1
|
Udupi A, Shetty S, Aranjani JM, Kumar R, Bharati S. Anticancer therapeutic potential of multimodal targeting agent- "phosphorylated galactosylated chitosan coated magnetic nanoparticles" against N-nitrosodiethylamine-induced hepatocellular carcinoma. Drug Deliv Transl Res 2025; 15:1023-1042. [PMID: 38990437 PMCID: PMC11782354 DOI: 10.1007/s13346-024-01655-1] [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: 06/19/2024] [Indexed: 07/12/2024]
Abstract
Superparamagnetic iron oxide nanoparticles (SPIONs) are extensively used as carriers in targeted drug delivery and has several advantages in the field of magnetic hyperthermia, chemodynamic therapy and magnet assisted radionuclide therapy. The characteristics of SPIONs can be tailored to deliver drugs into tumor via "passive targeting" and they can also be coated with tissue-specific agents to enhance tumor uptake via "active targeting". In our earlier studies, we developed HCC specific targeting agent- "phosphorylated galactosylated chitosan"(PGC) for targeting asialoglycoprotein receptors. Considering their encouraging results, in this study we developed a multifunctional targeting system- "phosphorylated galactosylated chitosan-coated magnetic nanoparticles"(PGCMNPs) for targeting HCC. PGCMNPs were synthesized by co-precipitation method and characterized by DLS, XRD, TEM, VSM, elemental analysis and FT-IR spectroscopy. PGCMNPs were evaluated for in vitro antioxidant properties, uptake in HepG2 cells, biodistribution, in vivo toxicity and were also evaluated for anticancer therapeutic potential against NDEA-induced HCC in mice model in terms of tumor status, electrical properties, antioxidant defense status and apoptosis. The characterization studies confirmed successful formation of PGCMNPs with superparamagnetic properties. The internalization studies demonstrated (99-100)% uptake of PGCMNPs in HepG2 cells. These results were also supported by biodistribution studies in which increased iron content (296%) was noted inside the hepatocytes. Further, PGCMNPs exhibited no in vivo toxicity. The anticancer therapeutic potential was evident from observation that PGCMNPs treatment decreased tumor bearing animals (41.6%) and significantly (p ≤ 0.05) lowered tumor multiplicity. Overall, this study indicated that PGCMNPs with improved properties are efficiently taken-up by hepatoma cells and has therapeutic potential against HCC. Further, this agent can be tagged with 32P and hence can offer multimodal cancer treatment options via radiation ablation as well as magnetic hyperthermia.
Collapse
Affiliation(s)
- Anushree Udupi
- Department of Nuclear Medicine, Manipal College of Health Professions, Manipal Academy of Higher Education, Manipal, 576104, Karnataka, India
| | - Sachin Shetty
- Department of Nuclear Medicine, Manipal College of Health Professions, Manipal Academy of Higher Education, Manipal, 576104, Karnataka, India
| | - Jesil Mathew Aranjani
- Department of Pharmaceutical Biotechnology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, 576104, Karnataka, India
| | - Rajesh Kumar
- Department of Nuclear Medicine, All India Institute of Medical Sciences, Jodhpur, 342005, Rajasthan, India
| | - Sanjay Bharati
- Department of Nuclear Medicine, Manipal College of Health Professions, Manipal Academy of Higher Education, Manipal, 576104, Karnataka, India.
| |
Collapse
|
2
|
Sangokoya C. The FIRE biosensor illuminates iron regulatory protein activity and cellular iron homeostasis. CELL REPORTS METHODS 2025; 5:100960. [PMID: 39824193 PMCID: PMC11840943 DOI: 10.1016/j.crmeth.2024.100960] [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: 10/21/2024] [Revised: 11/18/2024] [Accepted: 12/20/2024] [Indexed: 01/20/2025]
Abstract
On Earth, iron is abundant, bioavailable, and crucial for initiating the first catalytic reactions of life from prokaryotes to plants to mammals. Iron-complexed proteins are critical to biological pathways and essential cellular functions. While it is well known that the regulation of iron is necessary for mammalian development, little is known about the timeline of how specific transcripts network and interact in response to cellular iron regulation to shape cell fate, function, and plasticity in the developing embryo and beyond. Here, we present a ratiometric genetically encoded dual biosensor called FIRE (Fe-IRE [iron-responsive element]) to evaluate iron regulatory protein (IRP)-binding activity and cellular iron status in live cells, allowing for the study and dissection of dynamic changes in cellular iron and IRP activity over developmental time. FIRE reveals a previously unrecognized foundational timeline of IRP activity and cellular iron homeostasis during stem cell pluripotency transition and early differentiation.
Collapse
Affiliation(s)
- Carolyn Sangokoya
- Department of Pathology, University of California, San Francisco, San Francisco, CA, USA; The Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, CA, USA.
| |
Collapse
|
3
|
Ostertag F, Grimm VJ, Hinrichs J. Iron saturation and binding capacity of lactoferrin - development and validation of a colorimetric protocol for quality control. Food Chem 2025; 463:141365. [PMID: 39332363 DOI: 10.1016/j.foodchem.2024.141365] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Revised: 09/06/2024] [Accepted: 09/18/2024] [Indexed: 09/29/2024]
Abstract
Among the numerous biofunctional properties of lactoferrin, its ability to bind iron ions can be considered a core function. The saturation level with ferric iron affects the stability and functionality of the protein. To reliably quantify the iron saturation, an assay based on the color reagent Ferrozine was developed and validated concerning the lower detection (0.023 μg mL-1) and quantification limits (0.069 μg mL-1), as well as precision, recovery and accuracy values. The established assay was used to monitor iron uptake, comparing two commercially available bovine lactoferrin powders. Significant differences between the samples were observed. One sample exhibited nearly ideal binding behavior with a high affinity for ferric iron (saturation > 98 %), while the comparison sample did not exceed saturation values >80 %. This finding underscores the importance of assessing the iron status and binding capacity for the quality evaluation of lactoferrin products.
Collapse
Affiliation(s)
- Fabian Ostertag
- University of Hohenheim, Institute of Food Science and Biotechnology, Department of Soft Matter Science and Dairy Technology, Garbenstrasse 21, 70599 Stuttgart, Germany.
| | - Vanessa J Grimm
- University of Hohenheim, Institute of Food Science and Biotechnology, Department of Soft Matter Science and Dairy Technology, Garbenstrasse 21, 70599 Stuttgart, Germany
| | - Jörg Hinrichs
- University of Hohenheim, Institute of Food Science and Biotechnology, Department of Soft Matter Science and Dairy Technology, Garbenstrasse 21, 70599 Stuttgart, Germany
| |
Collapse
|
4
|
Ghosh S, Chigicherla KV, Dasgupta S, Goto Y, Mukherjee B. Oxidative stress-driven enhanced iron production and scavenging through Ferroportin reorientation worsens anemia in antimony-resistant Leishmania donovani infection. PLoS Pathog 2025; 21:e1012858. [PMID: 39888953 PMCID: PMC11785346 DOI: 10.1371/journal.ppat.1012858] [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: 04/22/2024] [Accepted: 12/23/2024] [Indexed: 02/02/2025] Open
Abstract
Despite the withdrawal of pentavalent-antimonials in treating Visceral leishmaniasis from India, recent clinical isolates of Leishmania donovani (LD) exhibit unresponsiveness towards pentavalent-antimony (LD-R). This antimony-unresponsiveness points towards a genetic adaptation that underpins LD-R's evolutionary persistence and dominance over sensitive counterparts (LD-S). This study highlights how LD evolutionarily tackled antimony exposure and gained increased potential of scavenging host-iron within its parasitophorous vacuoles (PV) to support its aggressive proliferation. Even though anti-leishmanial activity of pentavalent antimonials relies on triggering oxidative outburst, LD-R exhibits a surprising strategy of promoting reactive oxygen species (ROS) generation in infected macrophages. An inherent metabolic shift from glycolysis to Pentose Phosphate shunt allows LD-R to withstand elevated ROS by sustaining heightened levels of NADPH. Elevated ROS levels on the other hand trigger excess iron production, and LD-R capitalizes on this surplus iron by selectively reshuffling macrophage-surface iron exporter, Ferroportin, around its PV thereby gaining a survival edge as a heme-auxotroph. Higher iron utilization by LD-R leads to subsequent iron insufficiency, compensated by increased erythrophagocytosis through the breakdown of SIRPα-CD47 surveillance, orchestrated by a complex interplay of two proteases, Furin and ADAM10. Understanding these mechanisms is crucial for managing LD-R-infections and their associated complications like severe anemia, and may also provide valuable mechanistic insights into understanding drug unresponsiveness developed in other intracellular pathogens that rely on host iron.
Collapse
Affiliation(s)
- Souradeepa Ghosh
- School of Medical Science and Technology, Indian Institute of Technology, Kharagpur, West Bengal, India
| | | | - Shirin Dasgupta
- Dr B C Roy Multispeciality Medical Research Centre, Indian Institute of Technology, Kharagpur, West Bengal, India
| | - Yasuyuki Goto
- Laboratory of Molecular Immunology, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Tokyo, Japan
| | - Budhaditya Mukherjee
- School of Medical Science and Technology, Indian Institute of Technology, Kharagpur, West Bengal, India
| |
Collapse
|
5
|
Zhao YC, Wu LF, Wu SC. Amended Ferrozine Assay for Quantifying Magnetosome Iron Content in Magnetotactic Bacteria. ACS OMEGA 2024; 9:50650-50659. [PMID: 39741826 PMCID: PMC11683628 DOI: 10.1021/acsomega.4c08607] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/19/2024] [Revised: 11/28/2024] [Accepted: 12/02/2024] [Indexed: 01/03/2025]
Abstract
Magnetospirillum gryphiswaldense MSR-1 can biomineralize the magnetosome, nanoscale magnetite (Fe3O4) surrounded by a lipid bilayer, inside the cell. The magnetosome chain(s) enables MSR-1 to move along with the magnetic field (magnetoaerotaxis). Due to its unique characteristics, MSR-1 has attracted attention for biotechnological applications. During cultivation, not only the optical density but also the magnetosome content in MSR-1 should be monitored. The ferrozine assay had been utilized to quantify the iron content in magnetosomes. However, the effectiveness of the ferrozine assay on iron oxide nanoparticles is still unknown. Here, we examined the experimental factors, and the amended ferrozine assay demonstrates a recovery of 88.71% for Fe2O3 nanoparticles relative to the stock solution. Next, we apply the assay to analyze MSR-1 samples, which successfully reveals the difference in iron contents between magnetic and nonmagnetic MSR-1 samples and highlights the amount of MSR-1 cell density suitable for amended ferrozine assay. The assay further helps us examine the effects of centrifugation compared to magnetic separation (MS). The detection of residual magnetosomes in the supernatant indicates that MS remains a suitable method for collecting magnetosomes. We anticipate the amended ferrozine assay will facilitate research on MSR-1 by enabling investigators to measure iron content in cells in a fast, easy, and cost-effective manner.
Collapse
Affiliation(s)
- Ya-Chun Zhao
- Department of Environmental
Engineering, National Chung Hsing University, 145 Xingda Road, Taichung 40227, Taiwan
| | - Li-Fen Wu
- Department of Environmental
Engineering, National Chung Hsing University, 145 Xingda Road, Taichung 40227, Taiwan
| | - Siang Chen Wu
- Department of Environmental
Engineering, National Chung Hsing University, 145 Xingda Road, Taichung 40227, Taiwan
| |
Collapse
|
6
|
Khazaei M, Ardeshir RA. Protective effects of sulfated polysaccharides from Enteromorpha intestinalis on oxidative stress, liver iron overload and Ferroptosis in Zebra fish exposed to ethanol. Biomed Pharmacother 2024; 181:117715. [PMID: 39615168 DOI: 10.1016/j.biopha.2024.117715] [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: 08/29/2024] [Revised: 11/13/2024] [Accepted: 11/25/2024] [Indexed: 12/21/2024] Open
Abstract
The study investigates the protective effects of sulfated polysaccharides extracted from Enteromorpha intestinalis (EIP) against oxidative stress, liver iron overload, and ferroptosis in zebrafish exposed to ethanol, a model for alcohol-related liver disease (ALD). The extracted polysaccharides were characterized for sulfate and sugar content, molecular weight, and functional groups. Adult male zebrafish were divided into three groups: control, ethanol-exposed (EE) (0.2 % ethanol (v/v) in the water), and ethanol-exposed with EIP supplementation (1 % EIP incorporated into the basal diet) (EE+EIP) for 30 days. The study measured liver oxidative stress indexes, serum enzymological indexes, liver and serum lipid profiles, liver iron ion content, and expression of ferroptosis-related genes. Histological analysis was conducted to assess lipid accumulation and iron deposition in liver tissues. The findings indicate that EIP supplementation significantly mitigates ethanol-induced liver damage. Specifically, EIP reduced malondialdehyde levels, increased antioxidant enzyme and non-enzymatic antioxidant activity, and decreased iron ion accumulation and the area of iron granules in the liver tissue. Additionally, EIP treatment lowered lipids levels and aminotransferase enzyme activity in the serum. In the ALD model, EIP inhibited ethanol-induced ferroptosis by modulating the expression of key genes: it decreased the expression of transferrin (tf), transferrin receptor (tfr), ferroportin (fpn), and ferritin heavy chain (fth), while increasing the expression of glutathione peroxidase 4 (gpx4) and solute carrier family 7 member 11 (slc7a11). EIP has protective effects against ethanol-induced liver injury in zebrafish, offering a foundation for further research into its hepatoprotective action and potential application in preventing and treating ALD.
Collapse
Affiliation(s)
- Marziyeh Khazaei
- Institute of Pharmacy, Sechenov First Moscow State Medical University, Moscow, Russian Federation.
| | - Rashid Alijani Ardeshir
- Marine Biotechnology Department, College of Biotechnology, Amol University of Special Modern Technologies, Amol, Iran.
| |
Collapse
|
7
|
Bręborowicz A, Umezawa K. Effect of NF-κβ inhibitor - dehydroxymethylepoxyquinomicin on iron isomaltoside toxicity toward peritoneal mesothelial cells. Ren Fail 2024; 46:2304647. [PMID: 38240039 PMCID: PMC10802802 DOI: 10.1080/0886022x.2024.2304647] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2023] [Accepted: 01/08/2024] [Indexed: 01/23/2024] Open
Affiliation(s)
- Andrzej Bręborowicz
- Department of Pathophysiology, Poznan University Medical School, Poznan, Poland
- Collegium Medicum, University of Zielona Góra, Góra, Poland
| | - Kazuo Umezawa
- Department of Molecular Target Medicine Screening, Aichi Medical University School of Medicine, Nagakute, Japan
| |
Collapse
|
8
|
Manikanta, NaveenKumar SK, Thushara RM, Hemshekhar M, Sumedini ML, Sunitha K, Kemparaju K, Girish KS. Counteraction of unconjugated bilirubin against heme-induced toxicity in platelets. Thromb Res 2024; 244:109199. [PMID: 39467509 DOI: 10.1016/j.thromres.2024.109199] [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: 05/06/2024] [Revised: 10/09/2024] [Accepted: 10/22/2024] [Indexed: 10/30/2024]
Abstract
Platelets are essential for normal hemostasis and thrombosis but become hyperactive in hemolytic disorders. Cell-free heme is known to be toxic to platelets and endothelial cells, playing a significant role in the progression of pathological complications in various hemolytic conditions. The abnormal activation of circulatory platelets results in micro/macrovascular thrombosis and clot formation in the lungs, worsening the disease. This work aimed to establish the potent bioactive molecule that can regulate the heme-induced toxicity in platelets. We found that unconjugated bilirubin (UCB), an endogenous antioxidant and a byproduct of heme degradation, exhibited a higher protective effect against hemin-induced platelet aggregation and activation. This protective effect could mainly be due to reducing ROS and lipid peroxidation-mediated ferroptosis in hemin-treated platelets. Further experiments suggested that by blocking the interaction between hemin and the CLEC-2 receptor, UCB regulates the downstream Syk phosphorylation, a key event in hemin-induced platelet toxicity. Thus, UCB is emerging as a natural regulatory molecule that mitigates hemin-induced platelet toxicity and holds promise as an adjunctive therapy for managing platelet-associated complications, particularly in hemolytic disorders.
Collapse
Affiliation(s)
- Manikanta
- Department of Studies in Biochemistry, University of Mysore, Manasagangotri, Mysuru 570006, India
| | | | - Ram M Thushara
- Department of Studies in Biochemistry, University of Mysore, Manasagangotri, Mysuru 570006, India
| | - Mahadevappa Hemshekhar
- Department of Studies in Biochemistry, University of Mysore, Manasagangotri, Mysuru 570006, India
| | - Mysuru L Sumedini
- Department of Studies in Biochemistry, University of Mysore, Manasagangotri, Mysuru 570006, India
| | - Kabburahalli Sunitha
- Department of Studies and Research in Biochemistry, Tumkur University, Tumakuru 572103, India
| | - Kempaiah Kemparaju
- Department of Studies in Biochemistry, University of Mysore, Manasagangotri, Mysuru 570006, India.
| | - Kesturu S Girish
- Department of Studies and Research in Biochemistry, Tumkur University, Tumakuru 572103, India.
| |
Collapse
|
9
|
Sarkar AR, Mukherjee N, Sarkar AK, Jana NR. Designing Nano-Hemin for Ferroptosis-Mediated Cell Death via Enzymatic Hemin Digestion. ACS APPLIED MATERIALS & INTERFACES 2024; 16:64628-64637. [PMID: 39552348 DOI: 10.1021/acsami.4c17763] [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: 11/19/2024]
Abstract
Hemin is a protoporphyrin complex of ferric ion which catalyzes H2O2 degradation and produces reactive oxygen species (ROS). This ROS generation property induces oxidative stress to hemin-exposed cells that can lead to various situations such as intracellular Fenton reaction, ferroptosis, or autophagy. Therapeutic performance of hemin is hindered due to low bioavailability of the active monomeric form with an intact ROS generation property. Here, we demonstrate a colloidal nanoparticle form of hemin (nano-hemin) with a high ROS generation property and high cell uptake property. We have shown that nano-hemin produces ROS inside a cell that upregulate heme oxygenase-1 in order to metabolize hemin. This leads to the ferroptosis-mediated cell death. Furthermore, we show that the ROS generation property of nano-hemin can be modulated to control hemin cytotoxicity for either ferroptosis or autophagy. Our findings suggest that nano-hemin can be designed with modular cytotoxicity for different therapeutic applications.
Collapse
Affiliation(s)
- Abu Raihan Sarkar
- School of Materials Science, Indian Association for the Cultivation of Science, Kolkata 700 032, India
| | - Nayana Mukherjee
- School of Materials Science, Indian Association for the Cultivation of Science, Kolkata 700 032, India
| | - Ankan Kumar Sarkar
- School of Materials Science, Indian Association for the Cultivation of Science, Kolkata 700 032, India
| | - Nikhil R Jana
- School of Materials Science, Indian Association for the Cultivation of Science, Kolkata 700 032, India
| |
Collapse
|
10
|
Nonoyama S, Maeno S, Gotoh Y, Sugimoto R, Tanaka K, Hayashi T, Masuda S. Increased intracellular H 2S levels enhance iron uptake in Escherichia coli. mBio 2024; 15:e0199124. [PMID: 39324809 PMCID: PMC11481527 DOI: 10.1128/mbio.01991-24] [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: 07/01/2024] [Accepted: 09/02/2024] [Indexed: 09/27/2024] Open
Abstract
We investigated the impact of intracellular hydrogen sulfide (H2S) hyperaccumulation on the transcriptome of Escherichia coli. The wild-type (WT) strain overexpressing mstA, encoding 3-mercaptopyruvate sulfur transferase, produced significantly higher H2S levels than the control WT strain. The mstA-overexpressing strain exhibited increased resistance to antibiotics, supporting the prior hypothesis that intracellular H2S contributes to oxidative stress responses and antibiotic resistance. RNA-seq analysis revealed that over 1,000 genes were significantly upregulated or downregulated upon mstA overexpression. The upregulated genes encompassed those associated with iron uptake, including siderophore synthesis and iron import transporters. The mstA-overexpressing strain showed increased levels of intracellular iron content, indicating that H2S hyperaccumulation affects iron availability within cells. We found that the H2S-/supersulfide-responsive transcription factor YgaV is required for the upregulated expression of iron uptake genes in the mstA-overexpression conditions. These findings indicate that the expression of iron uptake genes is regulated by intracellular H2S, which is crucial for oxidative stress responses and antibiotic resistance in E. coli. IMPORTANCE H2S is recognized as a second messenger in bacteria, playing a vital role in diverse intracellular and extracellular activities, including oxidative stress responses and antibiotic resistance. Both H2S and iron serve as essential signaling molecules for gut bacteria. However, the intricate intracellular coordination between them, governing bacterial physiology, remains poorly understood. This study unveils a close relationship between intracellular H2S accumulation and iron uptake activity, a relationship critical for antibiotic resistance. We present additional evidence expanding the role of intracellular H2S synthesis in bacterial physiology.
Collapse
Affiliation(s)
- Shouta Nonoyama
- Department of Life Science and Technology, Tokyo Institute of Technology, Yokohama, Japan
| | - Shintaro Maeno
- Department of Biological Chemistry, College of Agriculture, Yamaguchi University, Yamaguchi, Japan
| | - Yasuhiro Gotoh
- Department of Bacteriology, Faculty of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Ryota Sugimoto
- Department of Life Science and Technology, Tokyo Institute of Technology, Yokohama, Japan
- Laboratory for Chemistry and Life Science, Institute of Innovative Research, Tokyo Institute of Technology, Yokohama, Japan
| | - Kan Tanaka
- Laboratory for Chemistry and Life Science, Institute of Innovative Research, Tokyo Institute of Technology, Yokohama, Japan
| | - Tetsuya Hayashi
- Department of Bacteriology, Faculty of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Shinji Masuda
- Department of Life Science and Technology, Tokyo Institute of Technology, Yokohama, Japan
| |
Collapse
|
11
|
Wedler V, Stiegler LMS, Gandziarowski T, Walter J, Peukert W, Distel LVR, Hirsch A, Klein S. Shell-by-Shell functionalized nanoparticles as radiosensitizers and radioprotectors in radiation therapy of cancer cells and tumor spheroids. Colloids Surf B Biointerfaces 2024; 245:114276. [PMID: 39353348 DOI: 10.1016/j.colsurfb.2024.114276] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Revised: 09/24/2024] [Accepted: 09/26/2024] [Indexed: 10/04/2024]
Abstract
Shell-by-Shell (SbS)-functionalized NPs can be tailor-made by combining a metal oxide NP core of choice with any desired phosphonic acids and amphiphiles as 1st or 2nd ligand shell building blocks. The complementary composition of such highly hierarchical structures makes them interesting candidates for various biomedical applications, as certain active ingredients can be incorporated into the structure. Here, we used TiO2 and CoFe2O4 NPs as drug delivery tools and coated them with a hexadecylphosphonic acid and with hexadecyl ammonium phenolates (caffeate, p-coumarate, ferulate), that possess anticancer as well as antioxidant properties. These architectures were then incubated in 2D and 3D cell cultures of non-tumorigenic and tumorigenic breast cells and irradiated to study their anticancer effect. It was found that both, the functionalized TiO2 and CoFe2O4 NPs acted as strong protective agents in non-tumorigenic spheroids. In contrast, the functionalized CoFe2O4 NPs induce a higher damage in irradiated tumor spheroids compared to the functionalized TiO2 NPs. CoFe3O4 NPs act additionally as radiosensitizing agents to the tumor spheroids. The radio-enhancement of the CoFe2O4 NPs is due to the generation of highly toxic hydroxyl radicals during X-ray irradiation. The irradiation exposed the CoFe2O4 surface, releasing the anticancer drugs into the cytoplasm and making the surface Co2+ ions accessible. These surface ions catalyze the Fenton reaction. This combination of radiosensitizer and anticancer drug delivery proved to be a very effective nanotherapeutic in 2D and 3D cell cultures of breast cancer cells.
Collapse
Affiliation(s)
- Vincent Wedler
- Department of Chemistry and Pharmacy, Chair of Organic Chemistry II, Friedrich-Alexander-Universität Erlangen-Nürnberg, Nikolaus-Fiebiger-Straße 10, Erlangen D-91058, Germany.
| | - Lisa M S Stiegler
- Institute of Particle Technology (LFG), Friedrich-Alexander-Universität Erlangen-Nürnberg, Cauerstr. 4, Erlangen 91058, Germany; Interdisciplinary Center for Functional Particle Systems (FPS), Friedrich-Alexander-Universität Erlangen-Nürnberg, Haberstrasse 9a, Erlangen 91058, Germany.
| | - Teresa Gandziarowski
- Department of Chemistry and Pharmacy, Physical Chemistry I, Friedrich-Alexander, Universität Erlangen-Nürnberg, Egerlandstr.3, Erlangen D-91058, Germany.
| | - Johannes Walter
- Institute of Particle Technology (LFG), Friedrich-Alexander-Universität Erlangen-Nürnberg, Cauerstr. 4, Erlangen 91058, Germany; Interdisciplinary Center for Functional Particle Systems (FPS), Friedrich-Alexander-Universität Erlangen-Nürnberg, Haberstrasse 9a, Erlangen 91058, Germany.
| | - Wolfgang Peukert
- Institute of Particle Technology (LFG), Friedrich-Alexander-Universität Erlangen-Nürnberg, Cauerstr. 4, Erlangen 91058, Germany; Interdisciplinary Center for Functional Particle Systems (FPS), Friedrich-Alexander-Universität Erlangen-Nürnberg, Haberstrasse 9a, Erlangen 91058, Germany.
| | - Luitpold V R Distel
- Department of Radiation Oncology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Universitätsstr. 27, Erlangen D-91054, Germany.
| | - Andreas Hirsch
- Department of Chemistry and Pharmacy, Chair of Organic Chemistry II, Friedrich-Alexander-Universität Erlangen-Nürnberg, Nikolaus-Fiebiger-Straße 10, Erlangen D-91058, Germany.
| | - Stefanie Klein
- Department of Chemistry and Pharmacy, Physical Chemistry I, Friedrich-Alexander, Universität Erlangen-Nürnberg, Egerlandstr.3, Erlangen D-91058, Germany.
| |
Collapse
|
12
|
García-Soriano D, Milán-Rois P, Lafuente-Gómez N, Rodríguez-Díaz C, Navío C, Somoza Á, Salas G. Multicore iron oxide nanoparticles for magnetic hyperthermia and combination therapy against cancer cells. J Colloid Interface Sci 2024; 670:73-85. [PMID: 38759270 DOI: 10.1016/j.jcis.2024.05.046] [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: 01/10/2024] [Revised: 04/24/2024] [Accepted: 05/07/2024] [Indexed: 05/19/2024]
Abstract
HYPOTHESIS Multicore flower-like iron oxide nanoparticles (IONPs) are among the best candidates for magnetic hyperthermia applications against cancers. However, they are rarely investigated in physiological environments and their efficacy against cancer cells has been even less studied. The combination of magnetic hyperthermia, using multicore IONPs, with selected bioactive molecules should lead to an enhanced activity against cancer cells. EXPERIMENTS Multicore IONPs were synthesized by a seeded-growth thermal decomposition approach. Then, the cytotoxicity, cell uptake, and efficacy of the magnetic hyperthermia approach were studied with six cancer cell lines: PANC1 (pancreatic carcinoma), Mel202 (uveal melanoma), MCF7 (breast adenocarcinoma), MB231 (triple-negative breast cancer line), A549 (lung cancer), and HCT116 (colon cancer). Finally, IONPs were modified with a chemotherapeutic drug (SN38) and tumor suppressor microRNAs (miR-34a, miR-182, let-7b, and miR-137), to study their activity against cancer cells with and without combination with magnetic hyperthermia. FINDINGS Two types of multicore IONPs with very good heating abilities under magnetic stimulation have been prepared. Their concentration-dependent cytotoxicity and internalization have been established, showing a strong dependence on the cell line and the nanoparticle type. Magnetic hyperthermia causes significant cell death that is dramatically enhanced in combination with the bioactive molecules.
Collapse
Affiliation(s)
- David García-Soriano
- Instituto Madrileño de Estudios Avanzados en Nanociencia, Campus Universitario de Cantoblanco, 28049 Madrid, Spain
| | - Paula Milán-Rois
- Instituto Madrileño de Estudios Avanzados en Nanociencia, Campus Universitario de Cantoblanco, 28049 Madrid, Spain
| | - Nuria Lafuente-Gómez
- Instituto Madrileño de Estudios Avanzados en Nanociencia, Campus Universitario de Cantoblanco, 28049 Madrid, Spain
| | - Ciro Rodríguez-Díaz
- Instituto Madrileño de Estudios Avanzados en Nanociencia, Campus Universitario de Cantoblanco, 28049 Madrid, Spain
| | - Cristina Navío
- Instituto Madrileño de Estudios Avanzados en Nanociencia, Campus Universitario de Cantoblanco, 28049 Madrid, Spain
| | - Álvaro Somoza
- Instituto Madrileño de Estudios Avanzados en Nanociencia, Campus Universitario de Cantoblanco, 28049 Madrid, Spain; Unidad Asociada de Nanobiotecnología (CNB-CSIC e IMDEA Nanociencia), 28049 Madrid, Spain
| | - Gorka Salas
- Instituto Madrileño de Estudios Avanzados en Nanociencia, Campus Universitario de Cantoblanco, 28049 Madrid, Spain; Unidad Asociada de Nanobiotecnología (CNB-CSIC e IMDEA Nanociencia), 28049 Madrid, Spain; Unidad de Nanomateriales Avanzados, IMDEA Nanociencia (Unidad de I+D+I Asociada al Instituto de Ciencia de Materiales de Madrid, CSIC), 28049 Madrid, Spain.
| |
Collapse
|
13
|
Fisher CE, Bak DW, Miller KE, Washington-Hughes CL, Dickfoss AM, Weerapana E, Py B, Outten FW. Escherichia coli monothiol glutaredoxin GrxD replenishes Fe-S clusters to the essential ErpA A-type carrier under low iron stress. J Biol Chem 2024; 300:107506. [PMID: 38944118 PMCID: PMC11327457 DOI: 10.1016/j.jbc.2024.107506] [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: 05/03/2024] [Revised: 06/01/2024] [Accepted: 06/18/2024] [Indexed: 07/01/2024] Open
Abstract
Iron-sulfur (Fe-S) clusters are required for essential biological pathways, including respiration and isoprenoid biosynthesis. Complex Fe-S cluster biogenesis systems have evolved to maintain an adequate supply of this critical protein cofactor. In Escherichia coli, two Fe-S biosynthetic systems, the "housekeeping" Isc and "stress responsive" Suf pathways, interface with a network of cluster trafficking proteins, such as ErpA, IscA, SufA, and NfuA. GrxD, a Fe-S cluster-binding monothiol glutaredoxin, also participates in Fe-S protein biogenesis in both prokaryotes and eukaryotes. Previous studies in E. coli showed that the ΔgrxD mutation causes sensitivity to iron depletion, spotlighting a critical role for GrxD under conditions that disrupt Fe-S homeostasis. Here, we utilized a global chemoproteomic mass spectrometry approach to analyze the contribution of GrxD to the Fe-S proteome. Our results demonstrate that (1) GrxD is required for biogenesis of a specific subset of Fe-S proteins under iron-depleted conditions, (2) GrxD is required for cluster delivery to ErpA under iron limitation, (3) GrxD is functionally distinct from other Fe-S trafficking proteins, and (4) GrxD Fe-S cluster binding is responsive to iron limitation. All these results lead to the proposal that GrxD is required to maintain Fe-S cluster delivery to the essential trafficking protein ErpA during iron limitation conditions.
Collapse
Affiliation(s)
- Claire E Fisher
- Department of Chemistry and Biochemistry, University of South Carolina, Columbia, South Carolina, USA
| | - Daniel W Bak
- Department of Chemistry, Boston College, Massachusetts, USA
| | - Kennedy E Miller
- Department of Chemistry and Biochemistry, University of South Carolina, Columbia, South Carolina, USA
| | | | - Anna M Dickfoss
- Department of Chemistry and Biochemistry, University of South Carolina, Columbia, South Carolina, USA
| | | | - Béatrice Py
- Aix-Marseille Université-Centre National de la Recherche Scientifique (UMR7283), Laboratoire de Chimie Bactérienne, Institut de Microbiologie de la Méditerranée, Institut Microbiologie Bioénergies et Biotechnologie, Marseille, France.
| | - F Wayne Outten
- Department of Chemistry and Biochemistry, University of South Carolina, Columbia, South Carolina, USA.
| |
Collapse
|
14
|
Wei P, Sun W, Hao S, Deng L, Zou W, Wu H, Lu W, He Y. Dietary Supplementation of Crossbred Pigs with Glycerol, Vitamin C, and Niacinamide Alters the Composition of Gut Flora and Gut Flora-Derived Metabolites. Animals (Basel) 2024; 14:2198. [PMID: 39123724 PMCID: PMC11311027 DOI: 10.3390/ani14152198] [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: 06/28/2024] [Revised: 07/24/2024] [Accepted: 07/26/2024] [Indexed: 08/12/2024] Open
Abstract
The addition of glycerin, vitamin C, and niacinamide to pig diets increased the redness of longissimus dorsi; however, it remains unclear how these supplements affect gut microbiota and metabolites. A total of 84 piglets (20.35 ± 2.14 kg) were randomly allotted to groups A (control), B (glycerin-supplemented), C (vitamin C and niacinamide-supplemented), and D (glycerin, vitamin C and niacinamide-supplemented) during a feeding experiment. Metagenomic and metabolomic technologies were used to analyze the fecal compositions of bile acids, metabolites, and microbiota. The results showed that compared to pigs in group A, pigs in group D had lower virulence factor expressions of lipopolysaccharide (p < 0.05), fatty acid resistance system (p < 0.05), and capsule (p < 0.01); higher fecal levels of ferric ion (p < 0.05), allolithocholic acid (p < 0.01), deoxycholic acid (p < 0.05), tauroursodeoxycholic acid dihydrate (p < 0.01), glycodeoxycholic acid (p < 0.05), L-proline (p < 0.01) and calcitriol (p < 0.01); and higher (p < 0.05) abundances of iron-acquiring microbiota (Methanobrevibacter, Clostridium, Clostridiaceae, Clostridium_sp_CAG_1000, Faecalibacterium_sp_CAG_74_58_120, Eubacteriales_Family_XIII_Incertae_Sedis, Alistipes_sp_CAG_435, Alistipes_sp_CAG_514 and Methanobrevibacter_sp_YE315). Supplementation with glycerin, vitamin C, and niacinamide to pigs significantly promoted the growth of iron-acquiring microbiota in feces, reduced the expression of some virulence factor genes of fecal pathogens, and increased the fecal levels of ferric ion, L-proline, and some secondary bile acids. The administration of glycerol, vitamin C, and niacinamide to pigs may serve as an effective measure for muscle redness improvement by altering the compositions of fecal microbiota and metabolites.
Collapse
Affiliation(s)
- Panting Wei
- Jiangxi Province Key Laboratory of Animal Nutrition and Feed, Engineering Research Center of Feed Development, Jiangxi Agricultural University, Nanchang 330045, China; (P.W.); (W.S.); (S.H.); (L.D.); (W.Z.); (W.L.)
- College of Animal Science and Technology, Jiangxi Agricultural University, Nanchang 330045, China;
| | - Wenchen Sun
- Jiangxi Province Key Laboratory of Animal Nutrition and Feed, Engineering Research Center of Feed Development, Jiangxi Agricultural University, Nanchang 330045, China; (P.W.); (W.S.); (S.H.); (L.D.); (W.Z.); (W.L.)
- College of Animal Science and Technology, Jiangxi Agricultural University, Nanchang 330045, China;
| | - Shaobin Hao
- Jiangxi Province Key Laboratory of Animal Nutrition and Feed, Engineering Research Center of Feed Development, Jiangxi Agricultural University, Nanchang 330045, China; (P.W.); (W.S.); (S.H.); (L.D.); (W.Z.); (W.L.)
- College of Animal Science and Technology, Jiangxi Agricultural University, Nanchang 330045, China;
| | - Linglan Deng
- Jiangxi Province Key Laboratory of Animal Nutrition and Feed, Engineering Research Center of Feed Development, Jiangxi Agricultural University, Nanchang 330045, China; (P.W.); (W.S.); (S.H.); (L.D.); (W.Z.); (W.L.)
- College of Animal Science and Technology, Jiangxi Agricultural University, Nanchang 330045, China;
| | - Wanjie Zou
- Jiangxi Province Key Laboratory of Animal Nutrition and Feed, Engineering Research Center of Feed Development, Jiangxi Agricultural University, Nanchang 330045, China; (P.W.); (W.S.); (S.H.); (L.D.); (W.Z.); (W.L.)
- College of Animal Science and Technology, Jiangxi Agricultural University, Nanchang 330045, China;
| | - Huadong Wu
- College of Animal Science and Technology, Jiangxi Agricultural University, Nanchang 330045, China;
| | - Wei Lu
- Jiangxi Province Key Laboratory of Animal Nutrition and Feed, Engineering Research Center of Feed Development, Jiangxi Agricultural University, Nanchang 330045, China; (P.W.); (W.S.); (S.H.); (L.D.); (W.Z.); (W.L.)
- College of Animal Science and Technology, Jiangxi Agricultural University, Nanchang 330045, China;
| | - Yuyong He
- Jiangxi Province Key Laboratory of Animal Nutrition and Feed, Engineering Research Center of Feed Development, Jiangxi Agricultural University, Nanchang 330045, China; (P.W.); (W.S.); (S.H.); (L.D.); (W.Z.); (W.L.)
- College of Animal Science and Technology, Jiangxi Agricultural University, Nanchang 330045, China;
| |
Collapse
|
15
|
Stavilă C, Minuti AE, Herea DD, Lăbuşcă L, Gherca D, Lupu N, Chiriac H. Synergistic Effect of Chemotherapy and Magnetomechanical Actuation of Fe-Cr-Nb-B Magnetic Particles on Cancer Cells. ACS OMEGA 2024; 9:30518-30533. [PMID: 39035922 PMCID: PMC11256100 DOI: 10.1021/acsomega.4c02189] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Revised: 06/12/2024] [Accepted: 06/14/2024] [Indexed: 07/23/2024]
Abstract
The present study is aimed at developing an innovative method for efficient cancer cell destruction by exploiting the magnetomechanical actuation (MMA) of Fe-Cr-Nb-B magnetic particles (MPs), which are loaded with clinically approved chemotherapeutic drugs. To achieve this objective, Fe68.2Cr11.5Nb0.3B20 magnetic nanoparticles were produced by mechanically grinding amorphous ribbon precursors with the same composition. These nanoparticles display high anisotropy, a parallelepipedic shape with an amorphous structure, and a ferromagnetic behavior. MPs were loaded with the antitumoral drugs mitoxantrone (MTX) or doxorubicin (DOX). In our study, we used adipose-derived mesenchymal stem cells and human osteosarcoma cells to test drug-loaded MPs for their biocompatibility, cytotoxicity, and cellular internalization. Further tests involved exposing cells to magnetomechanical actuation and simultaneous MPs-targeted chemotherapy followed by cell viability/death assays, such as MTT and LDH, and live/dead cell staining. Results demonstrate that cancer cell death was induced by the synergistic action of chemotherapeutic drugs and magnetomechanical actuation. The nanoparticle vehicles helped overcome drug resistance, decreasing the high dose of drugs used in conventional therapies as well as the time intervals needed for MMA to affect cancer cell viability. The proposed approach highlights the possibility of using a new, targeted, and effective cancer treatment with very few side effects.
Collapse
Affiliation(s)
- Cristina Stavilă
- National
Institute of Research and Development for Technical Physics, Iasi 700050, Romania
- Faculty
of Physics, “Alexandru Ioan Cuza”
University, Iasi 700506, Romania
| | - Anca Emanuela Minuti
- National
Institute of Research and Development for Technical Physics, Iasi 700050, Romania
| | - Dumitru Daniel Herea
- National
Institute of Research and Development for Technical Physics, Iasi 700050, Romania
| | - Luminiţa Lăbuşcă
- National
Institute of Research and Development for Technical Physics, Iasi 700050, Romania
| | - Daniel Gherca
- National
Institute of Research and Development for Technical Physics, Iasi 700050, Romania
| | - Nicoleta Lupu
- National
Institute of Research and Development for Technical Physics, Iasi 700050, Romania
| | - Horia Chiriac
- National
Institute of Research and Development for Technical Physics, Iasi 700050, Romania
| |
Collapse
|
16
|
Balsam SS, Zhong F, Pence N, Levintov L, Andhare D, Hammond JH, Ragusa MJ, Vashisth H, Hogan DA, Pletneva EV. Conserved C-Terminal Tail Is Responsible for Membrane Localization and Function of Pseudomonas aeruginosa Hemerythrin. Biochemistry 2024; 63:1795-1807. [PMID: 38951132 PMCID: PMC11481101 DOI: 10.1021/acs.biochem.4c00174] [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] [Indexed: 07/03/2024]
Abstract
Many bacteria have hemerythrin (Hr) proteins that bind O2, including Pseudomonas aeruginosa, in which microoxia-induced Hr (Mhr) provide fitness advantages under microoxic conditions. Mhr has a 23 amino-acid extension at its C-terminus relative to a well-characterized Hr from Methylococcus capsulatus, and similar extensions are also found in Hrs from other bacteria. The last 11 amino acids of this extended, C-terminal tail are highly conserved in gammaproteobacteria and predicted to form a helix with positively charged and hydrophobic faces. In cellular fractionation assays, wild-type (WT) Mhr was found in both membrane and cytosolic fractions, while a MhrW143* variant lacking the last 11 residues was largely in the cytosol and did not complement Mhr function in competition assays. MhrL112Y, a variant that has a much longer-lived O2-bound form, was fully functional and had a similar localization pattern to that of WT Mhr. Both MhrW143* and MhrL112Y had secondary structures, stabilities, and O2-binding kinetics similar to those of WT Mhr. Fluorescence studies revealed that the C-terminal tail, and particularly the fragment corresponding to its last 11 residues, was sufficient and necessary for association with lipid vesicles. Molecular dynamics simulations and subsequent cellular analysis of Mhr variants have demonstrated that conserved, positively charged residues in the tail are important for Mhr interactions with negatively charged membranes and the contribution of this protein to competitive fitness. Together, these data suggest that peripheral interactions of Mhr with membranes are guided by the C-terminal tail and are independent of O2-binding.
Collapse
Affiliation(s)
- Stacie Stuut Balsam
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Hanover, NH, 03755, USA
| | - Fangfang Zhong
- Department of Chemistry, Dartmouth College, Hanover, NH, 03755, USA
| | - Natasha Pence
- Department of Chemistry, Dartmouth College, Hanover, NH, 03755, USA
| | - Lev Levintov
- Department of Chemical Engineering and Bioengineering, University of New Hampshire, Durham, NH, 03824, USA
| | - Devika Andhare
- Department of Chemistry, Dartmouth College, Hanover, NH, 03755, USA
| | - John H. Hammond
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Hanover, NH, 03755, USA
| | | | - Harish Vashisth
- Department of Chemical Engineering and Bioengineering, University of New Hampshire, Durham, NH, 03824, USA
- Department of Chemistry, University of New Hampshire, Durham, NH, 03824, USA
- Integrated Applied Mathematics Program, University of New Hampshire, Durham, NH, 03824, USA
- Molecular and Cellular Biotechnology Program, University of New Hampshire, Durham, NH, 03824, USA
| | - Deborah A. Hogan
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Hanover, NH, 03755, USA
| | | |
Collapse
|
17
|
Chalivendra S, Shi S, Li X, Kuang Z, Giovinazzo J, Zhang L, Rossi J, Wang J, Saviola AJ, Welty R, Liu S, Vaeth KF, Zhou ZH, Hansen KC, Taliaferro JM, Zhao R. Selected humanization of yeast U1 snRNP leads to global suppression of pre-mRNA splicing and mitochondrial dysfunction in the budding yeast. RNA (NEW YORK, N.Y.) 2024; 30:1070-1088. [PMID: 38688558 PMCID: PMC11251525 DOI: 10.1261/rna.079917.123] [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: 12/08/2023] [Accepted: 04/16/2024] [Indexed: 05/02/2024]
Abstract
The recognition of the 5' splice site (5' ss) is one of the earliest steps of pre-mRNA splicing. To better understand, the mechanism and regulation of 5' ss recognition, we selectively humanized components of the yeast U1 (yU1) snRNP to reveal the function of these components in 5' ss recognition and splicing. We targeted U1C and Luc7, two proteins that interact with and stabilize the yU1 snRNA and the 5' ss RNA duplex. We replaced the zinc-finger (ZnF) domain of yeast U1C (yU1C) with its human counterpart, which resulted in a cold-sensitive growth phenotype and moderate splicing defects. We next added an auxin-inducible degron to yeast Luc7 (yLuc7) protein (to mimic the lack of Luc7Ls in human U1 snRNP). We found that Luc7-depleted yU1 snRNP resulted in the concomitant loss of Prp40 and Snu71 (two other essential yU1 snRNP proteins), and further biochemical analyses suggest a model of how these three proteins interact with each other in the U1 snRNP. The loss of these proteins resulted in a significant growth retardation accompanied by a global suppression of pre-mRNA splicing. The splicing suppression led to mitochondrial dysfunction as revealed by a release of Fe2+ into the growth medium and an induction of mitochondrial reactive oxygen species. Together, these observations indicate that the human U1C ZnF can substitute that of yeast, Luc7 is essential for the incorporation of the Luc7-Prp40-Snu71 trimer into yU1 snRNP, and splicing plays a major role in the regulation of mitochondrial function in yeast.
Collapse
Affiliation(s)
- Subbaiah Chalivendra
- Department of Biochemistry and Molecular Genetics, University of Colorado Anschutz Medical Campus, Aurora, Colorado 80045, USA
| | - Shasha Shi
- Department of Biochemistry and Molecular Genetics, University of Colorado Anschutz Medical Campus, Aurora, Colorado 80045, USA
| | - Xueni Li
- Department of Biochemistry and Molecular Genetics, University of Colorado Anschutz Medical Campus, Aurora, Colorado 80045, USA
| | - Zhiling Kuang
- Department of Biochemistry and Molecular Genetics, University of Colorado Anschutz Medical Campus, Aurora, Colorado 80045, USA
| | - Joseph Giovinazzo
- Department of Biochemistry and Molecular Genetics, University of Colorado Anschutz Medical Campus, Aurora, Colorado 80045, USA
| | - Lingdi Zhang
- Department of Biochemistry and Molecular Genetics, University of Colorado Anschutz Medical Campus, Aurora, Colorado 80045, USA
| | - John Rossi
- Department of Biochemistry and Molecular Genetics, University of Colorado Anschutz Medical Campus, Aurora, Colorado 80045, USA
| | - Jingxin Wang
- Department of Medicinal Chemistry, University of Kansas, Lawrence, Kansas 66047, USA
| | - Anthony J Saviola
- Department of Biochemistry and Molecular Genetics, University of Colorado Anschutz Medical Campus, Aurora, Colorado 80045, USA
| | - Robb Welty
- Department of Biochemistry and Molecular Genetics, University of Colorado Anschutz Medical Campus, Aurora, Colorado 80045, USA
| | - Shiheng Liu
- Department of Microbiology, Immunology, and Molecular Genetics, University of California, Los Angeles, California 90095, USA
| | - Katherine F Vaeth
- Department of Biochemistry and Molecular Genetics, University of Colorado Anschutz Medical Campus, Aurora, Colorado 80045, USA
| | - Z Hong Zhou
- Department of Microbiology, Immunology, and Molecular Genetics, University of California, Los Angeles, California 90095, USA
| | - Kirk C Hansen
- Department of Biochemistry and Molecular Genetics, University of Colorado Anschutz Medical Campus, Aurora, Colorado 80045, USA
| | - J Matthew Taliaferro
- Department of Biochemistry and Molecular Genetics, University of Colorado Anschutz Medical Campus, Aurora, Colorado 80045, USA
| | - Rui Zhao
- Department of Biochemistry and Molecular Genetics, University of Colorado Anschutz Medical Campus, Aurora, Colorado 80045, USA
| |
Collapse
|
18
|
Gromadzka G, Wilkaniec A, Tarnacka B, Hadrian K, Bendykowska M, Przybyłkowski A, Litwin T. The Role of Glia in Wilson's Disease: Clinical, Neuroimaging, Neuropathological and Molecular Perspectives. Int J Mol Sci 2024; 25:7545. [PMID: 39062788 PMCID: PMC11276698 DOI: 10.3390/ijms25147545] [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: 05/30/2024] [Revised: 07/07/2024] [Accepted: 07/08/2024] [Indexed: 07/28/2024] Open
Abstract
Wilson's disease (WD) is inherited in an autosomal recessive manner and is caused by pathogenic variants of the ATP7B gene, which are responsible for impaired copper transport in the cell, inhibition of copper binding to apoceruloplasmin, and biliary excretion. This leads to the accumulation of copper in the tissues. Copper accumulation in the CNS leads to the neurological and psychiatric symptoms of WD. Abnormalities of copper metabolism in WD are associated with impaired iron metabolism. Both of these elements are redox active and may contribute to neuropathology. It has long been assumed that among parenchymal cells, astrocytes have the greatest impact on copper and iron homeostasis in the brain. Capillary endothelial cells are separated from the neuropil by astrocyte terminal legs, putting astrocytes in an ideal position to regulate the transport of iron and copper to other brain cells and protect them if metals breach the blood-brain barrier. Astrocytes are responsible for, among other things, maintaining extracellular ion homeostasis, modulating synaptic transmission and plasticity, obtaining metabolites, and protecting the brain against oxidative stress and toxins. However, excess copper and/or iron causes an increase in the number of astrocytes and their morphological changes observed in neuropathological studies, as well as a loss of the copper/iron storage function leading to macromolecule peroxidation and neuronal loss through apoptosis, autophagy, or cuproptosis/ferroptosis. The molecular mechanisms explaining the possible role of glia in copper- and iron-induced neurodegeneration in WD are largely understood from studies of neuropathology in Parkinson's disease and Alzheimer's disease. Understanding the mechanisms of glial involvement in neuroprotection/neurotoxicity is important for explaining the pathomechanisms of neuronal death in WD and, in the future, perhaps for developing more effective diagnostic/treatment methods.
Collapse
Affiliation(s)
- Grażyna Gromadzka
- Department of Biomedical Sciences, Faculty of Medicine, Collegium Medicum, Cardinal Stefan Wyszynski University, Wóycickiego 1/3, 01-938 Warsaw, Poland
| | - Anna Wilkaniec
- Department of Cellular Signalling, Mossakowski Medical Research Centre, Polish Academy of Sciences, 5 Pawińskiego St., 02-106 Warsaw, Poland
| | - Beata Tarnacka
- Department of Rehabilitation, Medical University of Warsaw, Spartańska 1, 02-637 Warsaw, Poland
| | - Krzysztof Hadrian
- Department of Gastroenterology and Internal Medicine, Medical University of Warsaw, Banacha 1a, 02-097 Warsaw, Poland (A.P.)
| | - Maria Bendykowska
- Students Scientific Association “Immunis”, Cardinal Stefan Wyszynski University, Dewajtis 5, 01-815 Warsaw, Poland
| | - Adam Przybyłkowski
- Department of Gastroenterology and Internal Medicine, Medical University of Warsaw, Banacha 1a, 02-097 Warsaw, Poland (A.P.)
| | - Tomasz Litwin
- Second Department of Neurology, Institute of Psychiatry and Neurology, Sobieskiego 9, 02-957 Warsaw, Poland
| |
Collapse
|
19
|
Dash S, Majood M, Meena R, Mukherjee M, Dinda AK, Kuanr BK, Mohanty S. Biocompatible polymer-coated magneto-fluorescent super nanoparticles for the homing of mesenchymal stem cells. Int J Biol Macromol 2024; 273:132794. [PMID: 38834114 DOI: 10.1016/j.ijbiomac.2024.132794] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2023] [Revised: 04/04/2024] [Accepted: 05/29/2024] [Indexed: 06/06/2024]
Abstract
Stem cell plays an important role in the clinical field. However, the effective delivery of stem cells to the targeted site relies on the efficient homing of the cells to the site of injury. In view of that, fluorescent magnetic nanoparticles stick out due to their wide range of enabling functions including cellular homing and tracking. The present study unravels the synthesis of polymer-coated biocompatible and fluorescent magnetic nanoparticles (FMNPs) by a single-step hydrothermal synthesis method. Importantly, the facile method developed the biological super nanoparticles consisting of the magnetic core, which is surrounded by the fluorescent nanodot-decorated polymeric shell. The synthesized particles showed an amorphous nature, and superparamagnetic properties, with efficient fluorescence properties of emission at the blue range (̴ 410 nm). The FMNP labeling showed the mesenchymal stem cell (MSC) homing to the desired site in the presence of an external magnetic field. The in-house synthesized nanoparticles showed significant cytocompatibility and hemocompatibility in vitro as well as in vivo conditions owing to their surface coating. This unprecedented work advances the efficient internalization of FMNPs in MSCs and their enhanced migration potential provides a breakthrough in stem cell delivery for therapeutic applications. STATEMENT OF SIGNIFICANCE: The bi-modal fluorescent magnetic nanoparticles hold a promising role in the biomedical field for mesenchymal stem cell homing and tracking. Hence, in this study, for the first time, we have synthesized the fluorescent magnetic nanoparticle with polymer coating via an easy single-step method. The nanoparticle with a polymer coat enhanced the biocompatibility and effortless internalization of the nanoparticle into mesenchymal stem cells without hampering the native stem cell properties. Furthermore, the enhanced migration potential of such magnetized stem cells and their homing at the target site by applying an external magnetic field opened up avenues for the smart delivery of mesenchymal stem cells at complex sites such as retina for the tissue regeneration.
Collapse
Affiliation(s)
- Saumya Dash
- Stem Cell Facility, DBT-Centre of Excellence for Stem Cell Research, All India Institute of Medical Sciences, New Delhi 110029, India
| | - Misba Majood
- Stem Cell Facility, DBT-Centre of Excellence for Stem Cell Research, All India Institute of Medical Sciences, New Delhi 110029, India; Amity Institute of Click Chemistry Research and Studies, Amity University, Uttar Pradesh, 201303 Noida, India
| | - Ravindra Meena
- Special Centre for Nano Science, Jawaharlal Nehru University, New Delhi 110067, India
| | - Monalisa Mukherjee
- Amity Institute of Click Chemistry Research and Studies, Amity University, Uttar Pradesh, 201303 Noida, India
| | - Amit K Dinda
- Department of Pathology, All India Institute of Medical Sciences, New Delhi 110029, India
| | - Bijoy K Kuanr
- Special Centre for Nano Science, Jawaharlal Nehru University, New Delhi 110067, India
| | - Sujata Mohanty
- Stem Cell Facility, DBT-Centre of Excellence for Stem Cell Research, All India Institute of Medical Sciences, New Delhi 110029, India.
| |
Collapse
|
20
|
Sen Gupta P, Karmakar S, Biswas I, Ghosal J, Banerjee A, Roy S, Mandal DP, Bhattacharjee S. Vitamin E alleviates chlorpyrifos induced glutathione depletion, lipid peroxidation and iron accumulation to inhibit ferroptosis in hepatocytes and mitigate toxicity in zebrafish. CHEMOSPHERE 2024; 359:142252. [PMID: 38735493 DOI: 10.1016/j.chemosphere.2024.142252] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/09/2024] [Revised: 04/25/2024] [Accepted: 05/03/2024] [Indexed: 05/14/2024]
Abstract
Organophosphates, a widely used group of pesticides, can cause severe toxicity in human beings and other non-target organisms. Liver, being the primary site for xenobiotic metabolism, is extremely vulnerable to xenobiotic-induced toxicity. Considering the numerous vital functions performed by the liver, including xenobiotic detoxification, protecting this organ from the ubiquitous pesticides in our food and environment is essential for maintaining homeostasis. In this study, we have investigated the impact of the organophosphate pesticide, Chlorpyrifos (CPF), on zebrafish liver at a concentration (300 μg/L) which is environmentally realistic. We have also demonstrated the role of dietary supplementation of α-tocopherol or Vitamin E (Vit E) (500 mg/kg feed) in mitigating pesticide-induced liver toxicity. Mechanistically, we showed that Vit E resulted in significant elevation of the Nrf2 and its downstream antioxidant enzyme activities and gene expressions, especially that of GST and GPx, resulting in reduction of CPF-induced intracellular lipid ROS and hepatic LPO. Further interrogation, such as analysis of GSH: GSSG ratio, intracellular iron concentration, iron metabolizing genes, mitochondrial dysfunction etc. revealed that CPF induces ferroptosis which can be reversed by Vit E supplementation. Ultimately, reduced concentration of CPF in zebrafish serum and flesh highlighted the role of Vit E in ameliorating CPF toxicity.
Collapse
Affiliation(s)
- Poulami Sen Gupta
- Department of Zoology, West Bengal State University, Kolkata-700126, West Bengal, India
| | - Subrata Karmakar
- Department of Zoology, West Bengal State University, Kolkata-700126, West Bengal, India
| | - Ipsita Biswas
- Department of Zoology, West Bengal State University, Kolkata-700126, West Bengal, India
| | - Jahnabi Ghosal
- Department of Zoology, West Bengal State University, Kolkata-700126, West Bengal, India
| | - Ankur Banerjee
- Department of Zoology, West Bengal State University, Kolkata-700126, West Bengal, India
| | - Soumen Roy
- Department of Zoology, City College, Calcutta University, Kolkata-700009, West Bengal, India
| | - Deba Prasad Mandal
- Department of Zoology, West Bengal State University, Kolkata-700126, West Bengal, India.
| | - Shamee Bhattacharjee
- Department of Zoology, West Bengal State University, Kolkata-700126, West Bengal, India.
| |
Collapse
|
21
|
Renner N, Schöb F, Pape R, Suciu I, Spreng AS, Ückert AK, Cöllen E, Bovio F, Chilian B, Bauer J, Röpcke S, Bergemann J, Leist M, Schildknecht S. Modeling ferroptosis in human dopaminergic neurons: Pitfalls and opportunities for neurodegeneration research. Redox Biol 2024; 73:103165. [PMID: 38688061 PMCID: PMC11070765 DOI: 10.1016/j.redox.2024.103165] [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: 12/10/2023] [Revised: 03/17/2024] [Accepted: 04/18/2024] [Indexed: 05/02/2024] Open
Abstract
The activation of ferroptosis is being pursued in cancer research as a strategy to target apoptosis-resistant cells. By contrast, in various diseases that affect the cardiovascular system, kidneys, liver, and central and peripheral nervous systems, attention is directed toward interventions that prevent ferroptotic cell death. Mechanistic insights into both research areas stem largely from studies using cellular in vitro models. However, intervention strategies that show promise in cellular test systems often fail in clinical trials, which raises concerns regarding the predictive validity of the utilized in vitro models. In this study, the human LUHMES cell line, which serves as a model for human dopaminergic neurons, was used to characterize factors influencing the activation of ferroptosis. Erastin and RSL-3 induced cell death that was distinct from apoptosis. Parameters such as the differentiation state of LUHMES cells, cell density, and the number and timing of medium changes were identified as determinants of sensitivity to ferroptosis activation. In differentiated LUHMES cells, interventions at mechanistically divergent sites (iron chelation, coenzyme Q10, peroxidase mimics, or inhibition of 12/15-lipoxygenase) provide almost complete protection from ferroptosis. LUHMES cells allowed the experimental modulation of intracellular iron concentrations and demonstrated a correlation between intracellular iron levels, the rate of lipid peroxidation, as well as the sensitivity of the cells to ferroptotic cell death. These findings underscore the importance of understanding the various factors that influence ferroptosis activation and highlight the need for well-characterized in vitro models to enhance the reliability and predictive value of observations in ferroptosis research, particularly when translating findings into in vivo contexts.
Collapse
Affiliation(s)
- Nadine Renner
- Albstadt-Sigmaringen University, Faculty of Life Sciences, 72488, Sigmaringen, Germany
| | - Franziska Schöb
- In Vitro Toxicology and Biomedicine, Department of Biology, University of Konstanz, 78457, Konstanz, Germany
| | - Regina Pape
- In Vitro Toxicology and Biomedicine, Department of Biology, University of Konstanz, 78457, Konstanz, Germany
| | - Ilinca Suciu
- In Vitro Toxicology and Biomedicine, Department of Biology, University of Konstanz, 78457, Konstanz, Germany
| | - Anna-Sophie Spreng
- In Vitro Toxicology and Biomedicine, Department of Biology, University of Konstanz, 78457, Konstanz, Germany
| | - Anna-Katharina Ückert
- In Vitro Toxicology and Biomedicine, Department of Biology, University of Konstanz, 78457, Konstanz, Germany
| | - Eike Cöllen
- In Vitro Toxicology and Biomedicine, Department of Biology, University of Konstanz, 78457, Konstanz, Germany
| | - Federica Bovio
- Department of Biotechnology and Biosciences, University of Milano-Bicocca, 20126, Milano, Italy
| | - Bruno Chilian
- TRI Thinking Research Instruments GmbH, Große Freiheit 77, 22767, Hamburg, Germany
| | - Johannes Bauer
- TRI Thinking Research Instruments GmbH, Große Freiheit 77, 22767, Hamburg, Germany
| | - Stefan Röpcke
- Stemick GmbH, Byk-Gulden Str. 2, 78467, Konstanz, Germany
| | - Jörg Bergemann
- Albstadt-Sigmaringen University, Faculty of Life Sciences, 72488, Sigmaringen, Germany
| | - Marcel Leist
- In Vitro Toxicology and Biomedicine, Department of Biology, University of Konstanz, 78457, Konstanz, Germany
| | - Stefan Schildknecht
- Albstadt-Sigmaringen University, Faculty of Life Sciences, 72488, Sigmaringen, Germany.
| |
Collapse
|
22
|
Chen X, Tsvetkov AS, Shen HM, Isidoro C, Ktistakis NT, Linkermann A, Koopman WJ, Simon HU, Galluzzi L, Luo S, Xu D, Gu W, Peulen O, Cai Q, Rubinsztein DC, Chi JT, Zhang DD, Li C, Toyokuni S, Liu J, Roh JL, Dai E, Juhasz G, Liu W, Zhang J, Yang M, Liu J, Zhu LQ, Zou W, Piacentini M, Ding WX, Yue Z, Xie Y, Petersen M, Gewirtz DA, Mandell MA, Chu CT, Sinha D, Eftekharpour E, Zhivotovsky B, Besteiro S, Gabrilovich DI, Kim DH, Kagan VE, Bayir H, Chen GC, Ayton S, Lünemann JD, Komatsu M, Krautwald S, Loos B, Baehrecke EH, Wang J, Lane JD, Sadoshima J, Yang WS, Gao M, Münz C, Thumm M, Kampmann M, Yu D, Lipinski MM, Jones JW, Jiang X, Zeh HJ, Kang R, Klionsky DJ, Kroemer G, Tang D. International consensus guidelines for the definition, detection, and interpretation of autophagy-dependent ferroptosis. Autophagy 2024; 20:1213-1246. [PMID: 38442890 PMCID: PMC11210914 DOI: 10.1080/15548627.2024.2319901] [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: 08/25/2023] [Revised: 09/29/2023] [Accepted: 10/19/2023] [Indexed: 03/07/2024] Open
Abstract
Macroautophagy/autophagy is a complex degradation process with a dual role in cell death that is influenced by the cell types that are involved and the stressors they are exposed to. Ferroptosis is an iron-dependent oxidative form of cell death characterized by unrestricted lipid peroxidation in the context of heterogeneous and plastic mechanisms. Recent studies have shed light on the involvement of specific types of autophagy (e.g. ferritinophagy, lipophagy, and clockophagy) in initiating or executing ferroptotic cell death through the selective degradation of anti-injury proteins or organelles. Conversely, other forms of selective autophagy (e.g. reticulophagy and lysophagy) enhance the cellular defense against ferroptotic damage. Dysregulated autophagy-dependent ferroptosis has implications for a diverse range of pathological conditions. This review aims to present an updated definition of autophagy-dependent ferroptosis, discuss influential substrates and receptors, outline experimental methods, and propose guidelines for interpreting the results.Abbreviation: 3-MA:3-methyladenine; 4HNE: 4-hydroxynonenal; ACD: accidentalcell death; ADF: autophagy-dependentferroptosis; ARE: antioxidant response element; BH2:dihydrobiopterin; BH4: tetrahydrobiopterin; BMDMs: bonemarrow-derived macrophages; CMA: chaperone-mediated autophagy; CQ:chloroquine; DAMPs: danger/damage-associated molecular patterns; EMT,epithelial-mesenchymal transition; EPR: electronparamagnetic resonance; ER, endoplasmic reticulum; FRET: Försterresonance energy transfer; GFP: green fluorescent protein;GSH: glutathione;IF: immunofluorescence; IHC: immunohistochemistry; IOP, intraocularpressure; IRI: ischemia-reperfusion injury; LAA: linoleamide alkyne;MDA: malondialdehyde; PGSK: Phen Green™ SK;RCD: regulatedcell death; PUFAs: polyunsaturated fatty acids; RFP: red fluorescentprotein;ROS: reactive oxygen species; TBA: thiobarbituricacid; TBARS: thiobarbituric acid reactive substances; TEM:transmission electron microscopy.
Collapse
Affiliation(s)
- Xin Chen
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Protein Modification and Degradation, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, China
| | - Andrey S. Tsvetkov
- Department of Neurology, The University of Texas McGovern Medical School at Houston, Houston, TX, USA
| | - Han-Ming Shen
- Department of Biomedical Sciences, Faculty of Health Sciences, University of Macau, Macau, China
| | - Ciro Isidoro
- Department of Health Sciences, University of Piemonte Orientale, Novara, Italy
| | | | - Andreas Linkermann
- Division of Nephrology, Department of Internal Medicine 3, University Hospital Carl Gustav Carus at the Technische Universität Dresden, Germany
- Division of Nephrology, Department of Medicine, Albert Einstein College of Medicine, Bronx, New York, USA
| | - Werner J.H. Koopman
- Department of Pediatrics, Radboud Center for Mitochondrial Medicine, Radboud University Medical Center, Nijmegen, The Netherlands
- Human and Animal Physiology, Wageningen University, Wageningen, The Netherlands
| | - Hans-Uwe Simon
- Institute of Pharmacology, University of Bern, Bern, Switzerland
- Institute of Biochemistry, Brandenburg Medical School, Neuruppin, Germany
| | - Lorenzo Galluzzi
- Department of Radiation Oncology, Weill Cornell Medical College, New York, NY, USA
- Sandra and Edward Meyer Cancer Center, New York, NY, USA
- Caryl and Israel Englander Institute for Precision Medicine, New York, NY, USA
| | - Shouqing Luo
- Peninsula Medical School, University of Plymouth, Plymouth, UK
| | - Daqian Xu
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Institute of Translational Medicine, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, China
| | - Wei Gu
- Institute for Cancer Genetics, and Department of Pathology and Cell Biology, Vagelos College of Physicians & Surgeons, Columbia University, New York, NY, USA
| | - Olivier Peulen
- Metastasis Research Laboratory, GIGA Cancer-University of Liège, Liège, Belgium
| | - Qian Cai
- Department of Cell Biology and Neuroscience, Rutgers, The State University of New Jersey, Piscataway, NJ, USA
| | - David C. Rubinsztein
- Department of Medical Genetics, Cambridge Institute for Medical Research, University of Cambridge, Cambridge, UK
- UK Dementia Research Institute, University of Cambridge, Cambridge, UK
| | - Jen-Tsan Chi
- Department of Molecular Genetics and Microbiology, Duke University, Durham, NC, USA
| | - Donna D. Zhang
- Pharmacology and Toxicology, R. Ken Coit College of Pharmacy, University of Arizona, Tucson, AZ, USA
| | - Changfeng Li
- Department of Endoscopy Center, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Shinya Toyokuni
- Department of Pathology and Biological Response, Nagoya University Graduate School of Medicine, Nagoya, Japan
- Center for Low-temperature Plasma Sciences, Nagoya University, Nagoya, Japan
| | - Jinbao Liu
- Affiliated Cancer Hospital & Institute of Guangzhou Medical University, Guangzhou Municipal and Guangdong Provincial Key Laboratory of Protein Modification and Degradation, State Key Laboratory of Respiratory Disease, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, China
| | - Jong-Lyel Roh
- Department of Otorhinolaryngology-Head and Neck Surgery, CHA Bundang Medical Center, CHA University, Seongnam, Republic of Korea
| | - Enyong Dai
- The Second Department of Hematology and Oncology, China-Japan Union Hospital of Jilin University, Changchun, Jilin, China
| | - Gabor Juhasz
- Biological Research Center, Institute of Genetics, Szeged, Hungary
- Department of Anatomy, Cell and Developmental Biology, Eotvos Lorand University, Budapest, Hungary
| | - Wei Liu
- Department of Orthopedics, Changzheng Hospital, Second Affiliated Hospital of Naval Medical University, Shanghai, China
| | - Jianhua Zhang
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Minghua Yang
- Department of Pediatrics, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China
- Hunan Clinical Research Center of Pediatric Cancer, Changsha, China
| | - Jiao Liu
- DAMP Laboratory, Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Ling-Qiang Zhu
- Department of Pathophysiology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Weiping Zou
- Departments of Surgery and Pathology, University of Michigan Medical School, Ann Arbor, USA
| | - Mauro Piacentini
- Department of Biology, University of Rome “Tor Vergata”, Rome, Italy
- National Institute for Infectious Diseases IRCCS “Lazzaro Spallanzani”, Rome, Italy
| | - Wen-Xing Ding
- Department of Pharmacology, Toxicology and Therapeutics, The University of Kansas Medical Center, Kansas City, KS, USA
| | - Zhenyu Yue
- Department of Neurology, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Yangchun Xie
- Department of Oncology, Central South University, Changsha, Hunan, China
| | - Morten Petersen
- Functional genomics, Department of Biology, Copenhagen University, Denmark
| | - David A. Gewirtz
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Massey Cancer Center, Richmond, VA, USA
| | - Michael A. Mandell
- Department of Molecular Genetics and Microbiology, University of New Mexico, Albuquerque, USA
| | - Charleen T. Chu
- Pathology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Debasish Sinha
- Department of Ophthalmology, University of Pittsburgh, Pittsburgh, PA, USA; Wilmer Eye lnstitute, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Eftekhar Eftekharpour
- Department of Physiology and Pathophysiology, University of Manitoba, Winnipeg, Canada
- Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer, Villejuif, France; Gustave Roussy Cancer, Villejuif, France
| | - Boris Zhivotovsky
- Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden, Europe
- Faculty of Medicine, Lomonosov Moscow State University, Moscow, Russia
- Engelhardt Institute of Molecular Biology, Moscow, Russia
| | - Sébastien Besteiro
- LPHI, University Montpellier, CNRS, Montpellier, France
- Institut du Cancer Paris CARPEM, Department of Biology, Hôpital Européen Georges Pompidou, AP-HP, Paris, France
| | | | - Do-Hyung Kim
- Department of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota, Minneapolis, MN, USA
| | - Valerian E. Kagan
- Department of Environmental Health, University of Pittsburgh, Pittsburgh, PA, USA
| | - Hülya Bayir
- Department of Pediatrics, Columbia University, New York, USA
| | - Guang-Chao Chen
- Institute of Biological Chemistry, Academia Sinica, Taipei, Taiwan
| | - Scott Ayton
- Florey Institute, University of Melbourne, Parkville, Australia
| | - Jan D. Lünemann
- Department of Neurology with Institute of Translational Neurology, University of Münster, Münster, Germany
| | - Masaaki Komatsu
- Department of Physiology, Juntendo University School of Medicine, Bunkyo-ku Tokyo, Japan
| | - Stefan Krautwald
- Department of Nephrology and Hypertension, University Hospital Schleswig-Holstein, Kiel, Germany
| | - Ben Loos
- Department of Physiological Sciences, Stellenbosch University, Stellenbosch, South Africa
| | - Eric H. Baehrecke
- Department of Molecular, Cell and Cancer Biology, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Jiayi Wang
- Department of Clinical Laboratory, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Institute of Thoracic Oncology Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- College of Medical Technology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jon D. Lane
- School of Biochemistry, University of Bristol, Bristol, UK
| | - Junichi Sadoshima
- Rutgers New Jersey Medical School, Department of Cell Biology and Molecular Medicine, Newark, USA
| | - Wan Seok Yang
- Department of Biological Sciences, St. John’s University, New York City, NY, USA
| | - Minghui Gao
- The HIT Center for Life Sciences, School of Life Science and Technology, Harbin Institute of Technology, Harbin, China
| | - Christian Münz
- Institute of Experimental Immunology, University of Zürich, Zürich, Switzerland
| | - Michael Thumm
- Department of Cellular Biochemistry, University Medical Center Goettingen, Goettingen, Germany
| | - Martin Kampmann
- Department of Biochemistry & Biophysics, University of California, San Francisco, USA
- Institute for Neurodegenerative Diseases, University of California, San Francisco, USA
| | - Di Yu
- Faculty of Medicine, Frazer Institute, University of Queensland, Brisbane, Australia
- Faculty of Medicine, Ian Frazer Centre for Children’s Immunotherapy Research, Child Health Research Centre, University of Queensland, Brisbane, Australia
| | - Marta M. Lipinski
- Department of Anesthesiology & Department of Neurobiology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Jace W. Jones
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Maryland, Baltimore, MD, USA
| | - Xuejun Jiang
- Cell Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Herbert J. Zeh
- Department of Surgery, UT Southwestern Medical Center, Dallas, TX, USA
| | - Rui Kang
- Department of Surgery, UT Southwestern Medical Center, Dallas, TX, USA
| | - Daniel J. Klionsky
- Life Sciences Institute and Department of Molecular, Cellular and Developmental Biology, University of Michigan, Ann Arbor, MI, USA
| | - Guido Kroemer
- Centre de Recherche des Cordeliers, Université de Paris, Sorbonne Université, INSERM U1138, Institut Universitaire de France, Paris, France
- Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer, Villejuif, France; Gustave Roussy Cancer, Villejuif, France
- Institut du Cancer Paris CARPEM, Department of Biology, Hôpital Européen Georges Pompidou, AP-HP, Paris, France
| | - Daolin Tang
- Department of Surgery, UT Southwestern Medical Center, Dallas, TX, USA
| |
Collapse
|
23
|
Dubey Y, Mansuri S, Kanvah S. Detecting labile heme and ferroptosis through 'turn-on' fluorescence and lipid droplet localization post Fe 2+ sensing. J Mater Chem B 2024; 12:4962-4974. [PMID: 38687117 DOI: 10.1039/d4tb00353e] [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: 05/02/2024]
Abstract
Iron, a crucial biologically active ion essential for metabolic processes in living organisms, plays a vital role in biological functions, and imbalances in iron levels can lead to various diseases. In this study, we have developed two simple "turn-on" fluorescent probes, NOPy and NOCN, for the quick and selective detection of Fe2+ at nanomolar levels (LOD of 35 nM), accompanied by significant absorption and emission shifts, along with colorimetric demarcation. Both fluorophores exhibit an excellent "turn-on" emission response upon encountering Fe2+ in the cells. Flow cytometry and confocal fluorescence imaging studies demonstrate enhanced fluorescence signals in response to labile iron, efficiently detecting heme during erastin-induced ferroptosis. Interestingly, we also observed that the product formed after Fe2+ sensing localizes within the lipid droplets. These water-soluble and highly sensitive reactive probes, NOPy and NOCN, enable investigations of iron-dependent physiological and pathological conditions. The development of these probes represents an advancement in the field, offering a rapid and selective means for detecting Fe2+ with minimal cytotoxicity.
Collapse
Affiliation(s)
- Yogesh Dubey
- Department of Chemistry, Indian Institute of Technology Gandhinagar, Palaj, Gandhinagar, Gujarat-382055, India.
| | - Shabnam Mansuri
- Department of Chemistry, Indian Institute of Technology Gandhinagar, Palaj, Gandhinagar, Gujarat-382055, India.
| | - Sriram Kanvah
- Department of Chemistry, Indian Institute of Technology Gandhinagar, Palaj, Gandhinagar, Gujarat-382055, India.
| |
Collapse
|
24
|
Lewis NM, Kisgeropoulos EC, Lubner CE, Fixen KR. Characterization of ferredoxins involved in electron transfer pathways for nitrogen fixation implicates differences in electronic structure in tuning 2[4Fe4S] Fd activity. J Inorg Biochem 2024; 254:112521. [PMID: 38471286 DOI: 10.1016/j.jinorgbio.2024.112521] [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: 10/11/2023] [Revised: 03/03/2024] [Accepted: 03/05/2024] [Indexed: 03/14/2024]
Abstract
Ferredoxins (Fds) are small proteins which shuttle electrons to pathways like biological nitrogen fixation. Physical properties tune the reactivity of Fds with different pathways, but knowledge on how these properties can be manipulated to engineer new electron transfer pathways is lacking. Recently, we showed that an evolved strain of Rhodopseudomonas palustris uses a new electron transfer pathway for nitrogen fixation. This pathway involves a variant of the primary Fd of nitrogen fixation in R. palustris, Fer1, in which threonine at position 11 is substituted for isoleucine (Fer1T11I). To understand why this substitution in Fer1 enables more efficient electron transfer, we used in vivo and in vitro methods to characterize Fer1 and Fer1T11I. Electrochemical characterization revealed both Fer1 and Fer1T11I have similar redox transitions (-480 mV and - 550 mV), indicating the reduction potential was unaffected despite the proximity of T11 to an iron‑sulfur (FeS) cluster of Fer1. Additionally, disruption of hydrogen bonding around an FeS cluster in Fer1 by substituting threonine with alanine (T11A) or valine (T11V) did not increase nitrogenase activity, indicating that disruption of hydrogen bonding does not explain the difference in activity observed for Fer1T11I. Electron paramagnetic resonance spectroscopy studies revealed key differences in the electronic structure of Fer1 and Fer1T11I, which indicate changes to the high spin states and/or spin-spin coupling between the FeS clusters of Fer1. Our data implicates these electronic structure differences in facilitating electron flow and sets a foundation for further investigations to understand the connection between these properties and intermolecular electron transfer.
Collapse
Affiliation(s)
- Nathan M Lewis
- Department of Plant and Microbial Biology and the Biotechnology Institute, University of Minnesota, Minneapolis, MN, United States of America
| | | | - Carolyn E Lubner
- National Renewable Energy Laboratory, Golden, CO, United States of America.
| | - Kathryn R Fixen
- Department of Plant and Microbial Biology and the Biotechnology Institute, University of Minnesota, Minneapolis, MN, United States of America.
| |
Collapse
|
25
|
Marcora MS, Mattera VS, Goñi P, Aybar F, Correale JD, Pasquini JM. Iron deficiency in astrocytes alters cellular status and impacts on oligodendrocyte differentiation. J Neurosci Res 2024; 102:e25334. [PMID: 38656648 DOI: 10.1002/jnr.25334] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Revised: 04/01/2024] [Accepted: 04/04/2024] [Indexed: 04/26/2024]
Abstract
Iron deficiency (ID) has been shown to affect central nervous system (CNS) development and induce hypomyelination. Previous work from our laboratory in a gestational ID model showed that both oligodendrocyte (OLG) and astrocyte (AST) maturation was impaired. To explore the contribution of AST iron to the myelination process, we generated an in vitro ID model by silencing divalent metal transporter 1 (DMT1) in AST (siDMT1 AST) or treating AST with Fe3+ chelator deferoxamine (DFX; DFX AST). siDMT1 AST showed no changes in proliferation but remained immature. Co-cultures of oligodendrocyte precursors cells (OPC) with siDMT1 AST and OPC cultures incubated with siDMT1 AST-conditioned media (ACM) rendered a reduction in OPC maturation. These findings correlated with a decrease in the expression of AST-secreted factors IGF-1, NRG-1, and LIF, known to promote OPC differentiation. siDMT1 AST also displayed increased mitochondrial number and reduced mitochondrial size as compared to control cells. DFX AST also remained immature and DFX AST-conditioned media also hampered OPC maturation in culture, in keeping with a decrease in the expression of AST-secreted growth factors IGF-1, NRG-1, LIF, and CNTF. DFX AST mitochondrial morphology and number showed results similar to those observed in siDMT1 AST. In sum, our results show that ID, induced through two different methods, impacts AST maturation and mitochondrial functioning, which in turn hampers OPC differentiation.
Collapse
Affiliation(s)
- María Silvina Marcora
- Departamento de Química Biológica e Instituto de Química y Fisicoquímica Biológicas (IQUIFIB), Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires-CONICET, Buenos Aires, Argentina
| | - Vanesa Soledad Mattera
- Departamento de Química Biológica e Instituto de Química y Fisicoquímica Biológicas (IQUIFIB), Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires-CONICET, Buenos Aires, Argentina
| | - Pilar Goñi
- Departamento de Química Biológica e Instituto de Química y Fisicoquímica Biológicas (IQUIFIB), Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires-CONICET, Buenos Aires, Argentina
| | - Florencia Aybar
- Departamento de Química Biológica e Instituto de Química y Fisicoquímica Biológicas (IQUIFIB), Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires-CONICET, Buenos Aires, Argentina
| | - Jorge Daniel Correale
- Departamento de Neurología, Fleni e Instituto de Química y Fisicoquímica Biológicas (IQUIFIB), Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires-CONICET, Buenos Aires, Argentina
| | - Juana Maria Pasquini
- Departamento de Química Biológica e Instituto de Química y Fisicoquímica Biológicas (IQUIFIB), Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires-CONICET, Buenos Aires, Argentina
| |
Collapse
|
26
|
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
|
27
|
Jangid N, Sharma A, Srivastava N. Potential involvement of ferroptosis in BPA-induced neurotoxicity: An in vitro study. ENVIRONMENTAL TOXICOLOGY AND PHARMACOLOGY 2024; 106:104355. [PMID: 38154758 DOI: 10.1016/j.etap.2023.104355] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/23/2023] [Accepted: 12/22/2023] [Indexed: 12/30/2023]
Abstract
BACKGROUND Ferroptosis is a newly recognized cell death pathway having distinct characteristics compared to traditional cell death pathways such as apoptosis, necroptosis, or autophagy. However, the potential involvement of ferroptosis in bisphenol A (BPA)-induced neurotoxicity has not been well explored so far. In present study, we analyzed the relationship between ferroptosis and BPA-induced neurotoxicity. METHODS In this study, a human neuroblastoma cell line, SH-SY5Y, was treated with BPA, ferrostatin-1 (FS-1, ferroptosis inhibitor) and RSL-3 (ferroptosis inducer). The cell viability was measured using MTT assay. Additionally, the levels of lipid peroxidation, total iron content, reactive oxygen species (ROS) generation, and nitrite content were measured to evaluate the key markers of ferroptosis. To further confirm the involvement of ferroptosis in BPA-induced neurotoxicity, other ferroptosis markers such as glutathione peroxidase (GPx) activity, total glutathione contents and antioxidant parameters were also evaluated. RESULTS The cell viability of SH-SY5Y cells was down-regulated by BPA treatment in a concentration-dependent manner, the cell viability at 0.1 µM concentration was 97.63% whereas at highest BPA concentration i.e. 10 µM, the cell viability was 86.05% (p < 0.0001). Also the antioxidant parameters including catalase and superoxide dismutase activity of neuronal cells were down-regulated upon BPA exposure. However, the levels of lipid peroxidation, total iron, reactive oxygen species, and nitrite contents were increased in a concentration-dependent manner which could be rescued by FS-1 and exacerbated by RSL-3. The total iron in SH-SY5Y cells at 0.1 µM concentration was found to be 1.2 fold (p < 0.05) of control and at highest BPA concentration total iron was about 1.41 fold (p < 0.001) of control. CONCLUSIONS The present study indicated that, ferroptosis plays an important role in the progression of BPA-induced neurotoxicity, and ferroptosis may become a novel target in the treatment of various neurological disorders.
Collapse
Affiliation(s)
- Nita Jangid
- Department of Biotechnology, National Institute of Pharmaceutical Education and Research-Raebareli, Bijnor-Sisendi Road, Post Office Mati, Lucknow 226002, India
| | - Ankita Sharma
- Department of Biotechnology, National Institute of Pharmaceutical Education and Research-Raebareli, Bijnor-Sisendi Road, Post Office Mati, Lucknow 226002, India.
| | - Nidhi Srivastava
- Department of Biotechnology, National Institute of Pharmaceutical Education and Research-Raebareli, Bijnor-Sisendi Road, Post Office Mati, Lucknow 226002, India.
| |
Collapse
|
28
|
Sharifian Gh. M, Norouzi F, Sorci M, Zaid TS, Pier GB, Achimovich A, Ongwae GM, Liang B, Ryan M, Lemke M, Belfort G, Gadjeva M, Gahlmann A, Pires MM, Venter H, Harris TE, Laurie GW. Targeting Iron - Respiratory Reciprocity Promotes Bacterial Death. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.01.582947. [PMID: 38464199 PMCID: PMC10925246 DOI: 10.1101/2024.03.01.582947] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/12/2024]
Abstract
Discovering new bacterial signaling pathways offers unique antibiotic strategies. Here, through an unbiased resistance screen of 3,884 gene knockout strains, we uncovered a previously unknown non-lytic bactericidal mechanism that sequentially couples three transporters and downstream transcription to lethally suppress respiration of the highly virulent P. aeruginosa strain PA14 - one of three species on the WHO's 'Priority 1: Critical' list. By targeting outer membrane YaiW, cationic lacritin peptide 'N-104' translocates into the periplasm where it ligates outer loops 4 and 2 of the inner membrane transporters FeoB and PotH, respectively, to suppress both ferrous iron and polyamine uptake. This broadly shuts down transcription of many biofilm-associated genes, including ferrous iron-dependent TauD and ExbB1. The mechanism is innate to the surface of the eye and is enhanced by synergistic coupling with thrombin peptide GKY20. This is the first example of an inhibitor of multiple bacterial transporters.
Collapse
Affiliation(s)
| | - Fatemeh Norouzi
- Department of Cell Biology, University of Virginia, Charlottesville VA, USA
| | - Mirco Sorci
- Howard P. Isermann Department of Chemical and Biological Engineering, Rensselaer Polytechnic Institute, Troy NY, USA
| | - Tanweer S Zaid
- Division of Infectious Disease, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston MA
| | - Gerald B. Pier
- Division of Infectious Disease, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston MA
| | - Alecia Achimovich
- Department of Chemistry, University of Virginia, Charlottesville VA, USA
| | - George M. Ongwae
- Department of Chemistry, University of Virginia, Charlottesville VA, USA
| | - Binyong Liang
- Department of Molecular Physiology and Biological Physics, University of Virginia, Charlottesville VA, USA
| | - Margaret Ryan
- Department of Cell Biology, University of Virginia, Charlottesville VA, USA
| | - Michael Lemke
- Department of Pharmacology, University of Virginia, Charlottesville VA, USA
| | - Georges Belfort
- Howard P. Isermann Department of Chemical and Biological Engineering, Rensselaer Polytechnic Institute, Troy NY, USA
| | - Mihaela Gadjeva
- Division of Infectious Disease, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston MA
| | - Andreas Gahlmann
- Department of Chemistry, University of Virginia, Charlottesville VA, USA
| | - Marcos M. Pires
- Department of Chemistry, University of Virginia, Charlottesville VA, USA
| | - Henrietta Venter
- Sansom Institute for Health Research, University of South Australia, Adelaide, Australia
| | - Thurl E. Harris
- Department of Pharmacology, University of Virginia, Charlottesville VA, USA
| | - Gordon W. Laurie
- Department of Cell Biology, University of Virginia, Charlottesville VA, USA
- Department of Ophthalmology, University of Virginia, Charlottesville VA, USA
- Department of Biomedical Engineering, University of Virginia, Charlottesville VA, USA
- Contact author: Gordon Laurie
| |
Collapse
|
29
|
Dong CL, Wu T, Dong Y, Qu QW, Chen XY, Li YH. Exogenous methionine contributes to reversing the resistance of Streptococcus suis to macrolides. Microbiol Spectr 2024; 12:e0280323. [PMID: 38230928 PMCID: PMC10923279 DOI: 10.1128/spectrum.02803-23] [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: 07/10/2023] [Accepted: 12/21/2023] [Indexed: 01/18/2024] Open
Abstract
Streptococcus suis (S. suis) has been increasingly recognized as a porcine zoonotic pathogen that threatens the health of both pigs and humans. Multidrug-resistant Streptococcus suis is becoming increasingly prevalent, and novel strategies to treat bacterial infections caused by these organisms are desperately needed. In the present study, an untargeted metabolomics analysis showed that the significant decrease in methionine content and the methionine biosynthetic pathway were significantly affected by the Kyoto Encyclopedia of Genes and Genomes pathway enrichment analysis in drug-resistant S. suis. The addition of L-methionine restored the bactericidal activity of macrolides, doxycycline, and ciprofloxacin on S. suis in vivo and in vitro. Further studies showed that the exogenous addition of methionine affects methionine metabolism by reducing S-adenosylmethionine synthetase activity and the contents of S-adenosylmethionine, S-adenosyl homocysteine, and S-ribose homocysteine. Methionine can decrease the total methylation level and methylesterase activity in multidrug resistant S. suis. The drug transport proteins and efflux pump genes were significantly downregulated in S. suis by exogenous L-methionine. Moreover, the exogenous addition of methionine can reduce the survival of S. suis by affecting oxidative stress and metal starvation in bacteria. Thus, L-methionine may influence the development of resistance in S. suis through methyl metabolism and metal starvation. This study provides a new perspective on the mitigation of drug resistance in S. suis.IMPORTANCEBacterial antibiotic resistance has become a severe threat to human and animal health. Increasing the efficacy of existing antibiotics is a promising strategy against antibiotic resistance. Here, we report that L-methionine enhances the efficacy of macrolides, doxycycline, and ciprofloxacin antibiotics in killing Streptococcus suis, including multidrug-resistant pathogens. We investigated the mechanism of action of exogenous methionine supplementation in restoring macrolides in Streptococcus suis and the role of the methionine cycle pathway on methylation levels, efflux pump genes, oxidative stress, and metal starvation in Streptococcus suis. It provides a theoretical basis for the rational use of macrolides in clinical practice and also identifies a possible target for restoring drug resistance in Streptococcus suis.
Collapse
Affiliation(s)
- Chun-Liu Dong
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, Heilongjiang, China
- Heilongjiang Key Laboratory for Animal Disease Control and Pharmaceutical Development, Harbin, Heilongjiang, China
| | - Tong Wu
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, Heilongjiang, China
| | - Yue Dong
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, Heilongjiang, China
| | - Qian-Wei Qu
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, Heilongjiang, China
| | - Xue-Ying Chen
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, Heilongjiang, China
- Heilongjiang Key Laboratory for Animal Disease Control and Pharmaceutical Development, Harbin, Heilongjiang, China
| | - Yan-Hua Li
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, Heilongjiang, China
- Heilongjiang Key Laboratory for Animal Disease Control and Pharmaceutical Development, Harbin, Heilongjiang, China
| |
Collapse
|
30
|
Sharifabad ME, Soucaille R, Wang X, Rotherham M, Loughran T, Everett J, Cabrera D, Yang Y, Hicken R, Telling N. Optical Microscopy Using the Faraday Effect Reveals in Situ Magnetization Dynamics of Magnetic Nanoparticles in Biological Samples. ACS NANO 2024. [PMID: 38315113 PMCID: PMC10883041 DOI: 10.1021/acsnano.3c08955] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2024]
Abstract
The study of exogenous and endogenous nanoscale magnetic material in biology is important for developing biomedical nanotechnology as well as for understanding fundamental biological processes such as iron metabolism and biomineralization. Here, we exploit the magneto-optical Faraday effect to probe intracellular magnetic properties and perform magnetic imaging, revealing the location-specific magnetization dynamics of exogenous magnetic nanoparticles within cells. The opportunities enabled by this method are shown in the context of magnetic hyperthermia; an effect where local heating is generated in magnetic nanoparticles exposed to high-frequency AC magnetic fields. Magnetic hyperthermia has the potential to be used as a cellular-level thermotherapy for cancer, as well as for other biomedical applications that target heat-sensitive cellular function. However, previous experiments have suggested that the cellular environment modifies the magnetization dynamics of nanoparticles, thus dramatically altering their heating efficiency. By combining magneto-optical and fluorescence measurements, we demonstrate a form of biological microscopy that we used here to study the magnetization dynamics of nanoparticles in situ, in both histological samples and living cancer cells. Correlative magnetic and fluorescence imaging identified aggregated magnetic nanoparticles colocalized with cellular lysosomes. Nanoparticles aggregated within these lysosomes displayed reduced AC magnetic coercivity compared to the same particles measured in an aqueous suspension or aggregated in other areas of the cells. Such measurements reveal the power of this approach, enabling investigations of how cellular location, nanoparticle aggregation, and interparticle magnetic interactions affect the magnetization dynamics and consequently the heating response of nanoparticles in the biological milieu.
Collapse
Affiliation(s)
- Maneea Eizadi Sharifabad
- School of Pharmacy and Bioengineering, Keele University, Guy Hilton Research Centre, Thornburrow Drive, Stoke-on-Trent ST4 7QB, United Kingdom
| | - Rémy Soucaille
- Department of Physics and Astronomy, University of Exeter, Stocker Road, Exeter EX4 4QL, United Kingdom
| | - Xuyiling Wang
- School of Pharmacy and Bioengineering, Keele University, Guy Hilton Research Centre, Thornburrow Drive, Stoke-on-Trent ST4 7QB, United Kingdom
| | - Michael Rotherham
- School of Pharmacy and Bioengineering, Keele University, Guy Hilton Research Centre, Thornburrow Drive, Stoke-on-Trent ST4 7QB, United Kingdom
- Healthcare Technologies Institute, School of Chemical Engineering, University of Birmingham, Heritage Building, Mindelsohn Way, Edgbaston, Birmingham B15 2TH, United Kingdom
| | - Tom Loughran
- Department of Physics and Astronomy, University of Exeter, Stocker Road, Exeter EX4 4QL, United Kingdom
| | - James Everett
- School of Pharmacy and Bioengineering, Keele University, Guy Hilton Research Centre, Thornburrow Drive, Stoke-on-Trent ST4 7QB, United Kingdom
| | - David Cabrera
- School of Pharmacy and Bioengineering, Keele University, Guy Hilton Research Centre, Thornburrow Drive, Stoke-on-Trent ST4 7QB, United Kingdom
| | - Ying Yang
- School of Pharmacy and Bioengineering, Keele University, Guy Hilton Research Centre, Thornburrow Drive, Stoke-on-Trent ST4 7QB, United Kingdom
| | - Robert Hicken
- Department of Physics and Astronomy, University of Exeter, Stocker Road, Exeter EX4 4QL, United Kingdom
| | - Neil Telling
- School of Pharmacy and Bioengineering, Keele University, Guy Hilton Research Centre, Thornburrow Drive, Stoke-on-Trent ST4 7QB, United Kingdom
| |
Collapse
|
31
|
Mannella V, Chaabane L, Canu T, Zanardi A, Raia S, Conti A, Ferrini B, Caricasole A, Musco G, Alessio M. Lipid dysmetabolism in ceruloplasmin-deficient mice revealed both in vivo and ex vivo by MRI, MRS and NMR analyses. FEBS Open Bio 2024; 14:258-275. [PMID: 37986139 PMCID: PMC10839333 DOI: 10.1002/2211-5463.13740] [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: 10/27/2023] [Accepted: 11/17/2023] [Indexed: 11/22/2023] Open
Abstract
Ceruloplasmin (Cp) is a ferroxidase that plays a role in cellular iron homeostasis and is mainly expressed in the liver and secreted into the blood. Cp is also produced by adipose tissue, which releases it as an adipokine. Although a dysfunctional interaction of iron with the metabolism of lipids has been associated with several metabolic diseases, the role of Cp in adipose tissue metabolism and in the interplay between hepatocytes and adipocytes has been poorly investigated. We previously found that Cp-deficient (CpKO) mice become overweight and demonstrate adipose tissue accumulation together with liver steatosis during aging, suggestive of lipid dysmetabolism. In the present study, we investigated the lipid alterations which occur during aging in adipose tissue and liver of CpKO and wild-type mice both in vivo and ex vivo. During aging of CpKO mice, we observed adipose tissue accumulation and liver lipid deposition, both of which are associated with macrophage infiltration. Liver lipid deposition was characterized by accumulation of triglycerides, fatty acids and ω-3 fatty acids, as well as by a switch from unsaturated to saturated fatty acids, which is characteristic of lipid storage. Liver steatosis was preceded by iron deposition and macrophage infiltration, and this was observed to be already occurring in younger CpKO mice. The accumulation of ω-3 fatty acids, which can only be acquired through diet, was associated with body weight increase in CpKO mice despite food intake being equal to that of wild-type mice, thus underlining the alterations in lipid metabolism/catabolism in Cp-deficient animals.
Collapse
Affiliation(s)
- Valeria Mannella
- COSR‐Centre for Omics SciencesIRCCS‐San Raffaele HospitalMilanoItaly
| | - Linda Chaabane
- Preclinical Imaging, Experimental Imaging CentreIRCCS‐San Raffaele HospitalMilanoItaly
- Present address:
LC, Euro‐BioImaging ERIC, Med‐Hub section, Institute of Biostructures and Bioimaging (IBB)Italian National Research Council (CNR)TorinoItaly
- Present address:
SR, Deloitte & Touche SpAMilanoItaly
| | - Tamara Canu
- Preclinical Imaging, Experimental Imaging CentreIRCCS‐San Raffaele HospitalMilanoItaly
| | - Alan Zanardi
- Proteome Biochemistry, COSR‐Centre for Omics SciencesIRCCS‐San Raffaele HospitalMilanoItaly
| | - Sara Raia
- Proteome Biochemistry, COSR‐Centre for Omics SciencesIRCCS‐San Raffaele HospitalMilanoItaly
- Present address:
LC, Euro‐BioImaging ERIC, Med‐Hub section, Institute of Biostructures and Bioimaging (IBB)Italian National Research Council (CNR)TorinoItaly
- Present address:
SR, Deloitte & Touche SpAMilanoItaly
| | - Antonio Conti
- Proteome Biochemistry, COSR‐Centre for Omics SciencesIRCCS‐San Raffaele HospitalMilanoItaly
| | - Barbara Ferrini
- Proteome Biochemistry, COSR‐Centre for Omics SciencesIRCCS‐San Raffaele HospitalMilanoItaly
| | - Andrea Caricasole
- Department of Research & Innovation, Kedrion S.p.A.Loc BolognanaGallicanoItaly
| | - Giovanna Musco
- Biomolecular Nuclear Magnetic Resonance, Division of Genetics and Cell BiologyIRCCS‐San Raffaele HospitalMilanoItaly
| | - Massimo Alessio
- Proteome Biochemistry, COSR‐Centre for Omics SciencesIRCCS‐San Raffaele HospitalMilanoItaly
| |
Collapse
|
32
|
Stefos GC, Dalaka E, Papoutsi G, Palamidi I, Andreou V, Katsaros G, Bossis I, Politis I, Theodorou G. In vitro evaluation of the effect of yogurt acid whey fractions on iron bioavailability. J Dairy Sci 2024; 107:683-694. [PMID: 37709016 DOI: 10.3168/jds.2023-23643] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Accepted: 08/28/2023] [Indexed: 09/16/2023]
Abstract
A side effect of the raised consumption of Greek yogurt is the generation of massive amounts of yogurt acid whey (YAW). The dairy industry has tried several methods for handling these quantities, which constitute an environmental problem. Although the protein content of YAW is relatively low, given the huge amounts of produced YAW, the final protein amount in the produced YAW should not be underestimated. Taking into consideration the increased interest for bioactive peptides and the increased demand for dietary proteins, combined with protein and peptides content of YAW, efforts should be made toward reintroducing the latter in the food supply chain. In this context and in view of the prevalent dietary iron deficiency problem, the objective of the present study was the investigation of YAW fractions' effect on Fe bioavailability. With this purpose, an in vitro digest approach, following the INFOGEST protocol, was coupled with the Caco2 cell model. To evaluate whether YAW digest fractions exert positive, negative or neutral effect on Fe bioavailability, they were compared with the ones derived from milk, a well-studied food in this context. Milk and YAW showed the same effectiveness on both Fe bioavailability and the expression of relative genes (DCYTB, DMT1, FPN1, and HEPH). Focusing further on YAW fractions, by comparison with their blank digest control counterparts, it resulted that YAW 3- to 10-kDa digests fraction had a superior effect over the 0- to 3-kDa fraction on Fe-uptake, which was accompanied by a similar effect on the expression of Fe metabolism-related genes (DCYTB, FPN1, and HEPH). Finally, although the 3- to 10-kDa fraction of bovine YAW digests resulted in a nonsignificant increased Fe uptake, compared with the ovine and caprine YAW, the expression of DCYTB and FPN1 genes underlined this difference by showing a similar pattern with statistically significant higher expression of bovine compared with ovine and bovine compared with both ovine and caprine, respectively. The present study deals with the novel concept that YAW may contain factors affecting Fe bioavailability. The results show that it does not exert any negative effect and support the extensive investigation for specific peptides with positive effect as well as that YAW proteins should be further assessed on the prospect that they can be used in human nutrition.
Collapse
Affiliation(s)
- Georgios C Stefos
- Laboratory of Animal Breeding and Husbandry, Department of Animal Science, Agricultural University of Athens, 118 55 Athens, Greece.
| | - Eleni Dalaka
- Laboratory of Animal Breeding and Husbandry, Department of Animal Science, Agricultural University of Athens, 118 55 Athens, Greece
| | - Georgia Papoutsi
- Laboratory of Animal Breeding and Husbandry, Department of Animal Science, Agricultural University of Athens, 118 55 Athens, Greece
| | - Irida Palamidi
- Laboratory of Nutritional Physiology & Feeding, Department of Animal Science, Agricultural University of Athens, 112 55 Athens, Greece
| | - Varvara Andreou
- Institute of Technology of Agricultural Products, Hellenic Agricultural Organization-DEMETER, Lykovrissi 14123, Attica, Greece
| | - George Katsaros
- Institute of Technology of Agricultural Products, Hellenic Agricultural Organization-DEMETER, Lykovrissi 14123, Attica, Greece
| | - Ioannis Bossis
- Laboratory of Animal Husbandry, Department of Animal Production, School of Agriculture, Faculty of Agriculture, Forestry and Natural Environment, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece
| | - Ioannis Politis
- Laboratory of Animal Breeding and Husbandry, Department of Animal Science, Agricultural University of Athens, 118 55 Athens, Greece
| | - Georgios Theodorou
- Laboratory of Animal Breeding and Husbandry, Department of Animal Science, Agricultural University of Athens, 118 55 Athens, Greece.
| |
Collapse
|
33
|
Zhao H, Lu Y, Zhang J, Sun Z, Cheng C, Liu Y, Wu L, Zhang M, He W, Hao S, Li K. NCOA4 requires a [3Fe-4S] to sense and maintain the iron homeostasis. J Biol Chem 2024; 300:105612. [PMID: 38159858 PMCID: PMC10831263 DOI: 10.1016/j.jbc.2023.105612] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2023] [Revised: 12/04/2023] [Accepted: 12/22/2023] [Indexed: 01/03/2024] Open
Abstract
NCOA4 is a selective cargo receptor for ferritinophagy, the autophagic turnover of ferritin (FTH), a process critical for regulating intracellular iron bioavailability. However, how ferritinophagy flux is controlled through NCOA4 in iron-dependent processes needs to be better understood. Here, we show that the C-terminal FTH-binding domain of NCOA4 harbors a [3Fe-4S]-binding site with a stoichiometry of approximately one labile [3Fe-4S] cluster per NCOA4 monomer. By analyzing the interaction between NCOA4 and HERC2 ubiquitin ligase or NCOA4 and FTH, we demonstrate that NCOA4 regulates ferritinophagy by sensing the intracellular iron-sulfur cluster levels. Under iron-repletion conditions, HERC2 recognizes and recruits holo-NCOA4 as a substrate for polyubiquitination and degradation, favoring ferritin iron storage. Under iron-depletion conditions, NCOA4 exists in the form of apo-protein and binds ferritin to promote the occurrence of ferritinophagy and release iron. Thus, we identify an iron-sulfur cluster [3Fe-4S] as a critical cofactor in determining the fate of NCOA4 in favoring iron storage in ferritin or iron release via ferritinophagy and provide a dual mechanism for selective interaction between HERC2 and [3Fe-4S]-NCOA4 for proteasomal degradation or between ferritin and apo-NCOA4 for ferritinophagy in the control of iron homeostasis.
Collapse
Affiliation(s)
- Hongting Zhao
- State Key Laboratory of Pharmaceutical Biotechnology, Jiangsu Key Laboratory of Molecular Medicine, Medical School, Nanjing University, Nanjing, China
| | - Yao Lu
- State Key Laboratory of Pharmaceutical Biotechnology, Jiangsu Key Laboratory of Molecular Medicine, Medical School, Nanjing University, Nanjing, China
| | - Jinghua Zhang
- State Key Laboratory of Pharmaceutical Biotechnology, Jiangsu Key Laboratory of Molecular Medicine, Medical School, Nanjing University, Nanjing, China
| | - Zichen Sun
- State Key Laboratory of Pharmaceutical Biotechnology, Jiangsu Key Laboratory of Molecular Medicine, Medical School, Nanjing University, Nanjing, China
| | - Chen Cheng
- State Key Laboratory of Pharmaceutical Biotechnology, Jiangsu Key Laboratory of Molecular Medicine, Medical School, Nanjing University, Nanjing, China
| | - Yutong Liu
- State Key Laboratory of Pharmaceutical Biotechnology, Jiangsu Key Laboratory of Molecular Medicine, Medical School, Nanjing University, Nanjing, China
| | - Lin Wu
- State Key Laboratory of Pharmaceutical Biotechnology, Jiangsu Key Laboratory of Molecular Medicine, Medical School, Nanjing University, Nanjing, China
| | - Meng Zhang
- Department of General Surgery, Nanjing Drum Tower Hospital Clinical College of Nanjing Medical University, Nanjing, China
| | - Weijiang He
- School of Chemistry and Chemical Engineering, Chemistry and Biomedicine Innovation Center (ChemBIC), Nanjing University, Nanjing, China
| | - Shuangying Hao
- School of Medicine, Henan Polytechnic University, Jiaozuo, China.
| | - Kuanyu Li
- State Key Laboratory of Pharmaceutical Biotechnology, Jiangsu Key Laboratory of Molecular Medicine, Medical School, Nanjing University, Nanjing, China.
| |
Collapse
|
34
|
Singh G, Singh A, Mishra S, Singh D, Kumar A. Intracellular Iron Accumulation Induces Inflammatory and Oxidative Status of the Host After Japanese Encephalitis Viral Infection. Mol Neurobiol 2024; 61:175-187. [PMID: 37594653 DOI: 10.1007/s12035-023-03538-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2023] [Accepted: 07/18/2023] [Indexed: 08/19/2023]
Abstract
The factors mitigating the microglia/macrophage activation and inflammatory damage in Japanese encephalitis (JE) virus infected CNS are still being ascertained. We aim to characterize the changes in iron transporter and iron storage proteins along with inflammatory and oxidative stress-mediated signaling during the JE viral infection. Cortical tissue samples from mice with JE viral infection were processed for biochemical, histological, and molecular analysis. Iron storage protein, i.e., ferritin, was found significantly increased post-JE viral infection, and iron accumulation was noted in cortical tissue. Key proinflammatory associated markers, such as TNF-α, IL-6, and its regulator TLR4, were found to be increased, while SOCS1 (anti-inflammatory regulator) transcription decreased with increased levels of oxidative stress markers NOX2-mediated NF-ΚB/p65 and protein carbonyl. Furthermore, it is noted that hepcidin level increased and ferroportin level decreased, and iron transporter gene expression got imbalanced after JE viral infection. This observation was further confirmed by deferoxamine (DFO) treatment to JE viral infection mice model, where the decline in hepcidin transcription level and iron load in cortical tissue of JE viral infected animals was noted. However, no change was found in the ferroportin level compared to JE viral infected animals. Together, these findings suggest that iron overload and hepcidin-ferroportin regulation are involved in JE viral infection disease pathologies and associated with the inflammatory and oxidative status of the host during infection.
Collapse
Affiliation(s)
- Gajendra Singh
- Department of Molecular Medicine and Biotechnology, Sanjay Gandhi Postgraduate Institute of Medical Sciences, Lucknow, 226014, Uttar Pradesh, India
| | - Anjali Singh
- Department of Molecular Medicine and Biotechnology, Sanjay Gandhi Postgraduate Institute of Medical Sciences, Lucknow, 226014, Uttar Pradesh, India
| | - Sanjay Mishra
- Department of Pathology, King George's Medical University, Lucknow, 226003, Uttar Pradesh, India
| | - Devendra Singh
- Department of Pathology, King George's Medical University, Lucknow, 226003, Uttar Pradesh, India
| | - Alok Kumar
- Department of Molecular Medicine and Biotechnology, Sanjay Gandhi Postgraduate Institute of Medical Sciences, Lucknow, 226014, Uttar Pradesh, India.
| |
Collapse
|
35
|
Chalivendra S, Shi S, Li X, Kuang Z, Giovinazzo J, Zhang L, Rossi J, Saviola AJ, Wang J, Welty R, Liu S, Vaeth K, Zhou ZH, Hansen KC, Taliaferro JM, Zhao R. Selected humanization of yeast U1 snRNP leads to global suppression of pre-mRNA splicing and mitochondrial dysfunction in the budding yeast. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.12.15.571893. [PMID: 38168357 PMCID: PMC10760170 DOI: 10.1101/2023.12.15.571893] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2024]
Abstract
The recognition of 5' splice site (5' ss) is one of the earliest steps of pre-mRNA splicing. To better understand the mechanism and regulation of 5' ss recognition, we selectively humanized components of the yeast U1 snRNP to reveal the function of these components in 5' ss recognition and splicing. We targeted U1C and Luc7, two proteins that interact with and stabilize the yeast U1 (yU1) snRNA and the 5' ss RNA duplex. We replaced the Zinc-Finger (ZnF) domain of yU1C with its human counterpart, which resulted in cold-sensitive growth phenotype and moderate splicing defects. Next, we added an auxin-inducible degron to yLuc7 protein and found that Luc7-depleted yU1 snRNP resulted in the concomitant loss of PRP40 and Snu71 (two other essential yeast U1 snRNP proteins), and further biochemical analyses suggest a model of how these three proteins interact with each other in the U1 snRNP. The loss of these proteins resulted in a significant growth retardation accompanied by a global suppression of pre-mRNA splicing. The splicing suppression led to mitochondrial dysfunction as revealed by a release of Fe 2+ into the growth medium and an induction of mitochondrial reactive oxygen species. Together, these observations indicate that the human U1C ZnF can substitute that of yeast, Luc7 is essential for the incorporation of the Luc7-Prp40-Snu71 trimer into yeast U1 snRNP, and splicing plays a major role in the regulation of mitochondria function in yeast.
Collapse
|
36
|
Lafuente-Gómez N, de Lázaro I, Dhanjani M, García-Soriano D, Sobral MC, Salas G, Mooney DJ, Somoza Á. Multifunctional magnetic nanoparticles elicit anti-tumor immunity in a mouse melanoma model. Mater Today Bio 2023; 23:100817. [PMID: 37822453 PMCID: PMC10562177 DOI: 10.1016/j.mtbio.2023.100817] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Revised: 09/19/2023] [Accepted: 09/22/2023] [Indexed: 10/13/2023] Open
Abstract
Immunotherapy has emerged as a promising strategy to eradicate cancer cells. Particularly, the development of cancer vaccines to induce a potent and sustained antigen-specific T cell response has become a center of attention. Herein, we describe a novel immunotherapy based on magnetic nanoparticles (MNP) covalently modified with the OVA254-267 antigen and a CpG oligonucleotide via disulfide bonds. The MNP-CpG-COVA significantly enhances dendritic cell activation and CD8+ T cell antitumoral response against B16-OVA melanoma cells in vitro. Notably, the immune response induced by the covalently modified MNP is more potent and sustained over time than that triggered by the free components, highlighting the advantage of nanoformulations in immunotherapies. What is more, the nanoparticles are stable in the blood after in vivo administration and induce potent levels of systemic tumor-specific effector CD8 + T cells. Overall, our findings highlight the potential of covalently functionalized MNP to induce robust immune responses against mouse melanoma.
Collapse
Affiliation(s)
- Nuria Lafuente-Gómez
- Instituto Madrileño de Estudios Avanzados en Nanociencia (IMDEA Nanociencia), Madrid, 28049, Spain
- Harvard John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, 02138, USA
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, 02115, USA
- Immunology Service, Hospital Universitario de la Princesa, Instituto Investigación Sanitaria Princesa, Madrid, 28006, Spain
| | - Irene de Lázaro
- Harvard John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, 02138, USA
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, 02115, USA
- Department of Biomedical Engineering, NYU Tandon School of Engineering, New York University, New York, NY, 10010, USA
- NYU Cardiovascular Research Center, Division of Cardiology, Department of Medicine, NYU Grossman School of Medicine, New York University, New York, NY, 10010, USA
| | - Mónica Dhanjani
- Instituto Madrileño de Estudios Avanzados en Nanociencia (IMDEA Nanociencia), Madrid, 28049, Spain
| | - David García-Soriano
- Instituto Madrileño de Estudios Avanzados en Nanociencia (IMDEA Nanociencia), Madrid, 28049, Spain
| | - Miguel C. Sobral
- Harvard John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, 02138, USA
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, 02115, USA
| | - Gorka Salas
- Instituto Madrileño de Estudios Avanzados en Nanociencia (IMDEA Nanociencia), Madrid, 28049, Spain
- Unidad de Nanobiotecnología Asociada al Centro Nacional de Biotecnología (CNB-CSIC), Madrid, 28049, Spain
| | - David J. Mooney
- Harvard John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, 02138, USA
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, 02115, USA
| | - Álvaro Somoza
- Instituto Madrileño de Estudios Avanzados en Nanociencia (IMDEA Nanociencia), Madrid, 28049, Spain
- Unidad de Nanobiotecnología Asociada al Centro Nacional de Biotecnología (CNB-CSIC), Madrid, 28049, Spain
| |
Collapse
|
37
|
Baker IR, Matzen SL, Schuler CJ, Toner BM, Girguis PR. Aerobic iron-oxidizing bacteria secrete metabolites that markedly impede abiotic iron oxidation. PNAS NEXUS 2023; 2:pgad421. [PMID: 38111821 PMCID: PMC10727123 DOI: 10.1093/pnasnexus/pgad421] [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: 08/24/2023] [Accepted: 11/29/2023] [Indexed: 12/20/2023]
Abstract
Iron is one of the Earth's most abundant elements and is required for essentially all forms of life. Yet, iron's reactivity with oxygen and poor solubility in its oxidized form (Fe3+) mean that it is often a limiting nutrient in oxic, near-neutral pH environments like Earth's ocean. In addition to being a vital nutrient, there is a diversity of aerobic organisms that oxidize ferrous iron (Fe2+) to harness energy for growth and biosynthesis. Accordingly, these organisms rely on access to co-existing Fe2+ and O2 to survive. It is generally presumed that such aerobic iron-oxidizing bacteria (FeOB) are relegated to low-oxygen regimes where abiotic iron oxidation rates are slower, yet some FeOB live in higher oxygen environments where they cannot rely on lower oxygen concentrations to overcome abiotic competition. We hypothesized that FeOB chemically alter their environment to limit abiotic interactions between Fe2+ and O2. To test this, we incubated the secreted metabolites (collectively known as the exometabolome) of the deep-sea iron- and hydrogen-oxidizing bacterium Ghiorsea bivora TAG-1 with ferrous iron and oxygen. We found that this FeOB's iron-oxidizing exometabolome markedly impedes the abiotic oxidation of ferrous iron, increasing the half-life of Fe2+ 100-fold from ∼3 to ∼335 days in the presence of O2, while the exometabolome of TAG-1 grown on hydrogen had no effect. Moreover, the few precipitates that formed in the presence of TAG-1's iron-oxidizing exometabolome were poorly crystalline, compared with the abundant iron particles that mineralized in the absence of abiotic controls. We offer an initial exploration of TAG-1's iron-oxidizing exometabolome and discuss potential key contributors to this process. Overall, our findings demonstrate that the exometabolome as a whole leads to a sustained accumulation of ferrous iron in the presence of oxygen, consequently altering the redox equilibrium. This previously unknown adaptation likely enables these microorganisms to persist in an iron-oxidizing and iron-precipitating world and could have impacts on the bioavailability of iron to FeOB and other life in iron-limiting environments.
Collapse
Affiliation(s)
- Isabel R Baker
- Department of Organismic and Evolutionary Biology, Harvard University, Cambridge, MA 02138, USA
| | - Sarick L Matzen
- Department of Soil, Water, and Climate, University of Minnesota Twin Cities, Saint Paul, MN 55108, USA
| | - Christopher J Schuler
- Department of Earth and Environmental Sciences, University of Minnesota Twin Cities, Saint Paul, MN 55108, USA
| | - Brandy M Toner
- Department of Soil, Water, and Climate, University of Minnesota Twin Cities, Saint Paul, MN 55108, USA
- Department of Earth and Environmental Sciences, University of Minnesota Twin Cities, Saint Paul, MN 55108, USA
| | - Peter R Girguis
- Department of Organismic and Evolutionary Biology, Harvard University, Cambridge, MA 02138, USA
| |
Collapse
|
38
|
Ortiz S, Niks D, Wiley S, Lubner CE, Hille R. Rapid-reaction kinetics of the bifurcating NAD +-dependent NADPH:ferredoxin oxidoreductase NfnI from Pyrococcus furiosus. J Biol Chem 2023; 299:105403. [PMID: 38229399 PMCID: PMC10724689 DOI: 10.1016/j.jbc.2023.105403] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Revised: 10/02/2023] [Accepted: 10/22/2023] [Indexed: 01/18/2024] Open
Abstract
We have investigated the kinetics of NAD+-dependent NADPH:ferredoxin oxidoreductase (NfnI), a bifurcating transhydrogenase that takes two electron pairs from NADPH to reduce two ferredoxins and one NAD+ through successive bifurcation events. NADPH reduction takes place at the bifurcating FAD of NfnI's large subunit, with high-potential electrons transferred to the [2Fe-2S] cluster and S-FADH of the small subunit, ultimately on to NAD+; low-potential electrons are transferred to two [4Fe-4S] clusters of the large subunit and on to ferredoxin. Reduction of NfnI by NADPH goes to completion only at higher pH, with a limiting kred of 36 ± 1.6 s-1 and apparent KdNADPH of 5 ± 1.2 μM. Reduction of one of the [4Fe-4S] clusters of NfnI occurs within a second, indicating that in the absence of NAD+, the system can bifurcate and generate low-potential electrons without NAD+. When enzyme is reduced by NADPH in the absence of NAD+ but the presence of ferredoxin, up to three equivalents of ferredoxin become reduced, although the reaction is considerably slower than seen during steady-state turnover. Bifurcation appears to be limited by transfer of the first, high-potential electron into the high-potential pathway. Ferredoxin reduction without NAD+ demonstrates that electron bifurcation is an intrinsic property of the bifurcating FAD and is not dependent on the simultaneous presence of NAD+ and ferredoxin. The tight coupling between NAD+ and ferredoxin reduction observed under multiple-turnover conditions is instead simply due to the need to remove reducing equivalents from the high-potential electron pathway under multiple-turnover conditions.
Collapse
Affiliation(s)
- Steve Ortiz
- Department of Biochemistry and the Biophysics Graduate Program, University of California, Riverside, USA
| | - Dimitri Niks
- Department of Biochemistry and the Biophysics Graduate Program, University of California, Riverside, USA
| | - Seth Wiley
- Biosciences Center, National Renewable Energy Laboratory, Golden, Colorado, USA
| | - Carolyn E Lubner
- Biosciences Center, National Renewable Energy Laboratory, Golden, Colorado, USA.
| | - Russ Hille
- Department of Biochemistry and the Biophysics Graduate Program, University of California, Riverside, USA.
| |
Collapse
|
39
|
Hati D, Brault A, Gupta M, Fletcher K, Jacques JF, Labbé S, Outten CE. Iron homeostasis proteins Grx4 and Fra2 control activity of the Schizosaccharomyces pombe iron repressor Fep1 by facilitating [2Fe-2S] cluster removal. J Biol Chem 2023; 299:105419. [PMID: 37923140 PMCID: PMC10704371 DOI: 10.1016/j.jbc.2023.105419] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Revised: 10/12/2023] [Accepted: 10/26/2023] [Indexed: 11/07/2023] Open
Abstract
The Bol2 homolog Fra2 and monothiol glutaredoxin Grx4 together play essential roles in regulating iron homeostasis in Schizosaccharomyces pombe. In vivo studies indicate that Grx4 and Fra2 act as coinhibitory partners that inactivate the transcriptional repressor Fep1 in response to iron deficiency. In Saccharomyces cerevisiae, Bol2 is known to form a [2Fe-2S]-bridged heterodimer with the monothiol Grxs Grx3 and Grx4, with the cluster ligands provided by conserved residues in Grx3/4 and Bol2 as well as GSH. In this study, we characterized this analogous [2Fe-2S]-bridged Grx4-Fra2 complex in S. pombe by identifying the specific residues in Fra2 that act as ligands for the Fe-S cluster and are required to regulate Fep1 activity. We present spectroscopic and biochemical evidence confirming the formation of a [2Fe-2S]-bridged Grx4-Fra2 heterodimer with His66 and Cys29 from Fra2 serving as Fe-S cluster ligands in S. pombe. In vivo transcription and growth assays confirm that both His66 and Cys29 are required to fully mediate the response of Fep1 to low iron conditions. Furthermore, we analyzed the interaction between Fep1 and Grx4-Fra2 using CD spectroscopy to monitor changes in Fe-S cluster coordination chemistry. These experiments demonstrate unidirectional [2Fe-2S] cluster transfer from Fep1 to Grx4-Fra2 in the presence of GSH, revealing the Fe-S cluster dependent mechanism of Fep1 inactivation mediated by Grx4 and Fra2 in response to iron deficiency.
Collapse
Affiliation(s)
- Debolina Hati
- Department of Chemistry and Biochemistry, University of South Carolina, Columbia, South Carolina, USA
| | - Ariane Brault
- Département de Biochimie et de Génomique Fonctionnelle, Faculté de médecine et des sciences de la santé, Université de Sherbrooke, Sherbrooke, Quebec, Canada
| | - Malini Gupta
- Department of Chemistry and Biochemistry, University of South Carolina, Columbia, South Carolina, USA
| | - Kylie Fletcher
- Department of Chemistry and Biochemistry, University of South Carolina, Columbia, South Carolina, USA
| | - Jean-François Jacques
- Département de Biochimie et de Génomique Fonctionnelle, Faculté de médecine et des sciences de la santé, Université de Sherbrooke, Sherbrooke, Quebec, Canada
| | - Simon Labbé
- Département de Biochimie et de Génomique Fonctionnelle, Faculté de médecine et des sciences de la santé, Université de Sherbrooke, Sherbrooke, Quebec, Canada
| | - Caryn E Outten
- Department of Chemistry and Biochemistry, University of South Carolina, Columbia, South Carolina, USA.
| |
Collapse
|
40
|
Liu Z, Huang B, Liao X, Wang L, Yang X, Hu X. Salicylic acid doped silica nanoparticles as a fluorescent nanosensor for the detection of Fe 3+ in aqueous solution. ANALYTICAL METHODS : ADVANCING METHODS AND APPLICATIONS 2023; 15:6404-6411. [PMID: 37861085 DOI: 10.1039/d3ay01464a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/21/2023]
Abstract
A novel organic-inorganic hybrid nanosensor (SASP) was prepared by a one-step sol-gel method and characterized by scanning electron microscopy, transmission electron microscopy, Fourier transform infrared spectroscopy, thermogravimetric analysis, N2 adsorption-desorption, fluorescence spectroscopy, etc. The nanosensor showed almost 3-fold fluorescence emission quenching upon excitation with a 293 nm wavelength in the presence of 20 μM Fe3+ ions. The presence of 18 other metal ions had no observable effect on the sensitivity and selectivity of the nanosensor. A fluorescence analysis method based on the SASP for the selective detection of Fe3+ was established under optimal conditions. The results showed that there was a linear relationship between the log luminescence value and the concentration of Fe3+ over the range of 2.0 × 10-7-9.0 × 10-5 mol L-1 with a detection limit (3σ) of 2.5 × 10-8 mol L-1. Furthermore, the proposed method was successfully applied for the determination of trace Fe3+ in fetal bovine serum without the interference of other molecules and ions. Good recovery (96.5-104.5%) and a relative standard deviation of less than 8.6% were obtained from serum samples spiked with four levels of Fe3+. Additionally, the nanosensor showed a good reversibility; the fluorescence could be switched "off" and "on" in two ways, by adjusting the pH of the solution and adding metal chelating agent EDTA.
Collapse
Affiliation(s)
- Zhongyong Liu
- Guangzhou Key Laboratory of Analytical Chemistry for Biomedicine, GDMPA Key Laboratory for Process Control and Quality Evaluation of Chiral Pharmaceuticals, School of Chemistry, South China Normal University, Guangzhou, P. R. China.
| | - Bomao Huang
- Guangzhou Key Laboratory of Analytical Chemistry for Biomedicine, GDMPA Key Laboratory for Process Control and Quality Evaluation of Chiral Pharmaceuticals, School of Chemistry, South China Normal University, Guangzhou, P. R. China.
| | - Xianglin Liao
- Guangzhou Key Laboratory of Analytical Chemistry for Biomedicine, GDMPA Key Laboratory for Process Control and Quality Evaluation of Chiral Pharmaceuticals, School of Chemistry, South China Normal University, Guangzhou, P. R. China.
| | - Li Wang
- Guangzhou Key Laboratory of Analytical Chemistry for Biomedicine, GDMPA Key Laboratory for Process Control and Quality Evaluation of Chiral Pharmaceuticals, School of Chemistry, South China Normal University, Guangzhou, P. R. China.
| | - Xixiang Yang
- Guangzhou Key Laboratory of Analytical Chemistry for Biomedicine, GDMPA Key Laboratory for Process Control and Quality Evaluation of Chiral Pharmaceuticals, School of Chemistry, South China Normal University, Guangzhou, P. R. China.
| | - Xiaogang Hu
- Guangzhou Key Laboratory of Analytical Chemistry for Biomedicine, GDMPA Key Laboratory for Process Control and Quality Evaluation of Chiral Pharmaceuticals, School of Chemistry, South China Normal University, Guangzhou, P. R. China.
| |
Collapse
|
41
|
Angelini C, Durand CM, Fergelot P, Deforges J, Vital A, Menegon P, Sarrazin E, Bellance R, Mathis S, Gonzalez V, Renaud M, Frismand S, Schmitt E, Rouanet M, Burglen L, Chabrol B, Desnous B, Arveiler B, Stevanin G, Coupry I, Goizet C. Autosomal Dominant MPAN: Mosaicism Expands the Clinical Spectrum to Atypical Late-Onset Phenotypes. Mov Disord 2023; 38:2103-2115. [PMID: 37605305 DOI: 10.1002/mds.29576] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Revised: 05/31/2023] [Accepted: 07/24/2023] [Indexed: 08/23/2023] Open
Abstract
BACKGROUND Mitochondrial membrane protein-associated neurodegeneration (MPAN) is caused by mutations in the C19orf12 gene. MPAN typically appears in the first two decades of life and presents with progressive dystonia-parkinsonism, lower motor neuron signs, optic atrophy, and abnormal iron deposits predominantly in the basal ganglia. MPAN, initially considered as a strictly autosomal recessive disease (AR), turned out to be also dominantly inherited (AD). OBJECTIVES Our aim was to better characterize the clinical, molecular, and functional spectra associated with such dominant pathogenic heterozygous C19orf12 variants. METHODS We collected clinical, imaging, and molecular information of eight individuals from four AD-MPAN families and obtained brain neuropathology results for one. Functional studies, focused on energy and iron metabolism, were conducted on fibroblasts from AD-MPAN patients, AR-MPAN patients, and controls. RESULTS We identified four heterozygous C19orf12 variants in eight AD-MPAN patients. Two of them carrying the familial variant in mosaic displayed an atypical late-onset phenotype. Fibroblasts from AD-MPAN showed more severe alterations of iron storage metabolism and autophagy compared to AR-MPAN cells. CONCLUSION Our data add strong evidence of the realness of AD-MPAN with identification of novel monoallelic C19orf12 variants, including at the mosaic state. This has implications in diagnosis procedures. We also expand the phenotypic spectrum of MPAN to late onset atypical presentations. Finally, we demonstrate for the first time more drastic abnormalities of iron metabolism and autophagy in AD-MPAN than in AR-MPAN. © 2023 The Authors. Movement Disorders published by Wiley Periodicals LLC on behalf of International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
- Chloé Angelini
- Service de Génétique Médicale, Hôpital Pellegrin, CHU Bordeaux, Bordeaux, France
- Centre de Référence Maladies Rares «Neurogénétique», Service de Génétique Médicale, CHU Bordeaux, Bordeaux, France
- University of Bordeaux, CNRS, INCIA, UMR 5287, NRGen Team, Bordeaux, France
| | - Christelle Marie Durand
- Service de Génétique Médicale, Hôpital Pellegrin, CHU Bordeaux, Bordeaux, France
- Centre de Référence Maladies Rares «Neurogénétique», Service de Génétique Médicale, CHU Bordeaux, Bordeaux, France
- University of Bordeaux, CNRS, INCIA, UMR 5287, NRGen Team, Bordeaux, France
- MRGM, University of Bordeaux, INSERM U1211, Bordeaux, France
| | - Patricia Fergelot
- Service de Génétique Médicale, Hôpital Pellegrin, CHU Bordeaux, Bordeaux, France
- MRGM, University of Bordeaux, INSERM U1211, Bordeaux, France
| | - Julie Deforges
- Service de Génétique Médicale, Hôpital Pellegrin, CHU Bordeaux, Bordeaux, France
| | - Anne Vital
- Service d'Anatomie Pathologique, Hôpital Pellegrin, CHU Bordeaux, Bordeaux, France
| | - Patrice Menegon
- Service de Neuroradiologie, Hôpital Pellegrin, CHU Bordeaux, Bordeaux, France
| | - Elizabeth Sarrazin
- Centre de Référence Maladies Rares Neuromusculaires (AOC), Hôpital Pierre Zobda Quitman, CHU Martinique, Fort de France, Martinique
| | - Rémi Bellance
- Centre de Référence Maladies Rares Neuromusculaires (AOC), Hôpital Pierre Zobda Quitman, CHU Martinique, Fort de France, Martinique
| | - Stéphane Mathis
- Service de Neurologie (Unité Nerf-Muscle), Centre de Référence Maladies Rares, Neuromusculaires (AOC), Centre SLA, Hôpital Pellegrin, CHU Bordeaux, Bordeaux, France
| | - Victoria Gonzalez
- Service de neurologie, Hôpital Gui de Chauliac, CHU Montpellier, Montpellier, France
| | - Mathilde Renaud
- Service de Neurologie, CHRU Nancy, Nancy, France
- Service de Génétique Clinique, CHRU Nancy, Nancy, France
- NGERE, INSERM U1256, Faculté de Médecine, Université de Lorraine, Nancy, France
| | | | - Emmanuelle Schmitt
- Service de Neuroradiologie Diagnostique et Thérapeutique, CHRU Nancy, Nancy, France
| | - Marie Rouanet
- Service d'explorations Fonctionnelles du Système Nerveux, Hôpital Pellegrin, CHU Bordeaux, Bordeaux, France
| | - Lydie Burglen
- Laboratoire de Neurogénétique Pédiatrique, Département de Génétique, Hôpital Trousseau, APHP.Sorbonne Université, Paris, France
| | - Brigitte Chabrol
- Service de Neuropédiatrie, Hôpital Timone enfants, APHM, Marseille, France
| | - Béatrice Desnous
- Service de Neuropédiatrie, Hôpital Timone enfants, APHM, Marseille, France
| | - Benoît Arveiler
- Service de Génétique Médicale, Hôpital Pellegrin, CHU Bordeaux, Bordeaux, France
- MRGM, University of Bordeaux, INSERM U1211, Bordeaux, France
| | - Giovanni Stevanin
- University of Bordeaux, CNRS, INCIA, UMR 5287, NRGen Team, Bordeaux, France
- EPHE, CNRS, INCIA, UMR 5287, PSL Research University, Paris, France
| | - Isabelle Coupry
- University of Bordeaux, CNRS, INCIA, UMR 5287, NRGen Team, Bordeaux, France
- MRGM, University of Bordeaux, INSERM U1211, Bordeaux, France
| | - Cyril Goizet
- Service de Génétique Médicale, Hôpital Pellegrin, CHU Bordeaux, Bordeaux, France
- Centre de Référence Maladies Rares «Neurogénétique», Service de Génétique Médicale, CHU Bordeaux, Bordeaux, France
- University of Bordeaux, CNRS, INCIA, UMR 5287, NRGen Team, Bordeaux, France
- MRGM, University of Bordeaux, INSERM U1211, Bordeaux, France
| |
Collapse
|
42
|
Sun K, Li Y, Gai Y, Wang J, Jian Y, Liu X, Wu L, Shim WB, Lee YW, Ma Z, Haas H, Yin Y. HapX-mediated H2B deub1 and SreA-mediated H2A.Z deposition coordinate in fungal iron resistance. Nucleic Acids Res 2023; 51:10238-10260. [PMID: 37650633 PMCID: PMC10602907 DOI: 10.1093/nar/gkad708] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2022] [Revised: 07/26/2023] [Accepted: 08/15/2023] [Indexed: 09/01/2023] Open
Abstract
Plant pathogens are challenged by host-derived iron starvation or excess during infection, but the mechanism through which pathogens counteract iron stress is unclear. Here, we found that Fusarium graminearum encounters iron excess during the colonization of wheat heads. Deletion of heme activator protein X (FgHapX), siderophore transcription factor A (FgSreA) or both attenuated virulence. Further, we found that FgHapX activates iron storage under iron excess by promoting histone H2B deubiquitination (H2B deub1) at the promoter of the responsible gene. Meanwhile, FgSreA is shown to inhibit genes mediating iron acquisition during iron excess by facilitating the deposition of histone variant H2A.Z and histone 3 lysine 27 trimethylation (H3K27 me3) at the first nucleosome after the transcription start site. In addition, the monothiol glutaredoxin FgGrx4 is responsible for iron sensing and control of the transcriptional activity of FgHapX and FgSreA via modulation of their enrichment at target genes and recruitment of epigenetic regulators, respectively. Taken together, our findings elucidated the molecular mechanisms for adaptation to iron excess mediated by FgHapX and FgSreA during infection in F. graminearum and provide novel insights into regulation of iron homeostasis at the chromatin level in eukaryotes.
Collapse
Affiliation(s)
- Kewei Sun
- State Key Laboratory of Rice Biology, the Key Laboratory of Molecular Biology of Crop Pathogens and Insects, Institute of Biotechnology, Zhejiang University, Hangzhou, China
| | - Yiqing Li
- State Key Laboratory of Rice Biology, the Key Laboratory of Molecular Biology of Crop Pathogens and Insects, Institute of Biotechnology, Zhejiang University, Hangzhou, China
| | - Yunpeng Gai
- School of Grassland Science, Beijing Forestry University, Beijing, China
| | - Jingrui Wang
- State Key Laboratory of Rice Biology, the Key Laboratory of Molecular Biology of Crop Pathogens and Insects, Institute of Biotechnology, Zhejiang University, Hangzhou, China
| | - Yunqing Jian
- State Key Laboratory of Rice Biology, the Key Laboratory of Molecular Biology of Crop Pathogens and Insects, Institute of Biotechnology, Zhejiang University, Hangzhou, China
| | - Xin Liu
- Institute of Food Safety and Nutrition, Jiangsu Academy of Agricultural Sciences, Nanjing, China
| | - Liang Wu
- Institute of Crop Science, Zhejiang University, Hangzhou, China
| | - Won-Bo Shim
- Department of Plant Pathology and Microbiology, Texas A&M University, College Station, USA
| | - Yin-Won Lee
- Department of Agricultural Biotechnology, Seoul National University, Seoul, Korea
| | - Zhonghua Ma
- State Key Laboratory of Rice Biology, the Key Laboratory of Molecular Biology of Crop Pathogens and Insects, Institute of Biotechnology, Zhejiang University, Hangzhou, China
| | - Hubertus Haas
- Instiute of Molecular Biology, Biocenter, Medical University Innsbruck, Innsbruck A-6020, Austria
| | - Yanni Yin
- State Key Laboratory of Rice Biology, the Key Laboratory of Molecular Biology of Crop Pathogens and Insects, Institute of Biotechnology, Zhejiang University, Hangzhou, China
| |
Collapse
|
43
|
Breborowicz A. Iron increases the profibrotic properties of the senescent peritoneal mesothelial cells. Int J Artif Organs 2023; 46:473-480. [PMID: 37564017 DOI: 10.1177/03913988231192123] [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: 08/12/2023]
Abstract
BACKGROUND Treatment of anemia in peritoneal dialysis patients often requires intravenous iron supplementation. Iron diffuses into the peritoneal cavity and is injurious to the peritoneum. We studied how intermittent exposure to iron changes the properties of the senescent peritoneal mesothelial cells (MC). METHODS Replicative senescence was induced in MC in control medium (Con) or in control medium with intermittent exposure to iron isomaltoside 15 µg/dL (Con-IIS). After 10 passages properties of MC from both groups were compared to MC not exposed to replicative senescence. RESULTS In senescent MC population doubling time was elongated, intracellular generation of free radicals and staining for β-galactosidase was stronger than in MC not exposed to replicative senescence. All these effects were stronger in MC intermittently exposed to IIS. In these cells intracellular iron content was also higher. Also expression of genes p21 and p53 was stronger in MC intermittently treated with IIS. In senescent cells higher release and expression of IL6 and TGFβ1 was observed and that effect was stronger in MC treated with iron. Senescent MC had reduced fibrinolytic activity, what may predispose to the peritoneal fibrosis. Synthesis of collagen was higher in senescent cells, more in MC treated with iron. CONCLUSION MC aging results in change of their genotype and phenotype which lead to their profibrotic effect. Exposure to iron enhances these changes.
Collapse
Affiliation(s)
- Andrzej Breborowicz
- Department of Pathophysiology, Poznan University of Medical Sciences, Poznan, Greater Poland Voivodeship, Poland
- Department of Histology, Collegium Medicum, Zielona Gora University, Zielona Gora, Poland
| |
Collapse
|
44
|
Eckenberger E, Raczka T, Neuhuber W, Distel LVR, Klein S. Acriflavine-Functionalized Silica@Manganese Ferrite Nanostructures for Synergistic Radiation and Hypoxia Therapies. ACS APPLIED BIO MATERIALS 2023; 6:3089-3102. [PMID: 37433114 DOI: 10.1021/acsabm.2c01021] [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: 07/13/2023]
Abstract
Mesoporous and nonmesoporous SiO2@MnFe2O4 nanostructures were loaded with the hypoxia-inducible factor-1 inhibitor acriflavine for combined radiation and hypoxia therapies. The X-ray irradiation of the drug-loaded nanostructures not only triggered the release of the acriflavine inside the cells but also initiated an energy transfer from the nanostructures to surface-adsorbed oxygen to generate singlet oxygen. While the drug-loaded mesoporous nanostructures showed an initial drug release before the irradiation, the drug was primarily released upon X-ray radiation in the case of the nonmesoporous nanostructures. However, the drug loading capacity was less efficient for the nonmesoporous nanostructures. Both drug-loaded nanostructures proved to be very efficient in irradiated MCF-7 multicellular tumor spheroids. The damage of these nanostructures toward the nontumorigenic MCF-10A multicellular spheroids was very limited because of the small number of nanostructures that entered the MCF-10A spheroids, while similar concentrations of acriflavine without nanostructures were toxic for the MCF-10A spheroids.
Collapse
Affiliation(s)
- Elisabeth Eckenberger
- Department of Chemistry and Pharmacy, Physical Chemistry I and ICMM, Friedrich-Alexander University of Erlangen-Nuremberg, Egerlandstr.3, D-91058 Erlangen, Germany
| | - Theodor Raczka
- Department of Chemistry and Pharmacy, Physical Chemistry I and ICMM, Friedrich-Alexander University of Erlangen-Nuremberg, Egerlandstr.3, D-91058 Erlangen, Germany
| | - Winfried Neuhuber
- Institute of Anatomy and Cell Biology, Chair of Anatomy I, Friedrich-Alexander University Erlangen-Nuremberg, Krankenhausstr. 9, D-91054 Erlangen, Germany
| | - Luitpold V R Distel
- Department of Radiation Oncology, Friedrich-Alexander University of Erlangen-Nuremberg, Universitätsstr. 27, D-91054 Erlangen, Germany
| | - Stefanie Klein
- Department of Chemistry and Pharmacy, Physical Chemistry I and ICMM, Friedrich-Alexander University of Erlangen-Nuremberg, Egerlandstr.3, D-91058 Erlangen, Germany
| |
Collapse
|
45
|
Ger TR, Wu PS, Wang WJ, Chen CA, Abu PAR, Chen SL. Development of a Microfluidic Chip System with Giant Magnetoresistance Sensor for High-Sensitivity Detection of Magnetic Nanoparticles in Biomedical Applications. BIOSENSORS 2023; 13:807. [PMID: 37622894 PMCID: PMC10452397 DOI: 10.3390/bios13080807] [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: 07/10/2023] [Revised: 08/05/2023] [Accepted: 08/06/2023] [Indexed: 08/26/2023]
Abstract
Magnetic nanoparticles (MNPs) have been widely utilized in the biomedical field for numerous years, offering several advantages such as exceptional biocompatibility and diverse applications in biology. However, the existing methods for quantifying magnetic labeled sample assays are scarce. This research presents a novel approach by developing a microfluidic chip system embedded with a giant magnetoresistance (GMR) sensor. The system successfully detects low concentrations of MNPs with magnetic particle velocities of 20 mm/s. The stray field generated by the magnetic subject flowing through the microchannel above the GMR sensor causes variations in the signals. The sensor's output signals are appropriately amplified, filtered, and processed to provide valuable indications. The integration of the GMR microfluidic chip system demonstrates notable attributes, including affordability, speed, and user-friendly operation. Moreover, it exhibits a high detection sensitivity of 10 μg/μL for MNPs, achieved through optimizing the vertical magnetic field to 100 Oe and the horizontal magnetic field to 2 Oe. Additionally, the study examines magnetic labeled RAW264.7 cells. This quantitative detection of magnetic nanoparticles can have applications in DNA concentration detection, protein concentration detection, and other promising areas of research.
Collapse
Affiliation(s)
- Tzong-Rong Ger
- Department of Biomedical Engineering, Chung Yuan Christian University, Chung-Li 320314, Taiwan
| | - Pei-Sheng Wu
- Department of Biomedical Engineering, Chung Yuan Christian University, Chung-Li 320314, Taiwan
| | - Wei-Jie Wang
- Department of Biomedical Engineering, Chung Yuan Christian University, Chung-Li 320314, Taiwan
- Division of Nephrology, Department of Internal Medicine, Taoyuan General Hospital, Ministry of Health and Welfare, Taoyuan 115204, Taiwan
| | - Chiung-An Chen
- Department of Electrical Engineering, Ming Chi University of Technology, New Taipei City 243303, Taiwan
| | - Patricia Angela R. Abu
- Department of Information Systems and Computer Science, Ateneo de Manila University, Quezon City 1108, Philippines
| | - Shih-Lun Chen
- Department of Electronic Engineering, Chung Yuan Christian University, Taoyuan City 320314, Taiwan;
| |
Collapse
|
46
|
Banerjee S, Datta R. Localized Leishmania major infection disrupts systemic iron homeostasis that can be controlled by oral iron supplementation. J Biol Chem 2023; 299:105064. [PMID: 37468101 PMCID: PMC10448173 DOI: 10.1016/j.jbc.2023.105064] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2023] [Revised: 07/08/2023] [Accepted: 07/11/2023] [Indexed: 07/21/2023] Open
Abstract
Leishmania parasites are heavily dependent on efficient iron acquisition from a tightly regulated host iron pool for survival and virulence. Prior studies uncovered multiple strategies adopted by the parasite to hijack the iron-regulatory network of macrophages. Despite these extensive studies with infected macrophages, there is limited knowledge of the effect of Leishmania infection on systemic iron homeostasis. This issue is particularly relevant for Leishmania major, which causes localized skin infection with minimal lymphatic spread. We show for the first time that L. major infection in the mouse footpad induced influx of iron at the site of infection through blood with simultaneous upregulation of transferrin receptor 1 and downregulation of phagolysosomal iron exporter Nramp1 expression in the footpad tissue. Interestingly, localized L. major infection had far-reaching effects beyond the infection site triggering anemia-like symptoms. This was evident from depleted physiological iron stores from the liver and bone marrow as well as reduced hemoglobin levels and deformed erythrocytes. The infected mice also developed splenomegaly with signs of splenic stress erythropoiesis as indicated by upregulation of several erythroid-related genes. These observations prompted us to provide oral iron supplementations to the L. major-infected mice, which resulted in a drastic reduction of the parasite load and restoration of iron homeostasis.
Collapse
Affiliation(s)
- Sourav Banerjee
- Department of Biological Sciences, Indian Institute of Science Education and Research (IISER) Kolkata, Mohanpur, West Bengal, India
| | - Rupak Datta
- Department of Biological Sciences, Indian Institute of Science Education and Research (IISER) Kolkata, Mohanpur, West Bengal, India.
| |
Collapse
|
47
|
Pap R, Pandur E, Jánosa G, Sipos K, Fritz FR, Nagy T, Agócs A, Deli J. Protective Effects of 3'-Epilutein and 3'-Oxolutein against Glutamate-Induced Neuronal Damage. Int J Mol Sci 2023; 24:12008. [PMID: 37569384 PMCID: PMC10418699 DOI: 10.3390/ijms241512008] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Revised: 07/23/2023] [Accepted: 07/26/2023] [Indexed: 08/13/2023] Open
Abstract
Dietary lutein can be naturally metabolized to 3'-epilutein and 3'-oxolutein in the human body. The epimerization of lutein can happen in acidic pH, and through cooking, 3'-epilutein can be the product of the direct oxidation of lutein in the retina, which is also present in human serum. The 3'-oxolutein is the main oxidation product of lutein. Thus, the allylic oxidation of dietary lutein can result in the formation of 3'-oxolutein, which may undergo reduction either to revert to dietary lutein or epimerize to form 3'-epilutein. We focused on the effects of 3'-epilutein and 3'-oxolutein itself and on glutamate-induced neurotoxicity on SH-SY5Y human neuroblastoma cells to identify the possible alterations in oxidative stress, inflammation, antioxidant capacity, and iron metabolism that affect neurological function. ROS measurements were performed in the differently treated cells. The inflammatory state of cells was followed by TNFα, IL-6, and IL-8 cytokine ELISA measurements. The antioxidant status of the cells was determined by the total antioxidant capacity kit assay. The alterations of genes related to ferroptosis and lipid peroxidation were followed by gene expression measurements; then, thiol measurements were performed. Lutein metabolites 3'-epilutein and 3'-oxolutein differently modulated the effect of glutamate on ROS, inflammation, ferroptosis-related iron metabolism, and lipid peroxidation in SH-SY5Y cells. Our results revealed the antioxidant and anti-inflammatory features of 3'-epilutein and 3'-oxolutein as possible protective agents against glutamate-induced oxidative stress in SH-SY5Y cells, with greater efficacy in the case of 3'-epilutein.
Collapse
Affiliation(s)
- Ramóna Pap
- Department of Pharmaceutical Biology, Faculty of Pharmacy, University of Pécs, Rókus u. 2, H-7624 Pécs, Hungary; (R.P.); (E.P.); (G.J.); (K.S.); (F.R.F.)
| | - Edina Pandur
- Department of Pharmaceutical Biology, Faculty of Pharmacy, University of Pécs, Rókus u. 2, H-7624 Pécs, Hungary; (R.P.); (E.P.); (G.J.); (K.S.); (F.R.F.)
| | - Gergely Jánosa
- Department of Pharmaceutical Biology, Faculty of Pharmacy, University of Pécs, Rókus u. 2, H-7624 Pécs, Hungary; (R.P.); (E.P.); (G.J.); (K.S.); (F.R.F.)
| | - Katalin Sipos
- Department of Pharmaceutical Biology, Faculty of Pharmacy, University of Pécs, Rókus u. 2, H-7624 Pécs, Hungary; (R.P.); (E.P.); (G.J.); (K.S.); (F.R.F.)
| | - Ferenc Rómeó Fritz
- Department of Pharmaceutical Biology, Faculty of Pharmacy, University of Pécs, Rókus u. 2, H-7624 Pécs, Hungary; (R.P.); (E.P.); (G.J.); (K.S.); (F.R.F.)
| | - Tamás Nagy
- Department of Laboratory Medicine, Faculty of Medical Sciences, University of Pécs, Ifjúság út 13, H-7624 Pécs, Hungary;
| | - Attila Agócs
- Department of Biochemistry and Medical Chemistry, Medical School, University of Pécs, Szigeti út 12, H-7624 Pécs, Hungary;
| | - József Deli
- Department of Biochemistry and Medical Chemistry, Medical School, University of Pécs, Szigeti út 12, H-7624 Pécs, Hungary;
- Department of Pharmacognosy, Faculty of Pharmacy, University of Pécs, Rókus u. 2, H-7624 Pécs, Hungary
| |
Collapse
|
48
|
Bi G, Liang J, Shan G, Bian Y, Chen Z, Huang Y, Lu T, Li M, Besskaya V, Zhao M, Fan H, Wang Q, Gan B, Zhan C. Retinol Saturase Mediates Retinoid Metabolism to Impair a Ferroptosis Defense System in Cancer Cells. Cancer Res 2023; 83:2387-2404. [PMID: 37184371 DOI: 10.1158/0008-5472.can-22-3977] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Revised: 03/22/2023] [Accepted: 05/10/2023] [Indexed: 05/16/2023]
Abstract
Ferroptosis is an iron-dependent form of regulated cell death induced by the lethal overload of lipid peroxides in cellular membranes. In recent years, modulating ferroptosis has gained attention as a potential therapeutic approach for tumor suppression. In the current study, retinol saturase (RETSAT) was identified as a significant ferroptosis mediator using a publicly accessible CRISPR/Cas9 screening dataset. RETSAT depletion protected tumor cells from lipid peroxidation and subsequent cell death triggered by various ferroptosis inducers. Furthermore, exogenous supplementation with retinoids, including retinol (the substrate of RETSAT) and its derivatives retinal and retinoic acid, also suppressed ferroptosis, whereas the product of RETSAT, 13, 14-dihydroretinol, failed to do so. As effective radical-trapping antioxidant, retinoids protected the lipid membrane from autoxidation and subsequent fragmentation, thus terminating the cascade of ferroptosis. Pseudotargeted lipidomic analysis identified an association between retinoid regulation of ferroptosis and lipid metabolism. Retinoic acid, but not 13, 14-dihydroretinoic acid, interacted with its nuclear receptor and activated transcription of stearoyl-CoA desaturase, which introduces the first double bond into saturated fatty acid and thus catalyzes the generation of monounsaturated fatty acid, a known ferroptosis suppressor. Therefore, RETSAT promotes ferroptosis by transforming retinol to 13, 14-dihydroretinol, thereby turning a strong anti-ferroptosis regulator into a relatively weak one. SIGNIFICANCE Retinoids have ferroptosis-protective properties and can be metabolized by RETSAT to promote ferroptosis, suggesting the possibility of targeting retinoid metabolism in cancer as a treatment strategy to trigger ferroptosis.
Collapse
Affiliation(s)
- Guoshu Bi
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, Shanghai, P.R. China
| | - Jiaqi Liang
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, Shanghai, P.R. China
| | - Guangyao Shan
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, Shanghai, P.R. China
| | - Yunyi Bian
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, Shanghai, P.R. China
| | - Zhencong Chen
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, Shanghai, P.R. China
| | - Yiwei Huang
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, Shanghai, P.R. China
| | - Tao Lu
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, Shanghai, P.R. China
| | - Ming Li
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, Shanghai, P.R. China
| | - Valeria Besskaya
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, Shanghai, P.R. China
| | - Mengnan Zhao
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, Shanghai, P.R. China
| | - Hong Fan
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, Shanghai, P.R. China
| | - Qun Wang
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, Shanghai, P.R. China
| | - Boyi Gan
- Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Cheng Zhan
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, Shanghai, P.R. China
| |
Collapse
|
49
|
Xiao G, Li H, Zhao M, Zhou B. Assessing metal ion transporting activity of ZIPs: Intracellular zinc and iron detection. Methods Enzymol 2023; 687:157-184. [PMID: 37666631 DOI: 10.1016/bs.mie.2023.05.011] [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: 09/06/2023]
Abstract
Zrt/Irt-like proteins (ZIPs or SLC39A) are a large family of metal ion transporters mainly responsible for zinc uptake. Some ZIPs have been shown to specifically transport zinc, whereas others have broader substrate specificity in divalent metal ion trafficking, notably those of zinc and iron ions. Measuring intracellular zinc and iron levels helps assess their molecular and physiological activities. This chapter presents step-by-step methods for evaluating intracellular metal ion concentrations, including direct measurement using inductively coupled plasma-mass spectrometry (ICP-MS), chemical staining, fluorescent probes, and indirect reporter assays such as activity analysis of enzymes whose activities are dependent on metal ion availability.
Collapse
Affiliation(s)
- Guiran Xiao
- School of Food and Biological Engineering, Hefei University of Technology, Hefei, Anhui, P.R. China
| | - Huihui Li
- Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, P.R. China
| | - Mengran Zhao
- Beijing Key Laboratory for Precancerous Lesion of Digestive Disease, Department of Gastroenterology, Beijing Friendship Hospital, Capital Medical University, Beijing, P.R. China
| | - Bing Zhou
- Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, P.R. China.
| |
Collapse
|
50
|
Niemand Wolhuter N, Ngakane L, de Wet TJ, Warren RM, Williams MJ. The Mycobacterium smegmatis HesB Protein, MSMEG_4272, Is Required for In Vitro Growth and Iron Homeostasis. Microorganisms 2023; 11:1573. [PMID: 37375075 DOI: 10.3390/microorganisms11061573] [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: 05/17/2023] [Revised: 06/07/2023] [Accepted: 06/09/2023] [Indexed: 06/29/2023] Open
Abstract
A-type carrier (ATC) proteins are proposed to function in the biogenesis of Fe-S clusters, although their exact role remains controversial. The genome of Mycobacterium smegmatis encodes a single ATC protein, MSMEG_4272, which belongs to the HesB/YadR/YfhF family of proteins. Attempts to generate an MSMEG_4272 deletion mutant by two-step allelic exchange were unsuccessful, suggesting that the gene is essential for in vitro growth. CRISPRi-mediated transcriptional knock-down of MSMEG_4272 resulted in a growth defect under standard culture conditions, which was exacerbated in mineral-defined media. The knockdown strain displayed reduced intracellular iron levels under iron-replete conditions and increased susceptibility to clofazimine, 2,3-dimethoxy-1,4-naphthoquinone (DMNQ), and isoniazid, while the activity of the Fe-S containing enzymes, succinate dehydrogenase, and aconitase were not affected. This study suggests that MSMEG_4272 plays a role in the regulation of intracellular iron levels and is required for in vitro growth of M. smegmatis, particularly during exponential growth.
Collapse
Affiliation(s)
- Nandi Niemand Wolhuter
- NRF/DSI Centre of Excellence for Biomedical Tuberculosis Research, South African Medical Research Council Centre for Tuberculosis Research, Division of Molecular Biology and Human Genetics, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town 7505, South Africa
| | - Lerato Ngakane
- NRF/DSI Centre of Excellence for Biomedical Tuberculosis Research, South African Medical Research Council Centre for Tuberculosis Research, Division of Molecular Biology and Human Genetics, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town 7505, South Africa
| | - Timothy J de Wet
- SAMRC/NHLS/UCT Molecular Mycobacteriology Research Unit, Department of Pathology, University of Cape Town, Cape Town 7925, South Africa
- Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Cape Town 7925, South Africa
| | - Robin M Warren
- NRF/DSI Centre of Excellence for Biomedical Tuberculosis Research, South African Medical Research Council Centre for Tuberculosis Research, Division of Molecular Biology and Human Genetics, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town 7505, South Africa
| | - Monique J Williams
- NRF/DSI Centre of Excellence for Biomedical Tuberculosis Research, South African Medical Research Council Centre for Tuberculosis Research, Division of Molecular Biology and Human Genetics, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town 7505, South Africa
- Department of Molecular and Cell Biology, University of Cape Town, Cape Town 7700, South Africa
| |
Collapse
|