respectively. Injection of oxPAPC in vivo significantly upregulated the percentage of MDSCs in tumour microenvironment (TME) of wild-type LL2 tumour-bearing mice, but not CCL2-/- mice and LTB4R-/- mice. Critically, oxPAPC acted as a pro-tumor factor in LL2 tumour model. Indeed, cisplatin increased oxPAPC level in tumour tissues of WT mice, CCL2-/- and LTB4R-/- mice, but caused increased infiltration of Ly6Chigh monocytes and neutrophils only in WT LL2-bearing mice. Collectively, our work demonstrates cisplatin treatment induces an overproduction of oxPAPC and thus recruits MDSCs infiltration to promote the tumour growth through the MCP-1/CCL2 and LTB4/LTB4R pathways, which may restrict the effect of multiple chemotherapy. This provides evidence for a potential strategy to enhance the efficacy of multiple chemotherapeutic drugs in the treatment of lung cancer by targeting oxPAPC.
Collapse
Affiliation(s)
- Ji Nie
- Department of Biotherapy, Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy, National Clinical Research Center for GeriatricsWest China Hospital, Sichuan UniversityChengduSichuanChina
- Department of Pulmonary and Critical Care Medicine, The First People's Hospital of Yunnan ProvinceThe Affiliated Hospital of Kunming University of Science and TechnologyKunmingYunnanChina
| | - Jiayuan Ai
- Department of Biotherapy, Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy, National Clinical Research Center for GeriatricsWest China Hospital, Sichuan UniversityChengduSichuanChina
| | - Weiqi Hong
- Department of Biotherapy, Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy, National Clinical Research Center for GeriatricsWest China Hospital, Sichuan UniversityChengduSichuanChina
| | - Ziyi Bai
- Department of Biotherapy, Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy, National Clinical Research Center for GeriatricsWest China Hospital, Sichuan UniversityChengduSichuanChina
| | - Binhan Wang
- Department of Biotherapy, Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy, National Clinical Research Center for GeriatricsWest China Hospital, Sichuan UniversityChengduSichuanChina
| | - Jingyun Yang
- Department of Biotherapy, Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy, National Clinical Research Center for GeriatricsWest China Hospital, Sichuan UniversityChengduSichuanChina
| | - Ziqi Zhang
- Department of Biotherapy, Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy, National Clinical Research Center for GeriatricsWest China Hospital, Sichuan UniversityChengduSichuanChina
| | - Fei Mo
- Department of Biotherapy, Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy, National Clinical Research Center for GeriatricsWest China Hospital, Sichuan UniversityChengduSichuanChina
| | - Jing Yang
- Department of Biotherapy, Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy, National Clinical Research Center for GeriatricsWest China Hospital, Sichuan UniversityChengduSichuanChina
| | - Qiu Sun
- Department of Biotherapy, Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy, National Clinical Research Center for GeriatricsWest China Hospital, Sichuan UniversityChengduSichuanChina
- West China Medical Publishers, West China Hospital, Sichuan UniversityChengduSichuanChina
| | - Xiawei Wei
- Department of Biotherapy, Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy, National Clinical Research Center for GeriatricsWest China Hospital, Sichuan UniversityChengduSichuanChina
| |
Collapse
|
38
|
Chen T, Liang L, Wang Y, Li X, Yang C. Ferroptosis and cuproptposis in kidney Diseases: dysfunction of cell metabolism. Apoptosis 2024; 29:289-302. [PMID: 38095762 PMCID: PMC10873465 DOI: 10.1007/s10495-023-01928-z] [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] [Accepted: 12/03/2023] [Indexed: 02/18/2024]
Abstract
Metal ions play an important role in living organisms and are involved in essential physiological activities. However, the overload state of ions can cause excess free radicals, cell damage, and even cell death. Ferroptosis and cuproptosis are specific forms of cell death that are distinct from apoptosis, necroptosis, and other regulated cell death. These unique modalities of cell death, dependent on iron and copper, are regulated by multiple cellular metabolic pathways, including steady-state metal redox treatment mitochondrial activity of lipid, amino acid and glucose metabolism, and various signaling pathways associated with disease. Although the mechanisms of ferroptosis and cuproptosis are not yet fully understood, there is no doubt that ion overload plays a crucial act in these metal-dependent cell deaths. In this review, we discussed the core roles of ion overload in ferroptosis and cuproptosis, the association between metabolism imbalance and ferroptosis and cuproptosis, the extract the diseases caused by ion overload and current treatment modalities.
Collapse
Affiliation(s)
- Tingting Chen
- Department of Pharmacy, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Lifei Liang
- Department of Urology, Zhongshan Hospital, Fudan University, No. 180 Fenglin Road, Shanghai, China
- Shanghai Key Laboratory of Organ Transplantation, Shanghai, China
| | - Yuzhu Wang
- Department of Pharmacy, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Xiaoyu Li
- Department of Pharmacy, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Cheng Yang
- Department of Urology, Zhongshan Hospital, Fudan University, No. 180 Fenglin Road, Shanghai, China.
- Shanghai Key Laboratory of Organ Transplantation, Shanghai, China.
- Zhangjiang Institue of Fudan University, Shanghai, China.
| |
Collapse
|
39
|
Hacioglu C, Kar F, Ozbayer C, Gundogdu AC. Ex vivo investigation of betaine and boric acid function as preprotective agents on rat synaptosomes to be treated with Aβ (1-42). ENVIRONMENTAL TOXICOLOGY 2024; 39:2138-2149. [PMID: 38108610 DOI: 10.1002/tox.24098] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/15/2023] [Revised: 08/31/2023] [Accepted: 12/01/2023] [Indexed: 12/19/2023]
Abstract
Recent evidence suggests that ferroptosis, an iron-dependent cell death process, may be involved in Alzheimer's disease (AD) pathology. The study evaluated the therapeutic potential of betaine and boric acid (BA) pretreatment administered to rats for 21 days in AD. Then, the rats were sacrificed, and morphological and biochemical analyses were performed in brain tissues. Next, an ex vivo AD model was created by applying amyloid-β (Aβ1-42) to synaptosomes isolated from the brain tissues. Synaptosomes were analyzed with micrograph images, and protein and mRNA levels of ferroptotic markers were determined. Betaine and BA pretreatments did not cause any morphological and biochemical differences in the brain tissue. However, Aβ (1-42) administration in synaptosomes increased the levels of acyl-CoA synthetase long chain family member-4 (ACSL4), transferrin receptor-1 protein (TfR1), malondialdehyde (MDA), and 8-hydroxydeoxyguanosine (8-OHdG) and decreased the levels glutathione peroxidase-4 (GPx4) and glutathione (GSH). Moreover, ACSL4, GPx4, and TfR1 mRNA and protein levels were similar to the ELISA results. In contrast, betaine and BA pretreatments decreased the levels of ACSL4, TfR1, MDA, and 8-OHdG in synaptosomes incubated with Aβ1-42, while promoting increased levels of GPx4 and GSH. In addition, betaine and BA pretreatments completely reversed ACSL4, GPx4, and TfR1 mRNA and protein levels. Therefore, betaine and BA pretreatments may contribute to the prevention of neurodegenerative damage by supporting antiferroptotic activities.
Collapse
Affiliation(s)
- Ceyhan Hacioglu
- Faculty of Pharmacy, Department of Biochemistry, Düzce University, Düzce, Turkey
- Faculty of Medicine, Department of Medical Biochemistry, Düzce University, Düzce, Turkey
| | - Fatih Kar
- Faculty of Medicine, Department of Medical Biochemistry, Kütahya Health Sciences University, Kütahya, Turkey
| | - Cansu Ozbayer
- Faculty of Medicine, Department of Medical Biology, Kütahya Health Sciences University, Kütahya, Turkey
| | - Ayse Cakir Gundogdu
- Faculty of Medicine Department of Histology and Embryology, Kütahya Health Sciences University, Kütahya, Turkey
| |
Collapse
|
40
|
Bian Y, Qu X, Zhang F, Zhang Z, Kang J. The Monitoring and Cell Imaging of Fe 3+ Using a Chromone-Based Fluorescence Probe. Molecules 2024; 29:1504. [PMID: 38611784 PMCID: PMC11013790 DOI: 10.3390/molecules29071504] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Revised: 03/23/2024] [Accepted: 03/25/2024] [Indexed: 04/14/2024] Open
Abstract
A new structurally simple fluorescent CP probe based on chromone was designed and synthesized, and its structure was fully characterized using various analytical techniques. The CP probe displays a high selectivity and sensitivity for sensing Fe3+ with a "turn-off" fluorescence response over other metal ions in a DMSO/H2O (4:1, v/v) solution. The experiment results show that the CP probe is stable over a wide pH range of 2.0-12.0. The detection limit for Fe3+ was calculated to be 0.044 μmol•L-1. The molar ratio method indicated that the binding mode between the CP probe and Fe3+ is a 1:1 complex formation. HR-MS and density functional theory (DFT) calculations were also performed to further confirm the recognition mechanism. Both fluorescence imaging experiments and the MTT assay demonstrated that the CP probe was suitable for detecting intracellular Fe3+ and no significant cytotoxicity in living cells.
Collapse
Affiliation(s)
- Yongjun Bian
- College of Chemistry and Chemical Engineering, Jinzhong University, Jinzhong 030619, China; (X.Q.); (Z.Z.); (J.K.)
| | - Xingyu Qu
- College of Chemistry and Chemical Engineering, Jinzhong University, Jinzhong 030619, China; (X.Q.); (Z.Z.); (J.K.)
| | - Fengying Zhang
- Department of Materials Science and Engineering, Jinzhong University, Jinzhong 030619, China
| | - Zhengwei Zhang
- College of Chemistry and Chemical Engineering, Jinzhong University, Jinzhong 030619, China; (X.Q.); (Z.Z.); (J.K.)
| | - Jin Kang
- College of Chemistry and Chemical Engineering, Jinzhong University, Jinzhong 030619, China; (X.Q.); (Z.Z.); (J.K.)
| |
Collapse
|
41
|
Zaugg J, Lopez-Tello J, Musial B, Vaughan OR, Fowden AL, Albrecht C, Sferruzzi-Perri AN. Obesogenic diet in pregnancy disrupts placental iron handling and ferroptosis and stress signalling in association with fetal growth alterations. Cell Mol Life Sci 2024; 81:151. [PMID: 38526599 DOI: 10.1007/s00018-024-05192-5] [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: 11/27/2023] [Revised: 02/07/2024] [Accepted: 02/29/2024] [Indexed: 03/26/2024]
Abstract
Obesity and gestational diabetes (GDM) impact fetal growth during pregnancy. Iron is an essential micronutrient needed for energy-intense feto-placental development, but if mis-handled can lead to oxidative stress and ferroptosis (iron-dependent cell death). In a mouse model showing maternal obesity and glucose intolerance, we investigated the association of materno-fetal iron handling and placental ferroptosis, oxidative damage and stress signalling activation with fetal growth. Female mice were fed a standard chow or high fat, high sugar (HFHS) diet during pregnancy and outcomes were measured at day (d)16 or d19 of pregnancy. In HFHS-fed mice, maternal hepcidin was reduced and iron status maintained (tissue iron levels) at both d16 and d19. However, fetal weight, placental iron transfer capacity, iron deposition, TFR1 expression and ERK2-mediated signalling were reduced and oxidative damage-related lipofuscin accumulation in the placenta was increased in HFHS-fed mice. At d19, whilst TFR1 remained decreased, fetal weight was normal and placental weight, iron content and iron transporter genes (Dmt1, Zip14, and Fpn1) were reduced in HFHS-fed mice. Furthermore, there was stress kinase activation (increased phosphorylated p38MAPK, total ERK and JNK) in the placenta from HFHS-fed mice at d19. In summary, a maternal HFHS diet during pregnancy impacts fetal growth trajectory in association with changes in placental iron handling, ferroptosis and stress signalling. Downregulation of placental iron transporters in HFHS mice may protect the fetus from excessive oxidative iron. These findings suggest a role for alterations in placental iron homeostasis in determining perinatal outcomes of pregnancies associated with GDM and/or maternal obesity.
Collapse
Affiliation(s)
- Jonas Zaugg
- Department of Physiology, Development and Neuroscience, University of Cambridge, Downing Street, Cambridge, CB2 3EG, UK
- Institute of Biochemistry and Molecular Medicine, University of Bern, Bühlstrasse 28, CH-3012, Bern, Switzerland
- Swiss National Centre of Competence in Research (NCCR) TransCure, University of Bern, Bern, Switzerland
| | - Jorge Lopez-Tello
- Department of Physiology, Development and Neuroscience, University of Cambridge, Downing Street, Cambridge, CB2 3EG, UK
| | - Barbara Musial
- Department of Physiology, Development and Neuroscience, University of Cambridge, Downing Street, Cambridge, CB2 3EG, UK
| | - Owen R Vaughan
- Department of Physiology, Development and Neuroscience, University of Cambridge, Downing Street, Cambridge, CB2 3EG, UK
| | - Abigail L Fowden
- Department of Physiology, Development and Neuroscience, University of Cambridge, Downing Street, Cambridge, CB2 3EG, UK
| | - Christiane Albrecht
- Institute of Biochemistry and Molecular Medicine, University of Bern, Bühlstrasse 28, CH-3012, Bern, Switzerland.
- Swiss National Centre of Competence in Research (NCCR) TransCure, University of Bern, Bern, Switzerland.
| | - Amanda N Sferruzzi-Perri
- Department of Physiology, Development and Neuroscience, University of Cambridge, Downing Street, Cambridge, CB2 3EG, UK.
| |
Collapse
|
42
|
Ying Z, Yin M, Zhu Z, Shang Z, Pei Y, Liu J, Liu Q. Iron Stress Affects the Growth and Differentiation of Toxoplasma gondii. Int J Mol Sci 2024; 25:2493. [PMID: 38473741 PMCID: PMC10931281 DOI: 10.3390/ijms25052493] [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: 01/06/2024] [Revised: 02/07/2024] [Accepted: 02/14/2024] [Indexed: 03/14/2024] Open
Abstract
Iron is an indispensable nutrient for the survival of Toxoplasma gondii; however, excessive amounts can lead to toxicity. The parasite must overcome the host's "nutritional immunity" barrier and compete with the host for iron. Since T. gondii can infect most nucleated cells, it encounters increased iron stress during parasitism. This study assessed the impact of iron stress, encompassing both iron depletion and iron accumulation, on the growth of T. gondii. Iron accumulation disrupted the redox balance of T. gondii while enhancing the parasite's ability to adhere in high-iron environments. Conversely, iron depletion promoted the differentiation of tachyzoites into bradyzoites. Proteomic analysis further revealed proteins affected by iron depletion and identified the involvement of phosphotyrosyl phosphatase activator proteins in bradyzoite formation.
Collapse
Affiliation(s)
- Zhu Ying
- National Key Laboratory of Veterinary Public Health and Safety, College of Veterinary Medicine, China Agricultural University, Beijing 100083, China; (Z.Y.); (M.Y.); (Z.Z.); (Z.S.); (Y.P.); (J.L.)
- Key Laboratory of Animal Epidemiology of the Ministry of Agriculture, College of Veterinary Medicine, China Agricultural University, Beijing 100083, China
- National Animal Protozoa Laboratory, College of Veterinary Medicine, China Agricultural University, Beijing 100083, China
| | - Meng Yin
- National Key Laboratory of Veterinary Public Health and Safety, College of Veterinary Medicine, China Agricultural University, Beijing 100083, China; (Z.Y.); (M.Y.); (Z.Z.); (Z.S.); (Y.P.); (J.L.)
- Key Laboratory of Animal Epidemiology of the Ministry of Agriculture, College of Veterinary Medicine, China Agricultural University, Beijing 100083, China
- National Animal Protozoa Laboratory, College of Veterinary Medicine, China Agricultural University, Beijing 100083, China
| | - Zifu Zhu
- National Key Laboratory of Veterinary Public Health and Safety, College of Veterinary Medicine, China Agricultural University, Beijing 100083, China; (Z.Y.); (M.Y.); (Z.Z.); (Z.S.); (Y.P.); (J.L.)
- Key Laboratory of Animal Epidemiology of the Ministry of Agriculture, College of Veterinary Medicine, China Agricultural University, Beijing 100083, China
- National Animal Protozoa Laboratory, College of Veterinary Medicine, China Agricultural University, Beijing 100083, China
| | - Zheng Shang
- National Key Laboratory of Veterinary Public Health and Safety, College of Veterinary Medicine, China Agricultural University, Beijing 100083, China; (Z.Y.); (M.Y.); (Z.Z.); (Z.S.); (Y.P.); (J.L.)
- Key Laboratory of Animal Epidemiology of the Ministry of Agriculture, College of Veterinary Medicine, China Agricultural University, Beijing 100083, China
- National Animal Protozoa Laboratory, College of Veterinary Medicine, China Agricultural University, Beijing 100083, China
| | - Yanqun Pei
- National Key Laboratory of Veterinary Public Health and Safety, College of Veterinary Medicine, China Agricultural University, Beijing 100083, China; (Z.Y.); (M.Y.); (Z.Z.); (Z.S.); (Y.P.); (J.L.)
- Key Laboratory of Animal Epidemiology of the Ministry of Agriculture, College of Veterinary Medicine, China Agricultural University, Beijing 100083, China
- National Animal Protozoa Laboratory, College of Veterinary Medicine, China Agricultural University, Beijing 100083, China
| | - Jing Liu
- National Key Laboratory of Veterinary Public Health and Safety, College of Veterinary Medicine, China Agricultural University, Beijing 100083, China; (Z.Y.); (M.Y.); (Z.Z.); (Z.S.); (Y.P.); (J.L.)
- Key Laboratory of Animal Epidemiology of the Ministry of Agriculture, College of Veterinary Medicine, China Agricultural University, Beijing 100083, China
- National Animal Protozoa Laboratory, College of Veterinary Medicine, China Agricultural University, Beijing 100083, China
| | - Qun Liu
- National Key Laboratory of Veterinary Public Health and Safety, College of Veterinary Medicine, China Agricultural University, Beijing 100083, China; (Z.Y.); (M.Y.); (Z.Z.); (Z.S.); (Y.P.); (J.L.)
- Key Laboratory of Animal Epidemiology of the Ministry of Agriculture, College of Veterinary Medicine, China Agricultural University, Beijing 100083, China
- National Animal Protozoa Laboratory, College of Veterinary Medicine, China Agricultural University, Beijing 100083, China
| |
Collapse
|
43
|
Faraji P, Borchert A, Ahmadian S, Kuhn H. Butylated Hydroxytoluene (BHT) Protects SH-SY5Y Neuroblastoma Cells from Ferroptotic Cell Death: Insights from In Vitro and In Vivo Studies. Antioxidants (Basel) 2024; 13:242. [PMID: 38397840 PMCID: PMC10886092 DOI: 10.3390/antiox13020242] [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: 12/08/2023] [Revised: 02/06/2024] [Accepted: 02/13/2024] [Indexed: 02/25/2024] Open
Abstract
Ferroptosis is a special kind of programmed cell death that has been implicated in the pathogenesis of a large number of human diseases. It involves dysregulated intracellular iron metabolism and uncontrolled lipid peroxidation, which together initiate intracellular ferroptotic signalling pathways leading to cellular suicide. Pharmacological interference with ferroptotic signal transduction may prevent cell death, and thus patients suffering from ferroptosis-related diseases may benefit from such treatment. Butylated hydroxytoluene (BHT) is an effective anti-oxidant that is frequently used in oil chemistry and in cosmetics to prevent free-radical-mediated lipid peroxidation. Since it functions as a radical scavenger, it has previously been reported to interfere with ferroptotic signalling. Here, we show that BHT prevents RSL3- and ML162-induced ferroptotic cell death in cultured human neuroblastoma cells (SH-SY5Y) in a dose-dependent manner. It prevents the RSL3-induced oxidation of membrane lipids and normalises the RSL3-induced inhibition of the intracellular catalytic activity of glutathione peroxidase 4. The systemic application of BHT in a rat Alzheimer's disease model prevented the upregulation of the expression of ferroptosis-related genes. Taken together, these data indicate that BHT interferes with ferroptotic signalling in cultured neuroblastoma cells and may prevent ferroptotic cell death in an animal Alzheimer's disease model.
Collapse
Affiliation(s)
- Parisa Faraji
- Department of Biochemistry, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt Universität zu Berlin, Charitéplatz 1, 10117 Berlin, Germany; (P.F.); (A.B.)
- Institute of Biochemistry and Biophysics, University of Tehran, Tehran 1417614335, Iran
| | - Astrid Borchert
- Department of Biochemistry, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt Universität zu Berlin, Charitéplatz 1, 10117 Berlin, Germany; (P.F.); (A.B.)
| | - Shahin Ahmadian
- Institute of Biochemistry and Biophysics, University of Tehran, Tehran 1417614335, Iran
| | - Hartmut Kuhn
- Department of Biochemistry, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt Universität zu Berlin, Charitéplatz 1, 10117 Berlin, Germany; (P.F.); (A.B.)
| |
Collapse
|
44
|
Grüning NM, Ralser M. Monogenic Disorders of ROS Production and the Primary Anti-Oxidative Defense. Biomolecules 2024; 14:206. [PMID: 38397443 PMCID: PMC10887155 DOI: 10.3390/biom14020206] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2024] [Revised: 01/25/2024] [Accepted: 02/01/2024] [Indexed: 02/25/2024] Open
Abstract
Oxidative stress, characterized by an imbalance between the production of reactive oxygen species (ROS) and the cellular anti-oxidant defense mechanisms, plays a critical role in the pathogenesis of various human diseases. Redox metabolism, comprising a network of enzymes and genes, serves as a crucial regulator of ROS levels and maintains cellular homeostasis. This review provides an overview of the most important human genes encoding for proteins involved in ROS generation, ROS detoxification, and production of reduced nicotinamide adenine dinucleotide phosphate (NADPH), and the genetic disorders that lead to dysregulation of these vital processes. Insights gained from studies on inherited monogenic metabolic diseases provide valuable basic understanding of redox metabolism and signaling, and they also help to unravel the underlying pathomechanisms that contribute to prevalent chronic disorders like cardiovascular disease, neurodegeneration, and cancer.
Collapse
Affiliation(s)
- Nana-Maria Grüning
- Department of Biochemistry, Charité Universitätsmedizin Berlin, 10117 Berlin, Germany;
| | - Markus Ralser
- Department of Biochemistry, Charité Universitätsmedizin Berlin, 10117 Berlin, Germany;
- The Wellcome Centre for Human Genetics, Nuffield Department of Medicine, University of Oxford, Oxford OX3 7BN, UK
- Max Planck Institute for Molecular Genetics, 14195 Berlin, Germany
| |
Collapse
|
45
|
Wang R, Liu Y, Thabane L, Olier I, Li L, Ortega-Martorell S, Lip GYH, Li G. Relationship between trajectories of dietary iron intake and risk of type 2 diabetes mellitus: evidence from a prospective cohort study. Nutr J 2024; 23:15. [PMID: 38302934 PMCID: PMC10835921 DOI: 10.1186/s12937-024-00925-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2023] [Accepted: 01/30/2024] [Indexed: 02/03/2024] Open
Abstract
BACKGROUND The association between dietary iron intake and the risk of type 2 diabetes mellitus (T2DM) remains inconsistent. In this study, we aimed to investigate the relationship between trajectories of dietary iron intake and risk of T2DM. METHODS This study comprised a total of 61,115 participants without a prior T2DM from the UK Biobank database. We used the group-based trajectory model (GBTM) to identify different dietary iron intake trajectories. Cox proportional hazards models were used to evaluate the relationship between trajectories of dietary iron intake and risk of T2DM. RESULTS During a mean follow-up of 4.8 years, a total of 677 T2DM events were observed. Four trajectory groups of dietary iron intake were characterized by the GBTM: trajectory group 1 (with a mean dietary iron intake of 10.9 mg/day), 2 (12.3 mg/day), 3 (14.1 mg/day) and 4 (17.6 mg/day). Trajectory group 3 was significantly associated with a 38% decreased risk of T2DM when compared with trajectory group 1 (hazard ratio [HR] = 0.62, 95% confidence interval [CI]: 0.49-0.79), while group 4 was significantly related with a 30% risk reduction (HR = 0.70, 95% CI: 0.54-0.91). Significant effect modifications by obesity (p = 0.04) and history of cardiovascular disease (p < 0.01) were found to the relationship between trajectories of dietary iron intake and the risk of T2DM. CONCLUSIONS We found that trajectories of dietary iron intake were significantly associated with the risk of T2DM, where the lowest T2DM risk was observed in trajectory group 3 with a mean iron intake of 14.1 mg/day. These findings may highlight the importance of adequate dietary iron intake to the T2DM prevention from a public health perspective. Further studies to assess the relationship between dietary iron intake and risk of T2DM are needed, as well as intervention studies to mitigate the risks of T2DM associated with dietary iron changes.
Collapse
Affiliation(s)
- Ruoting Wang
- Center for Clinical Epidemiology and Methodology (CCEM), Guangdong Second Provincial General Hospital, Guangzhou, 510317, China
| | - Yingxin Liu
- Center for Clinical Epidemiology and Methodology (CCEM), Guangdong Second Provincial General Hospital, Guangzhou, 510317, China
| | - Lehana Thabane
- Department of Health Research Methods, Evidence, and Impact, McMaster University, 1280 Main St West, Hamilton, ON, L8S 4L8, Canada
| | - Ivan Olier
- Liverpool Centre for Cardiovascular Science, University of Liverpool, Liverpool John Moores University and Liverpool Heart & Chest Hospital, Liverpool, UK
- School of Computer Science and Mathematics, Liverpool John Moores University, Liverpool, UK
| | - Likang Li
- Center for Clinical Epidemiology and Methodology (CCEM), Guangdong Second Provincial General Hospital, Guangzhou, 510317, China
| | - Sandra Ortega-Martorell
- Liverpool Centre for Cardiovascular Science, University of Liverpool, Liverpool John Moores University and Liverpool Heart & Chest Hospital, Liverpool, UK
- School of Computer Science and Mathematics, Liverpool John Moores University, Liverpool, UK
| | - Gregory Y H Lip
- Liverpool Centre for Cardiovascular Science, University of Liverpool, Liverpool John Moores University and Liverpool Heart & Chest Hospital, Liverpool, UK
- Danish Center for Health Services Research, Aalborg University, Aalborg, Denmark
| | - Guowei Li
- Center for Clinical Epidemiology and Methodology (CCEM), Guangdong Second Provincial General Hospital, Guangzhou, 510317, China.
- Department of Health Research Methods, Evidence, and Impact, McMaster University, 1280 Main St West, Hamilton, ON, L8S 4L8, Canada.
- Father Sean O'Sullivan Research Centre, St Joseph's Healthcare Hamilton, 50 Charlton Ave E, Hamilton, ON, L8N 4A6, Canada.
| |
Collapse
|
46
|
Yu H, Gan D, Luo Z, Yang Q, An D, Zhang H, Hu Y, Ma Z, Zeng Q, Xu D, Ren H. α-Ketoglutarate improves cardiac insufficiency through NAD +-SIRT1 signaling-mediated mitophagy and ferroptosis in pressure overload-induced mice. Mol Med 2024; 30:15. [PMID: 38254035 PMCID: PMC10804789 DOI: 10.1186/s10020-024-00783-1] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Accepted: 01/11/2024] [Indexed: 01/24/2024] Open
Abstract
BACKGROUND In heart failure (HF), mitochondrial dysfunction and metabolic remodeling lead to a reduction in energy productivity and aggravate cardiomyocyte injury. Supplementation with α-ketoglutarate (AKG) alleviated myocardial hypertrophy and fibrosis in mice with HF and improved cardiac insufficiency. However, the myocardial protective mechanism of AKG remains unclear. We verified the hypothesis that AKG improves mitochondrial function by upregulating NAD+ levels and activating silent information regulator 2 homolog 1 (SIRT1) in cardiomyocytes. METHODS In vivo, 2% AKG was added to the drinking water of mice undergoing transverse aortic constriction (TAC) surgery. Echocardiography and biopsy were performed to evaluate cardiac function and pathological changes. Myocardial metabolomics was analyzed by liquid chromatography‒mass spectrometry (LC‒MS/MS) at 8 weeks after surgery. In vitro, the expression of SIRT1 or PINK1 proteins was inhibited by selective inhibitors and siRNA in cardiomyocytes stimulated with angiotensin II (AngII) and AKG. NAD+ levels were detected using an NAD test kit. Mitophagy and ferroptosis levels were evaluated by Western blotting, qPCR, JC-1 staining and lipid peroxidation analysis. RESULTS AKG supplementation after TAC surgery could alleviate myocardial hypertrophy and fibrosis and improve cardiac function in mice. Metabolites of the malate-aspartate shuttle (MAS) were increased, but the TCA cycle and fatty acid metabolism pathway could be inhibited in the myocardium of TAC mice after AKG supplementation. Decreased NAD+ levels and SIRT1 protein expression were observed in heart of mice and AngII-treated cardiomyocytes. After AKG treatment, these changes were reversed, and increased mitophagy, inhibited ferroptosis, and alleviated damage in cardiomyocytes were observed. When the expression of SIRT1 was inhibited by a selective inhibitor and siRNA, the protective effect of AKG was suppressed. CONCLUSION Supplementation with AKG can improve myocardial hypertrophy, fibrosis and chronic cardiac insufficiency caused by pressure overload. By increasing the level of NAD+, the SIRT-PINK1 and SIRT1-GPX4 signaling pathways are activated to promote mitophagy and inhibit ferroptosis in cardiomyocytes, which ultimately alleviates cardiomyocyte damage.
Collapse
Affiliation(s)
- Hao Yu
- State Key Laboratory of Organ Failure Research, Department of Cardiology, Nanfang Hospital, Southern Medical University, 1838 Northern Guangzhou Ave, Guangzhou, Guangdong, 510515, China
- Key Laboratory for Organ Failure Research, Ministry of Education of the People's Republic of China, 1838 Northern Guangzhou Ave, Guangzhou, Guangdong, 510515, China
| | - Daojing Gan
- State Key Laboratory of Organ Failure Research, Department of Cardiology, Nanfang Hospital, Southern Medical University, 1838 Northern Guangzhou Ave, Guangzhou, Guangdong, 510515, China
- Key Laboratory for Organ Failure Research, Ministry of Education of the People's Republic of China, 1838 Northern Guangzhou Ave, Guangzhou, Guangdong, 510515, China
| | - Zhen Luo
- State Key Laboratory of Organ Failure Research, Department of Cardiology, Nanfang Hospital, Southern Medical University, 1838 Northern Guangzhou Ave, Guangzhou, Guangdong, 510515, China
- Key Laboratory for Organ Failure Research, Ministry of Education of the People's Republic of China, 1838 Northern Guangzhou Ave, Guangzhou, Guangdong, 510515, China
| | - Qilin Yang
- State Key Laboratory of Organ Failure Research, Department of Cardiology, Nanfang Hospital, Southern Medical University, 1838 Northern Guangzhou Ave, Guangzhou, Guangdong, 510515, China
- Key Laboratory for Organ Failure Research, Ministry of Education of the People's Republic of China, 1838 Northern Guangzhou Ave, Guangzhou, Guangdong, 510515, China
| | - Dongqi An
- Key Laboratory for Organ Failure Research, Ministry of Education of the People's Republic of China, 1838 Northern Guangzhou Ave, Guangzhou, Guangdong, 510515, China
- Department of Cardiovascular Surgery, Nanfang Hospital, Southern Medical University, 1838 Northern Guangzhou Ave, Guangzhou, Guangdong, 510515, China
| | - Hao Zhang
- State Key Laboratory of Organ Failure Research, Department of Cardiology, Nanfang Hospital, Southern Medical University, 1838 Northern Guangzhou Ave, Guangzhou, Guangdong, 510515, China
- Key Laboratory for Organ Failure Research, Ministry of Education of the People's Republic of China, 1838 Northern Guangzhou Ave, Guangzhou, Guangdong, 510515, China
| | - Yingchun Hu
- State Key Laboratory of Organ Failure Research, Department of Cardiology, Nanfang Hospital, Southern Medical University, 1838 Northern Guangzhou Ave, Guangzhou, Guangdong, 510515, China
- Key Laboratory for Organ Failure Research, Ministry of Education of the People's Republic of China, 1838 Northern Guangzhou Ave, Guangzhou, Guangdong, 510515, China
| | - Zhuang Ma
- State Key Laboratory of Organ Failure Research, Department of Cardiology, Nanfang Hospital, Southern Medical University, 1838 Northern Guangzhou Ave, Guangzhou, Guangdong, 510515, China
- Key Laboratory for Organ Failure Research, Ministry of Education of the People's Republic of China, 1838 Northern Guangzhou Ave, Guangzhou, Guangdong, 510515, China
| | - Qingchun Zeng
- State Key Laboratory of Organ Failure Research, Department of Cardiology, Nanfang Hospital, Southern Medical University, 1838 Northern Guangzhou Ave, Guangzhou, Guangdong, 510515, China
- Key Laboratory for Organ Failure Research, Ministry of Education of the People's Republic of China, 1838 Northern Guangzhou Ave, Guangzhou, Guangdong, 510515, China
| | - Dingli Xu
- State Key Laboratory of Organ Failure Research, Department of Cardiology, Nanfang Hospital, Southern Medical University, 1838 Northern Guangzhou Ave, Guangzhou, Guangdong, 510515, China.
- Key Laboratory for Organ Failure Research, Ministry of Education of the People's Republic of China, 1838 Northern Guangzhou Ave, Guangzhou, Guangdong, 510515, China.
| | - Hao Ren
- Key Laboratory for Organ Failure Research, Ministry of Education of the People's Republic of China, 1838 Northern Guangzhou Ave, Guangzhou, Guangdong, 510515, China.
- Department of Rheumatology, Nanfang Hospital, Southern Medical University, 1838 Northern Guangzhou Ave, Guangzhou, Guangdong, 510515, China.
| |
Collapse
|
47
|
Confalonieri F, Ferraro V, Barone G, Di Maria A, Petrovski BÉ, Vallejo Garcia JL, Randazzo A, Vinciguerra P, Lumi X, Petrovski G. Outcomes in the Treatment of Subretinal Macular Hemorrhage Secondary to Age-Related Macular Degeneration: A Systematic Review. J Clin Med 2024; 13:367. [PMID: 38256501 PMCID: PMC10816885 DOI: 10.3390/jcm13020367] [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: 11/27/2023] [Revised: 12/29/2023] [Accepted: 01/04/2024] [Indexed: 01/24/2024] Open
Abstract
Background: Subretinal macular hemorrhage (SRMH) secondary to age-related macular degeneration (AMD) is a relatively rare condition in ophthalmology characterized by blood collection between the neurosensory retina and the retinal pigment epithelium (RPE). Without prompt treatment, visual prognosis is poor. A plethora of treatment approaches have been tried over the past years ranging from intravitreal anti-vascular endothelial growth factor (anti-VEGF) monotherapy to direct subretinal surgery, with no conclusive superiority of one over the other. Materials and Methods: We conducted a systematic review of the outcomes and treatment modalities of SRMH from inception to 14 June 2022, following the Preferred Reporting Items for Systematic Reviews and Meta-Analyses guidelines (PRISMA). The level of evidence was assessed for all included articles according to the quality of evidence according to the Grading of Recommendations Assessment, Development and Evaluation (GRADE) system. Results: A total of 2745 articles were initially extracted, out of which 1654 articles were obtained after duplicates were removed and their abstracts screened. A total of 155 articles were included for full-text review. Finally, 81 articles remained that fulfilled the inclusion criteria. Conclusions: Even though there are solid results supporting a variety of treatments for SRMH, the best treatment modality has still not been conclusively demonstrated and further research is needed.
Collapse
Affiliation(s)
- Filippo Confalonieri
- Department of Ophthalmology, IRCCS Humanitas Research Hospital, Rozzano, 20089 Milan, Italy; (V.F.); (G.B.); (A.D.M.); (J.L.V.G.); (P.V.)
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, 20090 Milan, Italy
- Center for Eye Research and Innovative Diagnostics, Department of Ophthalmology, Institute for Clinical Medicine, University of Oslo, Kirkeveien 166, 0450 Oslo, Norway; (B.É.P.); (X.L.)
- Department of Ophthalmology, Oslo University Hospital, Kirkeveien 166, 0450 Oslo, Norway
| | - Vanessa Ferraro
- Department of Ophthalmology, IRCCS Humanitas Research Hospital, Rozzano, 20089 Milan, Italy; (V.F.); (G.B.); (A.D.M.); (J.L.V.G.); (P.V.)
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, 20090 Milan, Italy
| | - Gianmaria Barone
- Department of Ophthalmology, IRCCS Humanitas Research Hospital, Rozzano, 20089 Milan, Italy; (V.F.); (G.B.); (A.D.M.); (J.L.V.G.); (P.V.)
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, 20090 Milan, Italy
| | - Alessandra Di Maria
- Department of Ophthalmology, IRCCS Humanitas Research Hospital, Rozzano, 20089 Milan, Italy; (V.F.); (G.B.); (A.D.M.); (J.L.V.G.); (P.V.)
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, 20090 Milan, Italy
| | - Beáta Éva Petrovski
- Center for Eye Research and Innovative Diagnostics, Department of Ophthalmology, Institute for Clinical Medicine, University of Oslo, Kirkeveien 166, 0450 Oslo, Norway; (B.É.P.); (X.L.)
| | - Josè Luis Vallejo Garcia
- Department of Ophthalmology, IRCCS Humanitas Research Hospital, Rozzano, 20089 Milan, Italy; (V.F.); (G.B.); (A.D.M.); (J.L.V.G.); (P.V.)
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, 20090 Milan, Italy
| | - Alessandro Randazzo
- Department of Ophthalmology, IRCCS Humanitas Research Hospital, Rozzano, 20089 Milan, Italy; (V.F.); (G.B.); (A.D.M.); (J.L.V.G.); (P.V.)
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, 20090 Milan, Italy
| | - Paolo Vinciguerra
- Department of Ophthalmology, IRCCS Humanitas Research Hospital, Rozzano, 20089 Milan, Italy; (V.F.); (G.B.); (A.D.M.); (J.L.V.G.); (P.V.)
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, 20090 Milan, Italy
| | - Xhevat Lumi
- Center for Eye Research and Innovative Diagnostics, Department of Ophthalmology, Institute for Clinical Medicine, University of Oslo, Kirkeveien 166, 0450 Oslo, Norway; (B.É.P.); (X.L.)
- Eye Hospital, University Medical Centre Ljubljana, Zaloška Cesta 2, 1000 Ljubljana, Slovenia
| | - Goran Petrovski
- Center for Eye Research and Innovative Diagnostics, Department of Ophthalmology, Institute for Clinical Medicine, University of Oslo, Kirkeveien 166, 0450 Oslo, Norway; (B.É.P.); (X.L.)
- Department of Ophthalmology, Oslo University Hospital, Kirkeveien 166, 0450 Oslo, Norway
- Department of Ophthalmology, University of Split School of Medicine and University Hospital Centre, 21000 Split, Croatia
- UKLONetwork, University St. Kliment Ohridski-Bitola, 7000 Bitola, North Macedonia
| |
Collapse
|
48
|
Lim J, Lee J, Boo Y, Kim WJ. A polymeric iron oxide nanocomplex loaded with sulfasalazine: an approach for inducing ferritinophagy-assisted ferroptosis for anti-cancer therapy. NANOSCALE 2024; 16:742-751. [PMID: 38086680 DOI: 10.1039/d3nr04733d] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2024]
Abstract
The approach of using ferroptosis to treat cancer has garnered attention due to its promising potential. However, the effectiveness of this therapy is often limited by the inherent redox system in cancer cells and the presence of ferritin as an iron ion storage molecule. To address this issue, we have designed a polymeric iron oxide nanocomplex loaded with sulfasalazine as a ferritinophagy-assisted ferroptosis inducing agent. The nanocomplex is based on a pH-responsive drug releasing platform that enables improved ferroptosis anti-cancer therapy. The nanocomplex was synthesized using polymerized phenylboronic acid decorated with iron oxide and further loaded with sulfasalazine by interacting with polymerized phenylboronic acid. Upon entering cancer cells, the nanocomplex releases sulfasalazine at the lysosomal acidic pH, which causes the complex to degrade into the labile iron ion (Fe2+). This process inhibits the production of GSH and reproduces the labile iron ion by degrading ferritin. As a result, an excess iron ion pool is formed, and the facilitated Fenton reaction induces an improved ferroptosis anti-cancer effect. Moreover, our research has demonstrated that the nanocomplex effectively regresses tumors, thereby representing significant inhibition of tumor growth using in vivo models. We believe that this ferritinophagy-assisted ferroptosis strategy using the nanocomplex provides a promising solution for iron-based anti-cancer therapy.
Collapse
Affiliation(s)
- Junha Lim
- Department of Chemistry, POSTECH-CATHOLIC Biomedical Engineering Institute, Pohang University of Science and Technology (POSTECH), Pohang 37673, Republic of Korea.
| | - Junseok Lee
- OmniaMed Co, Ltd, Pohang 37673, Republic of Korea
| | - Yeonju Boo
- Department of Chemistry, POSTECH-CATHOLIC Biomedical Engineering Institute, Pohang University of Science and Technology (POSTECH), Pohang 37673, Republic of Korea.
| | - Won Jong Kim
- Department of Chemistry, POSTECH-CATHOLIC Biomedical Engineering Institute, Pohang University of Science and Technology (POSTECH), Pohang 37673, Republic of Korea.
- OmniaMed Co, Ltd, Pohang 37673, Republic of Korea
| |
Collapse
|
49
|
Zhang Y, Roux C, Rouchaud A, Meddahi-Pellé A, Gueguen V, Mangeney C, Sun F, Pavon-Djavid G, Luo Y. Recent advances in Fe-based bioresorbable stents: Materials design and biosafety. Bioact Mater 2024; 31:333-354. [PMID: 37663617 PMCID: PMC10474570 DOI: 10.1016/j.bioactmat.2023.07.024] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2023] [Revised: 07/27/2023] [Accepted: 07/27/2023] [Indexed: 09/05/2023] Open
Abstract
Fe-based materials have received more and more interests in recent years as candidates to fabricate bioresorbable stents due to their appropriate mechanical properties and biocompatibility. However, the low degradation rate of Fe is a serious limitation for such application. To overcome this critical issue, many efforts have been devoted to accelerate the corrosion rate of Fe-based stents, through the structural and surface modification of Fe matrix. As stents are implantable devices, the released corrosion products (Fe2+ ions) in vessels may alter the metabolism, by generating reactive oxygen species (ROS), which might in turn impact the biosafety of Fe-based stents. These considerations emphasize the importance of combining knowledge in both materials and biological science for the development of efficient and safe Fe-based stents, although there are still only limited numbers of reviews regarding this interdisciplinary field. This review aims to provide a concise overview of the main strategies developed so far to design Fe-based stents with accelerated degradation, highlighting the fundamental mechanisms of corrosion and the methods to study them as well as the reported approaches to accelerate the corrosion rates. These approaches will be divided into four main sections, focusing on (i) increased active surface areas, (ii) tailored microstructures, (iii) creation of galvanic reactions (by alloying, ion implantation or surface coating of noble metals) and (iv) decreased local pH induced by degradable surface organic layers. Recent advances in the evaluation of the in vitro biocompatibility of the final materials and ongoing in vivo tests are also provided.
Collapse
Affiliation(s)
- Yang Zhang
- Université Paris Cité, CNRS, Laboratoire de Chimie et de Biochimie Pharmacologiques et Toxicologiques, F-75006, Paris, France
- Université Sorbonne Paris Nord, INSERM U1148, Laboratory for Vascular Translational Science, Cardiovascular Bioengineering, 99 Av. Jean-Baptiste Clément, 93430, Villetaneuse, France
| | - Charles Roux
- Univ. Limoges, CNRS, XLIM, UMR 7252, Limoges, France
| | | | - Anne Meddahi-Pellé
- Université Sorbonne Paris Nord, INSERM U1148, Laboratory for Vascular Translational Science, Cardiovascular Bioengineering, 99 Av. Jean-Baptiste Clément, 93430, Villetaneuse, France
| | - Virginie Gueguen
- Université Sorbonne Paris Nord, INSERM U1148, Laboratory for Vascular Translational Science, Cardiovascular Bioengineering, 99 Av. Jean-Baptiste Clément, 93430, Villetaneuse, France
| | - Claire Mangeney
- Université Paris Cité, CNRS, Laboratoire de Chimie et de Biochimie Pharmacologiques et Toxicologiques, F-75006, Paris, France
| | - Fan Sun
- PSL Université, Chimie Paris Tech, IRCP, CNRS UMR 8247, 11, Rue Pierre et Marie Curie, 75005, Paris, France
| | - Graciela Pavon-Djavid
- Université Sorbonne Paris Nord, INSERM U1148, Laboratory for Vascular Translational Science, Cardiovascular Bioengineering, 99 Av. Jean-Baptiste Clément, 93430, Villetaneuse, France
| | - Yun Luo
- Université Paris Cité, CNRS, Laboratoire de Chimie et de Biochimie Pharmacologiques et Toxicologiques, F-75006, Paris, France
| |
Collapse
|
50
|
Duarte TL, Lopes M, Oliveira M, Santos AG, Vasco C, Reis JP, Antunes AR, Gonçalves A, Chacim S, Oliveira C, Porto B, Teles MJ, Moreira AC, Silva AMN, Schwessinger R, Drakesmith H, Henrique R, Porto G, Duarte D. Iron overload induces dysplastic erythropoiesis and features of myelodysplasia in Nrf2-deficient mice. Leukemia 2024; 38:96-108. [PMID: 37857886 DOI: 10.1038/s41375-023-02067-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Revised: 10/04/2023] [Accepted: 10/09/2023] [Indexed: 10/21/2023]
Abstract
Iron overload (IOL) is hypothesized to contribute to dysplastic erythropoiesis. Several conditions, including myelodysplastic syndrome, thalassemia and sickle cell anemia, are characterized by ineffective erythropoiesis and IOL. Iron is pro-oxidant and may participate in the pathophysiology of these conditions by increasing genomic instability and altering the microenvironment. There is, however, lack of in vivo evidence demonstrating a role of IOL and oxidative damage in dysplastic erythropoiesis. NRF2 transcription factor is the master regulator of antioxidant defenses, playing a crucial role in the cellular response to IOL in the liver. Here, we crossed Nrf2-/- with hemochromatosis (Hfe-/-) or hepcidin-null (Hamp1-/-) mice. Double-knockout mice developed features of ineffective erythropoiesis and myelodysplasia including macrocytic anemia, splenomegaly, and accumulation of immature dysplastic bone marrow (BM) cells. BM cells from Nrf2/Hamp1-/- mice showed increased in vitro clonogenic potential and, upon serial transplantation, recipients disclosed cytopenias, despite normal engraftment, suggesting defective differentiation. Unstimulated karyotype analysis showed increased chromosome instability and aneuploidy in Nrf2/Hamp1-/- BM cells. In HFE-related hemochromatosis patients, NRF2 promoter SNP rs35652124 genotype TT (predicted to decrease NRF2 expression) associated with increased MCV, consistent with erythroid dysplasia. Our results suggest that IOL induces ineffective erythropoiesis and dysplastic hematologic features through oxidative damage in Nrf2-deficient cells.
Collapse
Affiliation(s)
- Tiago L Duarte
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal.
- IBMC - Instituto de Biologia Molecular e Celular, Universidade do Porto, Porto, Portugal.
| | - Marta Lopes
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
- IBMC - Instituto de Biologia Molecular e Celular, Universidade do Porto, Porto, Portugal
| | - Mónica Oliveira
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
- IBMC - Instituto de Biologia Molecular e Celular, Universidade do Porto, Porto, Portugal
| | - Ana G Santos
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
- IBMC - Instituto de Biologia Molecular e Celular, Universidade do Porto, Porto, Portugal
| | - Catarina Vasco
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
- IBMC - Instituto de Biologia Molecular e Celular, Universidade do Porto, Porto, Portugal
| | - Joana P Reis
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
- IBMC - Instituto de Biologia Molecular e Celular, Universidade do Porto, Porto, Portugal
- Instituto de Ciências Biomédicas Abel Salazar (ICBAS), Universidade do Porto, Porto, Portugal
| | - Ana Rita Antunes
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
- IBMC - Instituto de Biologia Molecular e Celular, Universidade do Porto, Porto, Portugal
| | - Andreia Gonçalves
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
- IBMC - Instituto de Biologia Molecular e Celular, Universidade do Porto, Porto, Portugal
| | - Sérgio Chacim
- Serviço de Hematologia e Transplantação de Medula Óssea, Instituto Português de Oncologia do Porto Francisco Gentil, E.P.E. (IPO Porto), Porto, Portugal
| | - Cláudia Oliveira
- Laboratório de Citogenética, Instituto de Ciências Biomédicas Abel Salazar (ICBAS), Universidade do Porto, Porto, Portugal
| | - Beatriz Porto
- Laboratório de Citogenética, Instituto de Ciências Biomédicas Abel Salazar (ICBAS), Universidade do Porto, Porto, Portugal
| | - Maria José Teles
- Departmento de Patologia Clínica, Centro Hospitalar Universitário São João, Porto, Portugal
| | - Ana C Moreira
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
- IBMC - Instituto de Biologia Molecular e Celular, Universidade do Porto, Porto, Portugal
- Instituto de Ciências Biomédicas Abel Salazar (ICBAS), Universidade do Porto, Porto, Portugal
| | - André M N Silva
- LAQV-REQUIMTE, Departamento de Química e Bioquímica, Faculdade de Ciências, Universidade do Porto, Porto, Portugal
- LAQV-REQUIMTE, Instituto de Ciências Biomédicas Abel Salazar (ICBAS), Universidade do Porto, Porto, Portugal
| | - Ron Schwessinger
- MRC Molecular Haematology Unit, MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, UK
- MRC WIMM Centre for Computational Biology, MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, UK
| | - Hal Drakesmith
- MRC Translational Immune Discovery Unit, MRC Weatherall Institute of Molecular Medicine, University of Oxford, John Radcliffe Hospital, Oxford, UK
| | - Rui Henrique
- Serviço de Anatomia Patológica, IPO Porto, Porto, Portugal
- Departamento de Patologia e Imunologia Molecular, Instituto de Ciências Biomédicas Abel Salazar (ICBAS), Universidade do Porto, Porto, Portugal
| | - Graça Porto
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
- IBMC - Instituto de Biologia Molecular e Celular, Universidade do Porto, Porto, Portugal
- Departamento de Patologia e Imunologia Molecular, Instituto de Ciências Biomédicas Abel Salazar (ICBAS), Universidade do Porto, Porto, Portugal
- Serviço de Imuno-hemoterapia, Centro Hospitalar Universitário de Santo António (CHUdSA), Porto, Portugal
| | - Delfim Duarte
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal.
- Serviço de Hematologia e Transplantação de Medula Óssea, Instituto Português de Oncologia do Porto Francisco Gentil, E.P.E. (IPO Porto), Porto, Portugal.
- Departmento de Biomedicina, Faculdade de Medicina da Universidade do Porto (FMUP), Porto, Portugal.
- P.CCC - Porto Comprehensive Cancer Center Raquel Seruca, Porto, Portugal.
| |
Collapse
|