1
|
Skrypnik K, Olejnik-Schmidt A, Mikołajczyk-Stecyna J, Schmidt M, Suliburska J. Influence of supplementation with probiotic bacteria Lactiplantibacillus plantarum and Latilactobacillus curvatus on selected parameters of duodenum iron metabolism in rats on a high-fat, iron-deficient diet. Nutrition 2025; 129:112591. [PMID: 39442381 DOI: 10.1016/j.nut.2024.112591] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Revised: 09/06/2024] [Accepted: 09/15/2024] [Indexed: 10/25/2024]
Abstract
OBJECTIVES A high-fat, iron (Fe)-deficient Western diet induces obesity and dysregulates Fe metabolism. We compared the influence of Lactiplantibacillus plantarum and Latilactobacillus curvatus with and without Fe supplementation on duodenal Fe uptake under high-fat diet conditions. METHODS Rats were fed a high-fat diet (HF group) or high-fat, Fe-deficient diet (HFDEF group) or control diet (C group) for 8 wk. For the next 8 wk, the rats in the C and HF groups continued on the same diet, whereas the rats in the HFDEF group were divided into six groups and fed high-fat, Fe-deficient diet combinations with L. plantarum (Lp), L. curvatus (Lc), and Fe supplementation (HFDEF, HFDEFFe, HFDEFLp, HFDEFLc, HFDEFFeLp, HFDEFFeLc). Duodenum and serum samples were collected for analysis. RESULTS In the duodenum, the Fe content was higher in the HFDEFFeLp and HFDEFFeLc groups; the ferroportin level was higher in the HFDEFFeLp and HFDEFFeLc groups versus the HF group; the divalent metal transporter 1 level was higher in the HFDEFFeLc group versus the C and HF groups; and duodenal cytochrome B was higher in the HFDEFLc versus all the other groups. In addition, duodenal expression of the solute carrier family 11 member 2 gene was higher in the HFDEF group versus the C, HF, HFDEFFe, HFDEFFeLp, and HFDEFFeLc groups; that of the TFRC gene was higher in the HFDEFFeLc group versus the C, HF, HFDEF, and HFDEFFe groups; and that of the HJV gene was higher in the HFDEFFeLp group versus the C, HF, HFDEF, HFDEFFe, and HFDEFLc groups. CONCLUSIONS L. plantarum and L. curvatus supplementation shows some potential to enhance duodenal cellular Fe uptake in rats on a high-fat, Fe-deficient diet.
Collapse
Affiliation(s)
- Katarzyna Skrypnik
- Institute of Human Nutrition and Dietetics, Poznań University of Life Sciences, Poznań, Poland.
| | - Agnieszka Olejnik-Schmidt
- Department of Food Biotechnology and Microbiology, Poznań University of Life Sciences, Poznań, Poland
| | | | - Marcin Schmidt
- Department of Food Biotechnology and Microbiology, Poznań University of Life Sciences, Poznań, Poland
| | - Joanna Suliburska
- Institute of Human Nutrition and Dietetics, Poznań University of Life Sciences, Poznań, Poland
| |
Collapse
|
2
|
Tan J, Chen J, Roxby D, Chooi WH, Nguyen TD, Ng SY, Han J, Chew SY. Using magnetic resonance relaxometry to evaluate the safety and quality of induced pluripotent stem cell-derived spinal cord progenitor cells. Stem Cell Res Ther 2024; 15:465. [PMID: 39639398 PMCID: PMC11622678 DOI: 10.1186/s13287-024-04070-y] [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: 07/26/2024] [Accepted: 11/20/2024] [Indexed: 12/07/2024] Open
Abstract
BACKGROUND The emergence of induced pluripotent stem cells (iPSCs) offers a promising approach for replacing damaged neurons and glial cells, particularly in spinal cord injuries (SCI). Despite its merits, iPSC differentiation into spinal cord progenitor cells (SCPCs) is variable, necessitating reliable assessment of differentiation and validation of cell quality and safety. Phenotyping is often performed via label-based methods including immunofluorescent staining or flow cytometry analysis. These approaches are often expensive, laborious, time-consuming, destructive, and severely limits their use in large scale cell therapy manufacturing settings. On the other hand, cellular biophysical properties have demonstrated a strong correlation to cell state, quality and functionality and can be measured with ingenious label-free technologies in a rapid and non-destructive manner. METHOD In this study, we report the use of Magnetic Resonance Relaxometry (MRR), a rapid and label-free method that indicates iron levels based on its readout (T2). Briefly, we differentiated human iPSCs into SCPCs and compared key iPSC and SCPC cellular markers to their intracellular iron content (Fe3+) at different stages of the differentiation process. RESULTS With MRR, we found that intracellular iron of iPSCs and SCPCs were distinctively different allowing us to accurately reflect varying levels of residual undifferentiated iPSCs (i.e., OCT4+ cells) in any given population of SCPCs. MRR was also able to predict Day 10 SCPC OCT4 levels from Day 1 undifferentiated iPSC T2 values and identified poorly differentiated SCPCs with lower T2, indicative of lower neural progenitor (SOX1) and stem cell (Nestin) marker expression levels. Lastly, MRR was able to provide predictive indications for the extent of differentiation to Day 28 spinal cord motor neurons (ISL-1/SMI-32) based on the T2 values of Day 10 SCPCs. CONCLUSION MRR measurements of iPSCs and SCPCs has clearly indicated its capabilities to identify and quantify key phenotypes of iPSCs and SCPCs for end-point validation of safety and quality parameters. Thus, our technology provides a rapid label-free method to determine critical quality attributes in iPSC-derived progenies and is ideally suited as a quality control tool in cell therapy manufacturing.
Collapse
Affiliation(s)
- Jerome Tan
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, Singapore, Singapore
- HealthTech @ NTU, Interdisciplinary Graduate Programme, Nanyang Technological University, Singapore, Singapore
- CAMP IRG, SMART Centre, CREATE, Singapore, Singapore
| | - Jiahui Chen
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, Singapore, Singapore
| | - Daniel Roxby
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, Singapore, Singapore
- CAMP IRG, SMART Centre, CREATE, Singapore, Singapore
| | - Wai Hon Chooi
- Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | | | - Shi Yan Ng
- Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Jongyoon Han
- CAMP IRG, SMART Centre, CREATE, Singapore, Singapore.
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, USA.
- Department of Electrical Engineering and Computer Science, Massachusetts Institute of Technology, Cambridge, USA.
| | - Sing Yian Chew
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, Singapore, Singapore.
- CAMP IRG, SMART Centre, CREATE, Singapore, Singapore.
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore.
- School of Materials Science and Engineering, Nanyang Technological University, Singapore, Singapore.
| |
Collapse
|
3
|
Cui Y, Du X, Li Y, Wang D, Lv Z, Yuan H, Chen Y, Liu J, Sun Y, Wang W. Imbalanced and Unchecked: The Role of Metal Dyshomeostasis in Driving COPD Progression. COPD 2024; 21:2322605. [PMID: 38591165 DOI: 10.1080/15412555.2024.2322605] [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: 10/25/2023] [Accepted: 02/19/2024] [Indexed: 04/10/2024]
Abstract
Chronic obstructive pulmonary disease (COPD) is a chronic respiratory condition characterized by persistent inflammation and oxidative stress, which ultimately leads to progressive restriction of airflow. Extensive research findings have cogently suggested that the dysregulation of essential transition metal ions, notably iron, copper, and zinc, stands as a critical nexus in the perpetuation of inflammatory processes and oxidative damage within the lungs of COPD patients. Unraveling the intricate interplay between metal homeostasis, oxidative stress, and inflammatory signaling is of paramount importance in unraveling the intricacies of COPD pathogenesis. This comprehensive review aims to examine the current literature on the sources, regulation, and mechanisms by which metal dyshomeostasis contributes to COPD progression. We specifically focus on iron, copper, and zinc, given their well-characterized roles in orchestrating cytokine production, immune cell function, antioxidant depletion, and matrix remodeling. Despite the limited number of clinical trials investigating metal modulation in COPD, the advent of emerging methodologies tailored to monitor metal fluxes and gauge responses to chelation and supplementation hold great promise in unlocking the potential of metal-based interventions. We conclude that targeted restoration of metal homeostasis represents a promising frontier for ameliorating pathological processes driving COPD progression.
Collapse
Affiliation(s)
- Ye Cui
- Department of Immunology, School of Basic Medical Sciences, Capital Medical University, Beijing, People's Republic of China
| | - Xinqian Du
- Department of Immunology, School of Basic Medical Sciences, Capital Medical University, Beijing, People's Republic of China
| | - Yunqi Li
- Department of Immunology, School of Basic Medical Sciences, Capital Medical University, Beijing, People's Republic of China
| | - Dan Wang
- Department of Immunology, School of Basic Medical Sciences, Capital Medical University, Beijing, People's Republic of China
| | - Zhe Lv
- Department of Immunology, School of Basic Medical Sciences, Capital Medical University, Beijing, People's Republic of China
| | - Huihui Yuan
- Department of Immunology, School of Basic Medical Sciences, Capital Medical University, Beijing, People's Republic of China
| | - Yan Chen
- Department of Immunology, School of Basic Medical Sciences, Capital Medical University, Beijing, People's Republic of China
| | - Jie Liu
- Department of Immunology, School of Basic Medical Sciences, Capital Medical University, Beijing, People's Republic of China
| | - Ying Sun
- Department of Immunology, School of Basic Medical Sciences, Capital Medical University, Beijing, People's Republic of China
| | - Wei Wang
- Department of Immunology, School of Basic Medical Sciences, Capital Medical University, Beijing, People's Republic of China
| |
Collapse
|
4
|
Wu Z, Xu W, Wang X, Peng D, Jiang Z. Exploring the Causal Relationship Between Inflammatory Cytokines and MRI-Derived Brain Iron: A Mendelian Randomization Study. Brain Behav 2024; 14:e70181. [PMID: 39643932 PMCID: PMC11624122 DOI: 10.1002/brb3.70181] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/28/2024] [Revised: 10/20/2024] [Accepted: 11/17/2024] [Indexed: 12/09/2024] Open
Abstract
BACKGROUND The association between inflammation and brain iron deposition is widely acknowledged. However, the precise causal impact of peripheral inflammatory cytokines on changes in brain iron content remains uncertain. METHODS The study utilized an available genome-wide association study (GWAS) summary associated with inflammatory cytokines from The Cardiovascular Risk in Young Finns Study and the FINRISK surveys. The GWAS data for brain iron markers were obtained from the UK Biobank. We assessed the iron content of each brain region using susceptibility-weighted magnetic resonance imaging, utilizing both quantitative susceptibility mapping and T2* measurements. The primary outcomes were susceptibility (χ) and T2*, which serve as indices of iron deposition. To investigate the causal relationship between exposure and outcome, we primarily employed inverse variance weighting, MR Egger, weighted median, simple mode, and weighted mode methods, collectively enhancing the robustness of our results. RESULTS The results of MR analyses demonstrate that our study unveiled that nerve growth factor-β, hepatocyte growth factor, interleukin-1 (IL-1), IL-8, macrophage inflammatory protein 1α, and tumor necrosis factor-α were associated with elevated brain iron content in the regions of left hippocampus, putamen, left thalamus, right pallidum, right hippocampus, left amygdala, respectively. Furthermore, our investigation provides evidence for a negative relationship between IL-1, IL-17, monocyte chemotactic protein-3, tumor necrosis factor-β, and brain iron content in distinct regions. CONCLUSIONS Our findings suggest a causal association between circulating inflammatory cytokines and brain iron deposition across various brain regions. This provides new insights into the immunopathogenesis of neurodegenerative diseases and potential preventive strategies targeting iron metabolism.
Collapse
Affiliation(s)
- Zhounan Wu
- Department of Orthopaedic SurgeryThe Second Xiangya Hospital, Central South UniversityChangshaHunanChina
| | - Wantong Xu
- Department of Orthopaedic SurgeryThe Second Xiangya Hospital, Central South UniversityChangshaHunanChina
| | - Xuemei Wang
- Department of Laboratory MedicineThe Second Xiangya Hospital, Central South UniversityChangshaHunanChina
| | - Dan Peng
- Department of Orthopaedic SurgeryThe Second Xiangya Hospital, Central South UniversityChangshaHunanChina
| | - Zhongbiao Jiang
- Department of RadiologyThe Second Xiangya Hospital, Central South UniversityChangshaHunanChina
| |
Collapse
|
5
|
Saleh AA, Bawahab AA, Bafail DA, Alosaimi ME, Abd-Elhakim YM, Mohamed AAR, Khamis T, Metwally MMM, Alotaibi BS, El-Gamal M, Dahran N, Alamri AS, ElAshmouny N. Biofabricated zinc oxide nanoparticles mitigate acrylamide-induced immune toxicity and modulate immune-related genes and microRNA in rats. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2024:10.1007/s00210-024-03566-x. [PMID: 39549065 DOI: 10.1007/s00210-024-03566-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/07/2024] [Accepted: 10/23/2024] [Indexed: 11/18/2024]
Abstract
This study evaluated the potential efficacy of eco-friendly biofabricated zinc oxide nanoparticles (GS-ZnONP) (10 mg/kg b.wt) to reduce the impacts of long-term oral acrylamide (ALD) exposure (20 mg/kg b.wt) on the blood cells, immune components, splenic oxidative status, and expression of CD20, CD3, CD4, CD8, TNF-α, caspase-3, microRNA-181a-5p, and microRNA-125-5p in rats in a 60-day experiment. The study findings revealed that GS-ZnONP significantly corrected the ALD-induced hematological alterations. Additionally, the ALD-induced increase in the serum C3, splenic ROS, CD4, CD8, and MDA and histological alterations were significantly repressed in the ALD + GS-ZnONP-treated rats. Instead, the depleted splenic antioxidants and Zn contents were markedly reestablished in the ALD + GS-ZnONP-treated group. Additionally, a significant upregulation of expression of splenic CD3, CD4, CD8, CD20, TNF-α, and caspase-3, but downregulation of microRNA-181a-5p and microRNA-125-5p was detected in the ALD-exposed group. Yet, the former deviations in the gene expressions were corrected in the ALD + GS-ZnONP-treated rats. Furthermore, GS-ZnONP treatment significantly minimized the increased caspase-3 and TNF-α immunoexpression in the splenic tissues of ALD-exposed rats. Conclusively, the study findings proved the efficacy of GS-ZnONP in rescuing ALD-induced disturbances in blood cell populations, immune function, splenic antioxidant status, and immune-related gene expression.
Collapse
Affiliation(s)
- Ayman A Saleh
- Department of Pathology, College of Medicine, University of Hail, Hail, Saudi Arabia
| | - Ahmed Abdulwahab Bawahab
- Department of Basic Medical Sciences, College of Medicine, University of Jeddah, Jeddah, Saudi Arabia
| | - Duaa Abdullah Bafail
- Department of Clinical Pharmacology, Faculty of Medicine, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Manal E Alosaimi
- Department of Basic Sciences, College of Medicine, Princess Nourah Bint Abdulrahman University, P.O Box 84428, 11671, Riyadh, Saudi Arabia.
| | - Yasmina M Abd-Elhakim
- Department of Forensic Medicine and Toxicology, Faculty of Veterinary Medicine, Zagazig University, Zagazig, 44519, Egypt
| | - Amany Abdel-Rahman Mohamed
- Department of Forensic Medicine and Toxicology, Faculty of Veterinary Medicine, Zagazig University, Zagazig, 44519, Egypt
| | - Tarek Khamis
- Department of Pharmacology, Faculty of Veterinary Medicine, Zagazig University, Zagazig, 44519, Egypt
- Laboratory of Biotechnology, Faculty of Veterinary Medicine, Zagazig University, Zagazig, 44519, Egypt
| | - Mohamed M M Metwally
- Department of Pathology and Clinical Pathology, Faculty of Veterinary Medicine, King Salman International University, Ras Sidr, Egypt
- Department of Pathology, Faculty of Veterinary Medicine, Zagazig University, Zagazig, Egypt
| | - Badriyah S Alotaibi
- Department of Pharmaceutical Sciences, College of Pharmacy, Princess Nourah Bint Abdulrahman University, P.O. Box 84428, 11671, Riyadh, Saudi Arabia
| | - Mohamed El-Gamal
- Forensic Medicine and Clinical Toxicology Department, Faculty of Medicine, Mansoura University, Mansoura, Egypt
- Department of Biological Sciences, Faculty of Science, New Mansoura University, New Mansoura City, Egypt
| | - Naief Dahran
- Department of Basic Medical Sciences, College of Medicine, University of Jeddah, Jeddah, Saudi Arabia
| | - Ahlam Saleh Alamri
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Al-Quwayiyah, Shaqra University, Riyadh, Saudi Arabia
| | - Naira ElAshmouny
- Histology and Cell Biology, Faculty of Medicine, Kafr Elsheikh University, Kafr Elsheikh, Egypt
| |
Collapse
|
6
|
Wen S, Xu D, Yuan Y, Xu Z, Li Y, Gong M, Yuan X, Zhou L. The Effect of Diabetic Ketoacidosis and Hyperosmolar Hyperglycemic on the Metabolic Tumor Markers: A Real-World Retrospective Study. Diabetes Metab Syndr Obes 2024; 17:4115-4133. [PMID: 39502450 PMCID: PMC11537188 DOI: 10.2147/dmso.s487398] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/17/2024] [Accepted: 10/26/2024] [Indexed: 11/08/2024] Open
Abstract
Objective Serum tumor markers have been discovered to be elevated in individuals with diabetes mellitus (DM); however, their significance in diabetic ketoacidosis (DKA) and hyperosmolar hyperglycemia (HHS) is unknown. We evaluated these indicators in the patients with DKA and HHS. Materials and Methods We retrospectively collected the laboratory data of 565 diabetic patients from Shanghai Pudong Hospital between Jan, 2019 and May, 2023, including 300 patients with type 2 diabetes mellitus (T2DM), 206 with DKA, and 59 with HHS. Serum tumor biomarkers and further clinical laboratory tests were compared among the three groups. Patients with conspicuous tumor evidence were excluded from the study. Results We found significantly higher levels of carbohydrate antigen 199 (CA199) in DKA (p<0.01), carcinoembryonic antigen (CEA), complex prostate specific antigen (CPSA) (p<0.01), prostate specific antigen (PSA) (p<0.05) in HHS, as well as the plasma lipid profile, iron, and electrolytes, in addition to decreased thyroid function, hepatic and renal function, and cardiac function (p<0.05). A Spearman correlational study revealed that osmolar levels were significantly positively correlated with myoglobin (MYO) and cardiac troponin I (cTNI), whereas serum iron concentration (r=0.520) was positively correlated with CEA levels in HHS. pH was negatively correlated with CA199 (r=-0.195), while HbA1c (r=0.22), globin (r=0.341), and total cholesterol (TC) (r=0.191) were positively correlated with elevated CA199 levels. Moreover, the multilinear regression investigation identified osmolarity as a significant determinant for CEA, as well as other parameters, and all proved to be proper predictors for CEA in HHS via ROC curve establishment. Conclusion Elevated CA199 levels in DKA were associated with acidosis, whereas HHS with elevated CEA levels may be related to iron homeostasis and could be predicted via the osmolar degree and other predictors.
Collapse
Grants
- Integrated Traditional Chinese and Western Medicine (YC-2023-0404), Fudan Zhangjiang Clinical Medicine Innovation Fund Project (KP0202118), Fudan Good Practice Program of Teaching and Learning (FD2023A227), Project of Key Medical Discipline of Pudong Hospital of Fudan University (Zdxk2020-11), Project of Key Medical Specialty and Treatment Center of Pudong Hospital of Fudan University (Zdzk2020-24), Integrative Medicine special fund of Shanghai Municipal Health Planning Committee (ZHYY- ZXYJHZX-2-201712), Special Department Fund of the Pudong New Area Health Planning Commission (PWZzk2017-03), Outstanding Leaders Training Program of Pudong Health Bureau of Shanghai (PWR12014-06), Pudong New Area Clinical Plateau Discipline Project (PWYgy-2021-03), the Natural Science Foundation of China (21675034), National Natural Science Foundation of China (81370932), Shanghai Natural Science Foundation (19ZR1447500), Pudong New Area Clinical Characteristic Discipline Project (PWYts2021-11), Pudong New Area Clinical Characteristic Discipline Project (PWYts2021-01), Wenzhou Medical University Education Grant (JG2021197)
Collapse
Affiliation(s)
- Song Wen
- Department of Endocrinology, Shanghai Pudong Hospital, Fudan University, Shanghai, 201399, People’s Republic of China
| | - Dongxiang Xu
- Department of Endocrinology, Shanghai Pudong Hospital, Fudan University, Shanghai, 201399, People’s Republic of China
| | - Yue Yuan
- Department of Endocrinology, Shanghai Pudong Hospital, Fudan University, Shanghai, 201399, People’s Republic of China
| | - Zhimin Xu
- Department of Endocrinology, Shanghai Pudong Hospital, Fudan University, Shanghai, 201399, People’s Republic of China
| | - Yanyan Li
- Department of Endocrinology, Shanghai Pudong Hospital, Fudan University, Shanghai, 201399, People’s Republic of China
| | - Min Gong
- Department of Endocrinology, Shanghai Pudong Hospital, Fudan University, Shanghai, 201399, People’s Republic of China
| | - Xinlu Yuan
- Department of Endocrinology, Shanghai Pudong Hospital, Fudan University, Shanghai, 201399, People’s Republic of China
| | - Ligang Zhou
- Department of Endocrinology, Shanghai Pudong Hospital, Fudan University, Shanghai, 201399, People’s Republic of China
- Shanghai Key Laboratory of Vascular Lesions Regulation and Remodeling, Shanghai Pudong Hospital, Shanghai, 201399, People’s Republic of China
| |
Collapse
|
7
|
Belbellaj W, Lona-Durazo F, Bodano C, Busseuil D, Cyr MC, Fiorillo E, Mulas A, Provost S, Steri M, Tanaka T, Vanderwerff B, Wang J, Byrne RP, Cucca F, Dubé MP, Ferrucci L, McLaughlin RL, Tardif JC, Zawistowski M, Gagliano Taliun SA. The role of genetically predicted serum iron levels on neurodegenerative and cardiovascular traits. Sci Rep 2024; 14:24588. [PMID: 39427026 PMCID: PMC11490554 DOI: 10.1038/s41598-024-76245-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Accepted: 10/11/2024] [Indexed: 10/21/2024] Open
Abstract
Iron is an essential mineral that supports numerous biological functions. Studies have reported associations between iron dysregulation and certain cardiovascular and neurodegenerative diseases, but the direction of influence is not clear. Our goal was to use computational approaches to better understand the role of genetically predicted iron levels on disease risk. We meta-analyzed genome-wide association study summary statistics for serum iron levels from two cohorts and two previous meta-analyses. We then obtained summary statistics from 11 neurodegenerative, cerebrovascular, cardiovascular or lipid traits to assess global and regional genetic correlation between iron levels and these traits. We used two-sample Mendelian randomization (MR) to estimate causal effects. Sex-stratified analyses were also carried out to identify effects potentially differing by sex. Overall, we identified three significant global correlations between iron levels and (i) coronary heart disease, (ii) triglycerides, and (iii) high-density lipoprotein (HDL) cholesterol levels. A total of 194 genomic regions had significant (after correction for multiple testing) local correlations between iron levels and the 11 tested traits. MR analysis revealed two potential causal relationships, between genetically predicted iron levels and (i) total cholesterol or (ii) non-HDL cholesterol. Sex-stratified analyses suggested a potential protective effect of iron levels on Parkinson's disease risk in females, but not in males. Our results will contribute to a better understanding of the genetic basis underlying iron in cardiovascular and neurological health in aging, and to the eventual identification of new preventive interventions or therapeutic avenues for diseases which affect women and men worldwide.
Collapse
Affiliation(s)
- Wiame Belbellaj
- Faculty of Medicine, Université de Montréal, Montreal, QC, H3C 3J7, Canada
- Research Centre, Montreal Heart Institute, 5000 Bélanger Street, Montreal, QC, H1T 1C8, Canada
| | - Frida Lona-Durazo
- Faculty of Medicine, Université de Montréal, Montreal, QC, H3C 3J7, Canada
- Research Centre, Montreal Heart Institute, 5000 Bélanger Street, Montreal, QC, H1T 1C8, Canada
| | - Cinzia Bodano
- Institute for Genetic and Biomedical Research, National Research Council (CNR), 09042, Monserrato-Cagliari, Italy
| | - David Busseuil
- Research Centre, Montreal Heart Institute, 5000 Bélanger Street, Montreal, QC, H1T 1C8, Canada
| | - Marie-Christyne Cyr
- Research Centre, Montreal Heart Institute, 5000 Bélanger Street, Montreal, QC, H1T 1C8, Canada
- Université de Montréal Beaulieu-Saucier Pharmacogenomics Centre, Montreal, QC, H1T 1C8, Canada
| | - Edoardo Fiorillo
- Institute for Genetic and Biomedical Research, National Research Council (CNR), 08045, Lanusei, Italy
| | - Antonella Mulas
- Institute for Genetic and Biomedical Research, National Research Council (CNR), 08045, Lanusei, Italy
| | - Sylvie Provost
- Research Centre, Montreal Heart Institute, 5000 Bélanger Street, Montreal, QC, H1T 1C8, Canada
- Université de Montréal Beaulieu-Saucier Pharmacogenomics Centre, Montreal, QC, H1T 1C8, Canada
| | - Maristella Steri
- Institute for Genetic and Biomedical Research, National Research Council (CNR), 09042, Monserrato-Cagliari, Italy
| | - Toshiko Tanaka
- Translational Gerontology Branch, National Institutes on Aging, Baltimore, MD, USA
| | - Brett Vanderwerff
- Department of Biostatistics and Center for Statistical Genetics, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Jiongming Wang
- Department of Biostatistics and Center for Statistical Genetics, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Ross P Byrne
- Smurfit Institute of Genetics, Trinity College Dublin, Dublin, D02 DK07, Republic of Ireland
| | - Francesco Cucca
- Department of Biomedical Sciences, University of Sassari, 07100, Sassari, Italy
| | - Marie-Pierre Dubé
- Research Centre, Montreal Heart Institute, 5000 Bélanger Street, Montreal, QC, H1T 1C8, Canada
- Université de Montréal Beaulieu-Saucier Pharmacogenomics Centre, Montreal, QC, H1T 1C8, Canada
- Department of Medicine, Faculty of Medicine, Université de Montréal, Montreal, QC, H3C 3J7, Canada
| | - Luigi Ferrucci
- Translational Gerontology Branch, National Institutes on Aging, Baltimore, MD, USA
| | - Russell L McLaughlin
- Smurfit Institute of Genetics, Trinity College Dublin, Dublin, D02 DK07, Republic of Ireland
| | - Jean-Claude Tardif
- Research Centre, Montreal Heart Institute, 5000 Bélanger Street, Montreal, QC, H1T 1C8, Canada
- Department of Medicine, Faculty of Medicine, Université de Montréal, Montreal, QC, H3C 3J7, Canada
| | - Matthew Zawistowski
- Department of Biostatistics and Center for Statistical Genetics, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Sarah A Gagliano Taliun
- Research Centre, Montreal Heart Institute, 5000 Bélanger Street, Montreal, QC, H1T 1C8, Canada.
- Department of Medicine, Faculty of Medicine, Université de Montréal, Montreal, QC, H3C 3J7, Canada.
- Department of Neurosciences, Faculty of Medicine, Université de Montréal, Montreal, QC, H3C 3J7, Canada.
| |
Collapse
|
8
|
Izuora K, Alver A, Basu A, Batra K, Williams SJ, Ebersole JL. The Association of Dietary Micronutrient Intake and Systemic Inflammation among Patients with Type 2 Diabetes: A Cross-Sectional Study. Healthcare (Basel) 2024; 12:1804. [PMID: 39337145 PMCID: PMC11431254 DOI: 10.3390/healthcare12181804] [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: 07/30/2024] [Revised: 08/30/2024] [Accepted: 09/03/2024] [Indexed: 09/30/2024] Open
Abstract
Inflammation contributes to the pathogenesis of type 2 diabetes (T2DM). This study sought to document how the systemic biomarkers of inflammation varied based on food choices among patients with T2DM. This cross-sectional study enrolled ambulatory patients with T2DM. Demographic and clinical information was collected. Five drops of fingerstick blood were collected using an absorbent paper device (HemaSpot HFR). C-reactive protein (CRP), serum amyloid A protein (SAA), and fibrinogen were measured using a Luminex assay. Patient-generated 7-day food diaries were analyzed using a validated food processor software. Data were analyzed by Pearson's correlation tests, linear regression and logistic regression with the significance level set at 0.05. Among the 71 participants, 43 (60.6%) were females. The average age and duration of T2DM were 64.1 ± 10.3 and 15.8 ± 9.1 years, respectively. In a simple linear regression run with selected micronutrients, iron [F (1, 53) = 5.319, p < 0.05, adj. R2 = 0.074] significantly predicted plasma CRP. This significance was lost with multiple linear regressions including age, gender, BMI, T2DM duration, T2DM complications, glycohemoglobin A1c (HbA1c) and other micronutrients. The average intake of most micronutrients by the participants was below the recommended daily intake. A higher intake of iron-rich foods was associated with higher levels of systemic inflammation in a simple linear regression model, but the association was not present after adjusting for patient factors like age, gender, BMI and T2DM-related variables. This relationship needs to be explored further given the key role of inflammation in the pathogenesis of T2DM and its associated complications.
Collapse
Affiliation(s)
- Kenneth Izuora
- Department of Internal Medicine, Kirk Kerkorian School of Medicine at UNLV, University of Nevada, Las Vegas, NV 89102, USA
| | - Amalie Alver
- Department of Internal Medicine, Kirk Kerkorian School of Medicine at UNLV, University of Nevada, Las Vegas, NV 89102, USA
| | - Arpita Basu
- Department of Kinesiology and Nutrition Sciences, School of Integrated Health Sciences, University of Nevada, Las Vegas, NV 89154, USA
| | - Kavita Batra
- Department of Medical Education and Office of Research, Kirk Kerkorian School of Medicine at UNLV, University of Nevada, Las Vegas, NV 89102, USA
| | - Shelley J Williams
- School of Dental Medicine, University of Nevada, Las Vegas, NV 89102, USA
| | - Jeffrey L Ebersole
- School of Dental Medicine, University of Nevada, Las Vegas, NV 89102, USA
| |
Collapse
|
9
|
Bolen ML, Nuñes Gomes B, Gill B, Menees KB, Staley H, Jernigan J, Tansey MG. Peripheral Blood Immune Cells from Individuals with Parkinson's Disease or Inflammatory Bowel Disease Share Deficits in Iron Storage and Transport that are Modulated by Non-Steroidal Anti-Inflammatory Drugs. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.19.608634. [PMID: 39229100 PMCID: PMC11370434 DOI: 10.1101/2024.08.19.608634] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 09/05/2024]
Abstract
Parkinson's Disease (PD) is a multisystem disorder in which dysregulated neuroimmune crosstalk and inflammatory relay via the gut-blood-brain axis have been implicated in PD pathogenesis. Although alterations in circulating inflammatory cytokines and reactive oxygen species (ROS) have been associated with PD, no biomarkers have been identified that predict clinical progression or disease outcome. Gastrointestinal (GI) dysfunction, which involves perturbation of the underlying immune system, is an early and often-overlooked symptom that affects up to 80% of individuals living with PD. Interestingly, 50-70% of individuals with inflammatory bowel disease (IBD), a GI condition that has been epidemiologically linked to PD, display chronic illness-induced anemia - which drives toxic accumulation of iron in the gut. Ferroptotic (or iron loaded) cells have small and dysmorphic mitochondria-suggesting that mitochondrial dysfunction is a consequence of iron accumulation. In pro-inflammatory environments, iron accumulates in immune cells, suggesting a possible connection and/or synergy between iron dysregulation and immune cell dysfunction. Peripheral blood mononuclear cells (PBMCs) recapitulate certain PD-associated neuropathological and inflammatory signatures and can act as communicating messengers in the gut-brain axis. Additionally, this communication can be modulated by several environmental factors; specifically, our data further support existing literature demonstrating a role for non-steroidal anti-inflammatory drugs (NSAIDs) in modulating immune transcriptional states in inflamed individuals. A mechanism linking chronic gut inflammation to iron dysregulation and mitochondrial function within peripheral immune cells has yet to be identified in conferring risk for PD. To that end, we isolated PBMCs and simultaneously evaluated their directed transcriptome and bioenergetic status, to investigate if iron dysregulation and mitochondrial sensitization are linked in individuals living with PD or IBD because of chronic underlying remittent immune activation. We have identified shared features of peripheral inflammation and immunometabolism in individuals living with IBD or PD that may contribute to the epidemiological association reported between IBD and risk for PD.
Collapse
Affiliation(s)
- MacKenzie L. Bolen
- Center for Translational Research in Neurodegenerative Disease, College of Medicine, University of Florida, Gainesville, FL, USA
- Department of Neuroscience, College of Medicine, University of Florida, Gainesville, FL, USA
- McKnight Brain Institute, University of Florida, Gainesville, FL, USA
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD, USA
| | - Beatriz Nuñes Gomes
- Center for Translational Research in Neurodegenerative Disease, College of Medicine, University of Florida, Gainesville, FL, USA
- Department of Neuroscience, College of Medicine, University of Florida, Gainesville, FL, USA
- McKnight Brain Institute, University of Florida, Gainesville, FL, USA
| | - Blake Gill
- Department of Surgery, Northwestern University
| | - Kelly B. Menees
- Center for Translational Research in Neurodegenerative Disease, College of Medicine, University of Florida, Gainesville, FL, USA
- Department of Neuroscience, College of Medicine, University of Florida, Gainesville, FL, USA
- McKnight Brain Institute, University of Florida, Gainesville, FL, USA
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD, USA
| | - Hannah Staley
- Center for Translational Research in Neurodegenerative Disease, College of Medicine, University of Florida, Gainesville, FL, USA
- Department of Neuroscience, College of Medicine, University of Florida, Gainesville, FL, USA
- McKnight Brain Institute, University of Florida, Gainesville, FL, USA
| | - Janna Jernigan
- Center for Translational Research in Neurodegenerative Disease, College of Medicine, University of Florida, Gainesville, FL, USA
- Department of Neuroscience, College of Medicine, University of Florida, Gainesville, FL, USA
- McKnight Brain Institute, University of Florida, Gainesville, FL, USA
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD, USA
| | - Malú Gámez Tansey
- Center for Translational Research in Neurodegenerative Disease, College of Medicine, University of Florida, Gainesville, FL, USA
- Department of Neuroscience, College of Medicine, University of Florida, Gainesville, FL, USA
- McKnight Brain Institute, University of Florida, Gainesville, FL, USA
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD, USA
- Norman Fixel Institute for Neurological Diseases, University of Florida, Gainesville, FL, USA
| |
Collapse
|
10
|
Goes JVC, Viana MDA, Sampaio LR, Cavalcante CBA, Melo MMDL, de Oliveira RTG, Borges DDP, Gonçalves PG, Pinheiro RF, Ribeiro-Junior HL. Gene expression patterns of Sirtuin family members (SIRT1 TO SIRT7): Insights into pathogenesis and prognostic of Myelodysplastic neoplasm. Gene 2024; 915:148428. [PMID: 38575099 DOI: 10.1016/j.gene.2024.148428] [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: 09/21/2023] [Revised: 01/02/2024] [Accepted: 04/01/2024] [Indexed: 04/06/2024]
Abstract
To assess and validate the gene expression profile of SIRTs (SIRT1, SIRT2, SIRT3, SIRT4, SIRT5, SIRT6, and SIRT7) in relation to the pathogenesis and prognostic progression of Myelodysplastic neoplasm (MDS). Eighty bone marrow samples of patients with de novo MDS were diagnosed according to WHO 2022 and IPSS-R criteria. Ten bone marrow samples were obtained from elderly healthy volunteers and used as control samples. Gene expression levels of all SIRTs were assessed using RT-qPCR assays. Downregulation of SIRT2 (p = 0.009), SIRT3 (p = 0.048), SIRT4 (p = 0.049), SIRT5 (p = 0.046), SIRT6 (p = 0.043), and SIRT7 (p = 0.047) was identified in MDS patients compared to control individuals. Also, we identified that while SIRT2-7 genes are typically down-regulated in MDS patients compared to normal controls, there are relative expression variations among MDS patient subgroups. Specifically, SIRT4 (p = 0.029) showed increased expression in patients aged 60 or above, and both SIRT2 (p = 0.016) and SIRT3 (p = 0.036) were upregulated in patients with hemoglobin levels below 8 g/dL. SIRT2 (p = 0.045) and SIRT3 (p = 0.033) were highly expressed in patients with chromosomal abnormalities. Different SIRTs exhibited altered expression patterns concerning specific MDS clinical and prognostic characteristics. The downregulation in SIRTs genes (e.g., SIRT2 to SIRT7) expression in Brazilian MDS patients highlights their role in the disease's development. The upregulation of SIRT2 and SIRT3 in severe anemia patients suggests a potential link to manage iron overload-related complications in transfusion-dependent patients. Moreover, the association of SIRT2/SIRT3 with genomic instability and their role in MDS progression signify promising areas for future research and therapeutic targets. These findings underscore the importance of SIRT family in understanding and addressing MDS, offering novel clinical, prognostic, and therapeutic insights for patients with this condition.
Collapse
Affiliation(s)
- João Vitor Caetano Goes
- Center for Research and Drug Development (NPDM), Federal University of Ceara, Fortaleza, Ceara, Brazil; Post-Graduate Program of Pathology, Federal University of Ceara, Fortaleza, Ceara, Brazil
| | - Mateus de Aguiar Viana
- Center for Research and Drug Development (NPDM), Federal University of Ceara, Fortaleza, Ceara, Brazil; Post-Graduate Program in Medical Science, Federal University of Ceara, Fortaleza, Ceara, Brazil.
| | - Leticia Rodrigues Sampaio
- Center for Research and Drug Development (NPDM), Federal University of Ceara, Fortaleza, Ceara, Brazil; Post-Graduate Program in Medical Science, Federal University of Ceara, Fortaleza, Ceara, Brazil.
| | | | - Mayara Magna de Lima Melo
- Center for Research and Drug Development (NPDM), Federal University of Ceara, Fortaleza, Ceara, Brazil; Post-Graduate Program in Medical Science, Federal University of Ceara, Fortaleza, Ceara, Brazil.
| | - Roberta Taiane Germano de Oliveira
- Center for Research and Drug Development (NPDM), Federal University of Ceara, Fortaleza, Ceara, Brazil; Post-Graduate Program in Medical Science, Federal University of Ceara, Fortaleza, Ceara, Brazil.
| | - Daniela de Paula Borges
- Center for Research and Drug Development (NPDM), Federal University of Ceara, Fortaleza, Ceara, Brazil; Post-Graduate Program in Medical Science, Federal University of Ceara, Fortaleza, Ceara, Brazil.
| | - Paola Gyuliane Gonçalves
- Molecular Oncology Research Center, Barretos Cancer Hospital, Barretos, São Paulo, Brazil; Department of Pathology, School of Medicine, Universidade Estadual Paulista, Botucatu, São Paulo, Brazil
| | - Ronald Feitosa Pinheiro
- Center for Research and Drug Development (NPDM), Federal University of Ceara, Fortaleza, Ceara, Brazil; Post-Graduate Program of Pathology, Federal University of Ceara, Fortaleza, Ceara, Brazil; Post-Graduate Program in Medical Science, Federal University of Ceara, Fortaleza, Ceara, Brazil.
| | - Howard Lopes Ribeiro-Junior
- Center for Research and Drug Development (NPDM), Federal University of Ceara, Fortaleza, Ceara, Brazil; Post-Graduate Program of Pathology, Federal University of Ceara, Fortaleza, Ceara, Brazil; Post-Graduate Program in Medical Science, Federal University of Ceara, Fortaleza, Ceara, Brazil.
| |
Collapse
|
11
|
Wu L, Zhang Y, Zhao F, Sheng J, Gu N. Magnetic Characterization of Human Intrinsically Magnetic Monocytes Through a Novel Optical Tracking-Based Magnetic Sensor. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2307306. [PMID: 38312110 DOI: 10.1002/smll.202307306] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Revised: 01/17/2024] [Indexed: 02/06/2024]
Abstract
Intrinsically magnetic cells naturally occur within organisms and are believed to be linked to iron metabolism and certain cellular functions while the functional significance of this magnetism is largely unexplored. To better understand this property, an approach named Optical Tracking-based Magnetic Sensor (OTMS) has been developed. This multi-target tracking system is designed to measure the magnetic moment of individual cells. The OTMS generates a tunable magnetic field and induces movement in magnetic cells that are subsequently analyzed through a learning-based tracking-by-detection system. The magnetic moment of numerous cells can be calculated simultaneously, thereby providing a quantitative tool to assess cellular magnetic properties within populations. Upon deploying the OTMS, a stable population of magnetic cells in human peripheral monocytes is discovered. Further application in the analysis of clinical blood samples reveals an intriguing pattern: the proportion of magnetic monocytes differs significantly between systemic lupus erythematosus (SLE) patients and healthy volunteers. This variation is positively correlated with disease activity, a trend not observed in patients with rheumatoid arthritis (RA). The study, therefore, presents a new frontier in the investigation of the magnetic characteristics of naturally occurring magnetic cells, opening the door to potential diagnostic and therapeutic applications that leverage cellular magnetism.
Collapse
Affiliation(s)
- Linyuan Wu
- Jiangsu Key Laboratory for Biomaterials and Devices, School of Biomedical Engineering, Southeast University, Nanjing, 210009, China
| | - Yuxin Zhang
- Jiangsu Key Laboratory for Biomaterials and Devices, School of Biomedical Engineering, Southeast University, Nanjing, 210009, China
| | - Fengfeng Zhao
- Center of Clinical Laboratory Medicine, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, 210009, China
| | - Jingyi Sheng
- Jiangsu Key Laboratory for Biomaterials and Devices, School of Biomedical Engineering, Southeast University, Nanjing, 210009, China
| | - Ning Gu
- Jiangsu Key Laboratory for Biomaterials and Devices, School of Biomedical Engineering, Southeast University, Nanjing, 210009, China
- Medical School, Nanjing University, Nanjing, 210093, China
| |
Collapse
|
12
|
Abbaszadeh F, Javadpour P, Mousavi Nasab MM, Jorjani M. The Role of Vitamins in Spinal Cord Injury: Mechanisms and Benefits. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2024; 2024:4293391. [PMID: 38938696 PMCID: PMC11211004 DOI: 10.1155/2024/4293391] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Revised: 03/18/2024] [Accepted: 06/06/2024] [Indexed: 06/29/2024]
Abstract
Spinal cord injury (SCI) is a common neurological disease worldwide, often resulting in a substantial decrease in quality of life, disability, and in severe cases, even death. Unfortunately, there is currently no effective treatment for this disease. Nevertheless, current basic and clinical evidence suggests that vitamins, with their antioxidant properties and biological functions, may play a valuable role in improving the quality of life for individuals with SCI. They can promote overall health and facilitate the healing process. In this review, we discuss the mechanisms and therapeutic potential of vitamins in the treatment of SCI.
Collapse
Affiliation(s)
- Fatemeh Abbaszadeh
- Neurobiology Research CenterShahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Pegah Javadpour
- Neuroscience Research CenterShahid Beheshti University of Medical Sciences, Tehran, Iran
| | | | - Masoumeh Jorjani
- Neurobiology Research CenterShahid Beheshti University of Medical Sciences, Tehran, Iran
- Department of PharmacologySchool of MedicineShahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
13
|
Wang C, Xiang Y, Shao Y, Li C. Ferroptosis resists intracellular Vibrio splendidus AJ01 mediated by ferroportin in sea cucumber Apostichopus japonicus. FISH & SHELLFISH IMMUNOLOGY 2024; 149:109585. [PMID: 38663462 DOI: 10.1016/j.fsi.2024.109585] [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: 02/29/2024] [Revised: 04/16/2024] [Accepted: 04/23/2024] [Indexed: 04/29/2024]
Abstract
Ferroptosis, a kind of programmed cell death, is characterized with iron-dependent lipid ROS buildup, which is considered as an important cellular immunity in resisting intracellular bacterial infection in mammalian macrophages. In this process, lipid ROS oxidizes the bacterial biofilm to inhibit intracellular bacteria. However, the function of ferroptosis in invertebrate remains unknown. In this study, the existence of ferroptosis in Apostichopus japonicus coelomocytes was confirmed, and its antibacterial mechanism was investigated. First, our results indicated that the expression of glutathione peroxidase (AjGPX4) was significantly inhibited by 0.21-fold (p < 0.01) after injecting A. japonicus with the ferroptosis inducer RSL3, and the contents of MDA (3.93-fold, p < 0.01), ferrous iron (1.40-fold, p < 0.01), and lipid ROS (3.10-fold, p < 0.01) were all significantly increased under this condition and simultaneously accompanied with mitochondrial contraction and disappearance of cristae, indicating the existence of ferroptosis in the coelomocytes of A. japonicus. Subsequently, the contents of ferrous iron (1.40-fold, p < 0.05), MDA (2.10-fold, p < 0.01), ROS (1.70-fold, p < 0.01), and lipid ROS (2.50-fold, p < 0.01) were all significantly increased, whereas the mitochondrial membrane potential and GSH/GSSG were markedly decreased by 0.68-fold (p < 0.05) and 0.69-fold (p < 0.01) under Vibrio splendidus (AJ01) infection. This process could be reversed by the iron-chelating agent deferoxamine mesylate, which indicated that AJ01 could induce coelomocytic ferroptosis. Moreover, the results demonstrated that the intracellular AJ01 load was clearly decreased to 0.49-fold (p < 0.05) and 0.06-fold (p < 0.01) after treating coelomocytes with RSL3 and ferrous iron, which indicated that enhanced ferroptosis could inhibit bacterial growth. Finally, subcellular localization demonstrated that ferrous iron efflux protein ferroportin (AjFPN) and intracellular AJ01 were co-localized in coelomocytes. After AjFPN interference (0.58-fold, p < 0.01), the signals of ferrous iron and lipid ROS levels in intracellular AJ01 were significantly reduced by 0.38-fold (p < 0.01) and 0.48-fold (p < 0.01), indicating that AjFPN was an important factor in the introduction of ferroptosis into intracellular bacteria. Overall, our findings indicated that ferroptosis could resist intracellular AJ01 infection via AjFPN. These findings provide a novel defense mechanism for aquatic animals against intracellular bacterial infection.
Collapse
Affiliation(s)
- Chengyang Wang
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Ningbo University, Ningbo, PR China
| | - Yangxi Xiang
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Ningbo University, Ningbo, PR China.
| | - Yina Shao
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Ningbo University, Ningbo, PR China.
| | - Chenghua Li
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Ningbo University, Ningbo, PR China; Laboratory for Marine Fisheries Science and Food Production Processes, Qingdao, PR China.
| |
Collapse
|
14
|
Chen J, Ou L, Liu W, Gao F. Exploring the molecular mechanisms of ferroptosis-related genes in periodontitis: a multi-dataset analysis. BMC Oral Health 2024; 24:611. [PMID: 38802844 PMCID: PMC11129485 DOI: 10.1186/s12903-024-04342-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Accepted: 05/07/2024] [Indexed: 05/29/2024] Open
Abstract
PURPOSE This study aims to elucidate the biological functions of ferroptosis-related genes in periodontitis, along with their correlation to tumor microenvironment (TME) features such as immune infiltration. It aims to provide potential diagnostic markers of ferroptosis for clinical management of periodontitis. METHODS Utilizing the periodontitis-related microarray dataset GSE16134 from the Gene Expression Omnibus (GEO) and a set of 528 ferroptosis-related genes identified in prior studies, this research unveils differentially expressed ferroptosis-related genes in periodontitis. Subsequently, a protein-protein interaction network was constructed. Subtyping of periodontitis was explored, followed by validation through immune cell infiltration and gene set enrichment analyses. Two algorithms, randomForest and SVM(Support Vector Machine), were employed to reveal potential ferroptosis diagnostic markers for periodontitis. The diagnostic efficacy, immune correlation, and potential transcriptional regulatory networks of these markers were further assessed. Finally, potential targeted drugs for differentially expressed ferroptosis markers in periodontitis were predicted. RESULTS A total of 36 ferroptosis-related genes (30 upregulated, 6 downregulated) were identified from 829 differentially expressed genes between 9 periodontitis samples and the control group. Subsequent machine learning algorithm screening highlighted 4 key genes: SLC1A5(Solute Carrier Family 1 Member 5), SLC2A14(Solute Carrier Family 1 Member 14), LURAP1L(Leucine Rich Adaptor Protein 1 Like), and HERPUD1(Homocysteine Inducible ER Protein With Ubiquitin Like Domain 1). Exploration of these 4 key genes, supported by time-correlated ROC analysis, demonstrated reliability, while immune infiltration results indicated a strong correlation between key genes and immune factors. Furthermore, Gene Set Enrichment Analysis (GSEA) was conducted for the four key genes, revealing enrichment in GO/KEGG pathways that have a significant impact on periodontitis. Finally, the study predicted potential transcriptional regulatory networks and targeted drugs associated with these key genes in periodontitis. CONCLUSIONS The ferroptosis-related genes identified in this study, including SLC1A5, SLC2A14, LURAP1L, and HERPUD1, may serve as novel diagnostic and therapeutic targets for periodontitis. They are likely involved in the occurrence and development of periodontitis through mechanisms such as immune infiltration, cellular metabolism, and inflammatory chemotaxis, potentially linking the ferroptosis pathway to the progression of periodontitis. Targeted drugs such as flurofamide, L-733060, memantine, tetrabenazine, and WAY-213613 hold promise for potential therapeutic interventions in periodontitis associated with these ferroptosis-related genes.
Collapse
Affiliation(s)
- Jili Chen
- Department of Periodontics, Panyu Branch, Stomatological Hospital, School of Stomatology, Southern Medical University, No.366 Jiangnan Dadao Nan, Haizhu District, Guangzhou, Guangdong, 510220, China
| | - Lijia Ou
- Department of Histology and Embryology, Xiangya School of Medicine, Central South University, No. 172 Tongzipo Road, Yuelu District, Changsha, 410006, China
| | - Weizhen Liu
- Department of Periodontics, Panyu Branch, Stomatological Hospital, School of Stomatology, Southern Medical University, No.366 Jiangnan Dadao Nan, Haizhu District, Guangzhou, Guangdong, 510220, China
| | - Feng Gao
- Department of Periodontics, Panyu Branch, Stomatological Hospital, School of Stomatology, Southern Medical University, No.366 Jiangnan Dadao Nan, Haizhu District, Guangzhou, Guangdong, 510220, China.
| |
Collapse
|
15
|
Min JH, Sarlus H, Harris RA. Glycyl-l-histidyl-l-lysine prevents copper- and zinc-induced protein aggregation and central nervous system cell death in vitro. Metallomics 2024; 16:mfae019. [PMID: 38599632 PMCID: PMC11135135 DOI: 10.1093/mtomcs/mfae019] [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: 10/09/2023] [Accepted: 04/09/2024] [Indexed: 04/12/2024]
Abstract
Common features of neurodegenerative diseases are oxidative and inflammatory imbalances as well as the misfolding of proteins. An excess of free metal ions can be pathological and contribute to cell death, but only copper and zinc strongly promote protein aggregation. Herein we demonstrate that the endogenous copper-binding tripeptide glycyl-l-histidyl-l-lysine (GHK) has the ability to bind to and reduce copper redox activity and to prevent copper- and zinc-induced cell death in vitro. In addition, GHK prevents copper- and zinc-induced bovine serum albumin aggregation and reverses aggregation through resolubilizing the protein. We further demonstrate the enhanced toxicity of copper during inflammation and the ability of GHK to attenuate this toxicity. Finally, we investigated the effects of copper on enhancing paraquat toxicity and report a protective effect of GHK. We therefore conclude that GHK has potential as a cytoprotective compound with regard to copper and zinc toxicity, with positive effects on protein solubility and aggregation that warrant further investigation in the treatment of neurodegenerative diseases.
Collapse
Affiliation(s)
- Jin-Hong Min
- Department of Clinical Neuroscience, Karolinska Institutet, Center for Molecular Medicine, Karolinska University Hospital, S-171 76 Stockholm, Sweden
| | - Heela Sarlus
- Department of Clinical Neuroscience, Karolinska Institutet, Center for Molecular Medicine, Karolinska University Hospital, S-171 76 Stockholm, Sweden
| | - Robert A Harris
- Department of Clinical Neuroscience, Karolinska Institutet, Center for Molecular Medicine, Karolinska University Hospital, S-171 76 Stockholm, Sweden
| |
Collapse
|
16
|
Christodoulou M, Aspray TJ, Piec I, Fraser WD, Schoenmakers I. Alterations in regulators of the renal-bone axis, inflammation and iron status in older people with early renal impairment and the effect of vitamin D supplementation. Age Ageing 2024; 53:afae096. [PMID: 38770543 PMCID: PMC11106582 DOI: 10.1093/ageing/afae096] [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: 07/25/2023] [Revised: 03/21/2024] [Indexed: 05/22/2024] Open
Abstract
CONTEXT Chronic kidney disease (CKD) leads to alterations in fibroblast growth factor 23 (FGF23) and the renal-bone axis. This may be partly driven by altered inflammation and iron status. Vitamin D supplementation may reduce inflammation. OBJECTIVE AND METHODS Older adults with early CKD (estimated glomerular filtration rate (eGFR) 30-60 ml/min/1.73 m2; CKDG3a/b; n = 35) or normal renal function (eGFR >90 ml/min/1.73 m2; CKDG1; n = 35) received 12,000, 24,000 or 48,000 IU D3/month for 1 year. Markers of the renal-bone axis, inflammation and iron status were investigated pre- and post-supplementation. Predictors of c-terminal and intact FGF23 (cFGF23; iFGF23) were identified by univariate and multivariate regression. RESULTS Pre-supplementation, comparing CKDG3a/b to CKDG1, plasma cFGF23, iFGF23, PTH, sclerostin and TNFα were significantly higher and Klotho, 1,25-dihydroxyvitamin D and iron were lower. Post-supplementation, only cFGF23, 25(OH)D and IL6 differed between groups. The response to supplementation differed between eGFR groups. Only in the CKDG1 group, phosphate decreased, cFGF23, iFGF23 and procollagen type I N-propeptide increased. In the CKDG3a/b group, TNFα significantly decreased, and iron increased. Plasma 25(OH)D and IL10 increased, and carboxy-terminal collagen crosslinks decreased in both groups. In univariate models cFGF23 and iFGF23 were predicted by eGFR and regulators of calcium and phosphate metabolism at both time points; IL6 predicted cFGF23 (post-supplementation) and iFGF23 (pre-supplementation) in univariate models. Hepcidin predicted post-supplementation cFGF23 in multivariate models with eGFR. CONCLUSION Alterations in regulators of the renal-bone axis, inflammation and iron status were found in early CKD. The response to vitamin D3 supplementation differed between eGFR groups. Plasma IL6 predicted both cFGF23 and iFGF23 and hepcidin predicted cFGF23.
Collapse
Affiliation(s)
| | - Terence J Aspray
- Freeman Hospital, Bone Clinic, University of Newcastle upon Tyne, Newcastle upon Tyne, UK
| | - Isabelle Piec
- University of East Anglia, Norwich Medical School, Norwich, UK
| | - William D Fraser
- University of East Anglia, Norwich Medical School, Norwich, UK
- Clinical Biochemistry, Department of Laboratory Medicine and Department of Diabetes and Endocrinology, Norfolk and Norwich University Hospital NHS Foundation Trust, Norwich, UK
| | - Inez Schoenmakers
- University of East Anglia, Norwich Medical School, Norwich, UK
- MRC Human Nutrition Research, Cambridge, UK
| |
Collapse
|
17
|
Yi W, Zhang J, Huang Y, Zhan Q, Zou M, Cheng X, Zhang X, Yin Z, Tao S, Cheng H, Wang F, Guo J, Ju Z, Chen Z. Ferritin-mediated mitochondrial iron homeostasis is essential for the survival of hematopoietic stem cells and leukemic stem cells. Leukemia 2024; 38:1003-1018. [PMID: 38402368 DOI: 10.1038/s41375-024-02169-y] [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: 09/07/2023] [Revised: 01/26/2024] [Accepted: 02/02/2024] [Indexed: 02/26/2024]
Abstract
Iron metabolism plays a crucial role in cell viability, but its relationship with adult stem cells and cancer stem cells is not fully understood. The ferritin complex, responsible for intracellular iron storage, is important in this process. We report that conditional deletion of ferritin heavy chain 1 (Fth1) in the hematopoietic system reduced the number and repopulation capacity of hematopoietic stem cells (HSCs). These effects were associated with a decrease in cellular iron level, leading to impaired mitochondrial function and the initiation of apoptosis. Iron supplementation, antioxidant, and apoptosis inhibitors reversed the reduced cell viability of Fth1-deleted hematopoietic stem and progenitor cells (HSPCs). Importantly, leukemic stem cells (LSCs) derived from MLL-AF9-induced acute myeloid leukemia (AML) mice exhibited reduced Fth1 expression, rendering them more susceptible to apoptosis induced by the iron chelation compared to normal HSPCs. Modulating FTH1 expression using mono-methyl fumarate increased LSCs resistance to iron chelator-induced apoptosis. Additionally, iron supplementation, antioxidant, and apoptosis inhibitors protected LSCs from iron chelator-induced cell death. Fth1 deletion also extended the survival of AML mice. These findings unveil a novel mechanism by which ferritin-mediated iron homeostasis regulates the survival of both HSCs and LSCs, suggesting potential therapeutic strategies for blood cancer with iron dysregulation.
Collapse
Affiliation(s)
- Weiwei Yi
- Department of Cardiology, the First Affiliated Hospital of Jinan University, Guangzhou, 510630, Guangdong, China
- Key Laboratory of Regenerative Medicine of Ministry of Education, Institute of Aging and Regenerative Medicine, College of Life Science and Technology, Jinan University, Guangzhou, 510632, Guangdong, China
| | - Jinhua Zhang
- Department of Cardiology, the First Affiliated Hospital of Jinan University, Guangzhou, 510630, Guangdong, China
| | - Yingxin Huang
- Key Laboratory of Regenerative Medicine of Ministry of Education, Institute of Aging and Regenerative Medicine, College of Life Science and Technology, Jinan University, Guangzhou, 510632, Guangdong, China
| | - Qiang Zhan
- Key Laboratory of Regenerative Medicine of Ministry of Education, Institute of Aging and Regenerative Medicine, College of Life Science and Technology, Jinan University, Guangzhou, 510632, Guangdong, China
| | - Mi Zou
- Key Laboratory of Regenerative Medicine of Ministry of Education, Institute of Aging and Regenerative Medicine, College of Life Science and Technology, Jinan University, Guangzhou, 510632, Guangdong, China
| | - Xiang Cheng
- Department of Hematology, Children's Hospital, Capital Institute of Pediatrics, Beijing, 100020, China
| | - Xuguang Zhang
- Mengniu Institute of Nutrition Science, Global R&D Innovation Center, Shanghai, China
- Shanghai Institute of Nutrition and Health, The Chinese Academy of Sciences, Shanghai, China
| | - Zhinan Yin
- Guangdong Provincial Key Laboratory of Tumor Interventional Diagnosis and Treatment, Institute of Translational Medicine, Zhuhai People's Hospital Affiliated with Jinan University, Jinan University, Zhuhai, 519000, Guangdong, China
- The Biomedical Translational Research Institute, Health Science Center (School of Medicine), Jinan University, Guangzhou, 510632, Guangdong, China
| | - Si Tao
- Department of Oncology, The Second Affiliated Hospital of Nanchang University, Nanchang, 330006, Jiangxi, China
| | - Hui Cheng
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, 300020, China
| | - Fudi Wang
- The Second Affiliated Hospital, School of Public Health, State Key Laboratory of Experimental Hematology, Zhejiang University School of Medicine, Hangzhou, 310058, Zhejiang, China
- The First Affiliated Hospital, Basic Medical Sciences, School of Public Health, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China
| | - Jun Guo
- Department of Cardiology, the First Affiliated Hospital of Jinan University, Guangzhou, 510630, Guangdong, China.
| | - Zhenyu Ju
- Key Laboratory of Regenerative Medicine of Ministry of Education, Institute of Aging and Regenerative Medicine, College of Life Science and Technology, Jinan University, Guangzhou, 510632, Guangdong, China.
| | - Zhiyang Chen
- Key Laboratory of Regenerative Medicine of Ministry of Education, Institute of Aging and Regenerative Medicine, College of Life Science and Technology, Jinan University, Guangzhou, 510632, Guangdong, China.
| |
Collapse
|
18
|
Fang Z, Wang C, Zhu J, Gou Y. Iron overload promotes hemochromatosis-associated osteoarthritis via the mTORC1-p70S6K/4E-BP1 pathway. Int Immunopharmacol 2024; 131:111848. [PMID: 38479156 DOI: 10.1016/j.intimp.2024.111848] [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: 01/20/2024] [Revised: 02/23/2024] [Accepted: 03/09/2024] [Indexed: 04/10/2024]
Abstract
BACKGROUNDS Joint iron overload in hemochromatosis induces M1 polarization in synovial macrophages, releasing pro-inflammatory factors and leading to osteoarthritis development. However, the mechanism by which iron overload regulates M1 polarization remains unclear. This study aims to elucidate the mechanism by which synovial iron overload promotes macrophage M1 polarization. METHODS In vitro, RAW264.7 macrophages were treated with iron and divided into five groups based on the concentration of the iron chelator, desferrioxamine (DFO): Ctrl, Fe, DFO1, DFO2, and DFO3. In vivo, rats were categorized into five groups based on iron overload and intra-articular DFO injection: A-Ctrl, A-Fe, A-DFO1, A-DFO2, and A-DFO3. Osteoarthritis was induced by transecting the left knee anterior cruciate ligament. Macrophage morphology was observed; Prussian Blue staining quantified iron deposition in macrophages, synovium, and liver; serum iron concentration was measured using the ferrozine method; cartilage damage was assessed using H&E and Safranin O-Fast Green staining; qPCR detected iNOS and Arg-1 expression; Western Blot analyzed the protein expression of iNOS, Arg-1, 4E-BP1, phosphorylated 4E-BP1, p70S6K, and phosphorylated p70S6K; ELISA measured TNF-α and IL-6 concentrations in supernatants; and immunohistochemistry examined the protein expression of F4/80, iNOS, Arg-1, 4E-BP1, phosphorylated 4E-BP1, p70S6K, and phosphorylated p70S6K in the synovium. RESULTS In vitro, iron-treated macrophages exhibited Prussian Blue staining indicative of iron overload and morphological changes towards M1 polarization. qPCR and Western Blot revealed increased expression of the M1 polarization markers iNOS and its protein. ELISA showed elevated TNF-α and IL-6 levels in supernatants. In vivo, ferrozine assay indicated significantly increased serum iron concentrations in all groups except A-Ctrl; Prussian Blue staining showed increased liver iron deposition in all groups except A-Ctrl. Iron deposition in rat synovium decreased in a DFO concentration-dependent manner; immunohistochemistry showed a corresponding decrease in iNOS and phosphorylated 4E-BP1 expression, and an increase in Arg-1 expression. CONCLUSION Intracellular iron overload may exacerbate joint cartilage damage by promoting synovial macrophage M1 polarization through phosphorylation of 4E-BP1 in the mTORC1-p70S6K/4E-BP1 pathway.
Collapse
Affiliation(s)
- Zhiyuan Fang
- Department of Orthopaedic Surgery, The Third Affiliated Hospital of Xinjiang Medical University, Xinjiang Medical University, Urumqi 830000, China.
| | - Chengwei Wang
- Department of Orthopaedic Surgery, The Third Affiliated Hospital of Xinjiang Medical University, Xinjiang Medical University, Urumqi 830000, China.
| | - Jiang Zhu
- General Surgery department, Prevention and Treatment of High Incidence Diseases in Central Asia, Clinical Medicine Institute, First Teaching Hospital of Xinjiang Medical University, Urumqi 830011, China.
| | - Yangyang Gou
- The Sixth Affiliated Hospital of Xinjiang Medical University, Xinjiang Medical University, Urumqi 830000, China.
| |
Collapse
|
19
|
Xia Y, Chen Z, Huang C, Shi L, Ma W, Chen X, Liu Y, Wang Y, Cai C, Huang Y, Liu W, Shi R, Luo Q. Investigation the mechanism of iron overload-induced colonic inflammation following ferric citrate exposure. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2024; 275:116241. [PMID: 38522287 DOI: 10.1016/j.ecoenv.2024.116241] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/21/2024] [Revised: 03/06/2024] [Accepted: 03/18/2024] [Indexed: 03/26/2024]
Abstract
Iron overload occurs due to excessive iron intake compared to the body's demand, leading to iron deposition and impairment of multiple organ functions. Our previous study demonstrated that chronic oral administration of ferric citrate (FC) caused colonic inflammatory injury. However, the precise mechanism underlying this inflammatory response remains unclear. The current study aims to investigate the mechanism by which iron overload induced by FC exposure leads to colonic inflammation. To accomplish this, mice were orally exposed to three different concentrations of FC (71 mg/kg/bw (L), 143 mg/kg/bw (M) and 286 mg/kg/bw (H)) for continuous 16 weeks, with the control group receiving ultrapure water (C). Exposure to FC caused disturbances in the excretory system, altered colonic flora alpha diversity, and enriched pathogenic bacteria, such as Mucispirillum, Helicobacter, Desulfovibrio, and Shigella. These changes led to structural disorders of the colonic flora and an inflammatory response phenotype characterized by inflammatory cells infiltration, atrophy of intestinal glands, and irregular thickening of the intestinal wall. Mechanistic studies revealed that FC-exposure activated the NF-κB signaling pathway by up-regulating TLR4, MyD88, and NF-κB mRNA levels and protein expression. This activation resulted in increased production of pro-inflammatory cytokines, further contributing to the colonic inflammation. Additionally, in vitro experiments in SW480 cells confirmed the activation of NF-κB signaling pathway by FC exposure, consistent with the in vivo findings. The significance of this study lies in its elucidation of the mechanism by which iron overload caused by FC exposure leads to colonic inflammation. By identifying the role of pathogenic bacteria and the NF-κB signaling pathway, this study could potentially offer a crucial theoretical foundation for the research on iron overload, as well as provide valuable insights for clinical iron supplementation.
Collapse
Affiliation(s)
- Yu Xia
- Laboratory of Experimental Animal Disease Model, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China; Animal Disease Prevention and Control and Healthy Breeding Engineering Technology Research Centre, Mianyang Normal University, Mianyang 621000, China
| | - Zhengli Chen
- Laboratory of Experimental Animal Disease Model, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China
| | - Chao Huang
- Laboratory of Experimental Animal Disease Model, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China
| | - Liangqin Shi
- Chengdu University of Traditional Chinese Medicine, College of Basic Medicine, Chengdu 611130, China
| | - Wenjing Ma
- Laboratory of Experimental Animal Disease Model, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China
| | - Xiwen Chen
- Animal Disease Prevention and Control and Healthy Breeding Engineering Technology Research Centre, Mianyang Normal University, Mianyang 621000, China
| | - Yucong Liu
- Laboratory of Experimental Animal Disease Model, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China
| | - Yao Wang
- Laboratory of Experimental Animal Disease Model, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China
| | - Chunyu Cai
- Laboratory of Experimental Animal Disease Model, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China
| | - Yixiang Huang
- Laboratory of Experimental Animal Disease Model, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China
| | - Wentao Liu
- Laboratory of Experimental Animal Disease Model, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China
| | - Riyi Shi
- Department of Basic Medical Sciences, Center for Paralysis Research, College of Veterinary Medicine, Purdue University, West Lafayette, IN 47907, USA
| | - Qihui Luo
- Laboratory of Experimental Animal Disease Model, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China.
| |
Collapse
|
20
|
Martínez-Quintana E, Alcántara-Castellano M, García-Suárez MI, Rodríguez-González F. C-Reactive Protein and Long-Term Prognosis in Adult Patients with Congenital Heart Disease. J Clin Med 2024; 13:2199. [PMID: 38673472 PMCID: PMC11050825 DOI: 10.3390/jcm13082199] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2024] [Revised: 03/21/2024] [Accepted: 04/07/2024] [Indexed: 04/28/2024] Open
Abstract
Background/Objectives: Prognostic biomarkers may provide information about the patient's cardiovascular outcomes. However, there are doubts regarding how high-sensitivity C-reactive protein (hs-CRP) impacts patients with congenital heart disease (CHD). The main objective is to evaluate whether high hs-CRP levels predict a worse prognosis in patients with CHD. Methods: Observational and prospective cohort study. Adult CHD patients and controls were matched for age and sex. Results: In total, 434 CHD patients (cases) and 820 controls were studied. The median age in the CHD patients was 30 (18-62) years and 256 (59%) were male. A total of 51%, 30%, and 19% of patients with CHD had mild, moderate, and great complexity defects, respectively. The body mass index [1.07 (1.01-1.13), p = 0.022)], diabetes mellitus [3.57 (1.07-11.97), p = 0.039], high NT-pro-BNP levels [1.00 (1.00-1.01), p = 0.021], and low serum iron concentrations [0.98 (0.97-0.99), p = 0.001] predicted high hs-CRP levels (≥0.3 mg/dL) in patients with CHD. During a follow-up time of 6.81 (1.17-10.46) years, major cardiovascular events (MACE) occurred in 40 CHD patients, showing the Kaplan-Meier test demonstrated a worse outcome among patients with hs-CRP levels above 0.3 mg/dL (p = 0.012). Also, hs-CRP showed statistical significance in the univariate Cox regression survival analysis. However, after adjusting for other variables, this significance was lost and the remaining predictors of MACE were age [HR 1.03 (1.01-1.06), p = 0.001], great complexity defects [HR 2.46 (1.07-5.69), p = 0.035], and an NT pro-BNP cutoff value for heart failure > 125 pg/mL [HR 7.73 (2.54-23.5), p < 0.001]. Conclusions: Hs-CRP obtained statistical significance in the univariate survival analysis. However, this significance was lost in the multivariate analysis in favor of age, CHD complexity, and heart failure.
Collapse
Affiliation(s)
- Efrén Martínez-Quintana
- Cardiology Service, Complejo Hospitalario Universitario Insular-Materno Infantil, Avd. Marítima del Sur s/n, 35016 Las Palmas de Gran Canaria, Spain
- Department of Medical and Surgical Sciences, Faculty of Health Sciences, Universidad de Las Palmas de Gran Canaria, 35016 Las Palmas de Gran Canaria, Spain
| | - María Alcántara-Castellano
- Department of Medical and Surgical Sciences, Faculty of Health Sciences, Universidad de Las Palmas de Gran Canaria, 35016 Las Palmas de Gran Canaria, Spain
| | - Marta Isabel García-Suárez
- Department of Medical and Surgical Sciences, Faculty of Health Sciences, Universidad de Las Palmas de Gran Canaria, 35016 Las Palmas de Gran Canaria, Spain
| | | |
Collapse
|
21
|
Iddrisu I, Monteagudo-Mera A, Poveda C, Shahzad M, Walton GE, Andrews SC. A review of the effect of iron supplementation on the gut microbiota of children in developing countries and the impact of prebiotics. Nutr Res Rev 2024:1-9. [PMID: 38586996 DOI: 10.1017/s0954422424000118] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/09/2024]
Abstract
Iron is essential for many physiological functions of the body, and it is required for normal growth and development. Iron deficiency (ID) is the most common form of micronutrient malnutrition and is particularly prevalent in infants and young children in developing countries. Iron supplementation is considered the most effective strategy to combat the risk of ID and ID anaemia (IDA) in infants, although iron supplements cause a range of deleterious gut-related problems in malnourished children. The purpose of this review is to assess the available evidence on the effect of iron supplementation on the gut microbiota during childhood ID and to further assess whether prebiotics offer any benefits for iron supplementation. Prebiotics are well known to improve gut-microbial health in children, and recent reports indicate that prebiotics can mitigate the adverse gut-related effects of iron supplementation in children with ID and IDA. Thus, provision of prebiotics alongside iron supplements has the potential for an enhanced strategy for combatting ID and IDA among children in the developing world. However, further understanding is required before the benefit of such combined treatments of ID in nutritionally deprived children across populations can be fully confirmed. Such enhanced understanding is of high relevance in resource-poor countries where ID, poor sanitation and hygiene, alongside inadequate access to good drinking water and poor health systems, are serious public health concerns.
Collapse
Affiliation(s)
- Ishawu Iddrisu
- Rose Ward, Prospect Park Hospital, Berkshire Healthcare NHS Foundation Trust, Reading, RG30 4EJ, UK
- Department of Food and Nutritional Sciences, University of Reading, Whiteknights, Reading, RG6 6AP, UK
- School of Biological Sciences, University of Reading, Whiteknights, Reading, RG6 6EX, UK
| | - Andrea Monteagudo-Mera
- Department of Food and Nutritional Sciences, University of Reading, Whiteknights, Reading, RG6 6AP, UK
| | - Carlos Poveda
- Department of Food and Nutritional Sciences, University of Reading, Whiteknights, Reading, RG6 6AP, UK
| | - Muhammed Shahzad
- Institute of Basic Medical Sciences, Khyber Medical University, Peshawar, Pakistan
- Faculty of Dentistry, Zarqa University, Zarqa, 13110, Jordan
| | - Gemma E Walton
- Department of Food and Nutritional Sciences, University of Reading, Whiteknights, Reading, RG6 6AP, UK
| | - Simon C Andrews
- School of Biological Sciences, University of Reading, Whiteknights, Reading, RG6 6EX, UK
| |
Collapse
|
22
|
Liu P, Lv M, Rong Y, Yu S, Wu R. No genetic causal association between iron status and pulmonary artery hypertension: Insights from a two-sample Mendelian randomization. Pulm Circ 2024; 14:e12370. [PMID: 38774814 PMCID: PMC11108639 DOI: 10.1002/pul2.12370] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/17/2024] [Revised: 04/08/2024] [Accepted: 04/09/2024] [Indexed: 05/24/2024] Open
Abstract
To explore the genetic causal association between pulmonary artery hypertension (PAH) and iron status through Mendelian randomization (MR), we conducted MR analysis using publicly available genome-wide association study (GWAS) summary data. Five indicators related to iron status (serum iron, ferritin, total iron binding capacity (TIBC), soluble transferrin receptor (sTfR), and transferrin saturation) served as exposures, while PAH was the outcome. The genetic causal association between these iron status indicators and PAH was assessed using the inverse variance weighted (IVW) method. Cochran's Q statistic was employed to evaluate heterogeneity. We assessed pleiotropy using MR-Egger regression and MR-Presso test. Additionally, we validated our results using the Weighted median, Simple mode, and Weighted mode methods. Based on the IVW method, we found no causal association between iron status (serum iron, ferritin, TIBC, sTfR, and transferrin saturation) and PAH (p β > 0.05). The Weighted median, Simple mode, and Weighted mode methods showed no potential genetic causal association (p β > 0.05 in the three analyses). Additionally, no heterogeneity or horizontal pleiotropy was detected in any of the analyses. Our results show that there are no genetic causal association between iron status and PAH.
Collapse
Affiliation(s)
- Peng‐Cheng Liu
- Department of Rheumatology, The First Affiliated Hospital, Jiangxi Medical CollegeNanchang UniversityNanchangChina
| | - Meng‐Na Lv
- The First Clinical Medical College of Nanchang UniversityNanchangChina
| | - Yan‐Yan Rong
- Department of Hematology, The First Affiliated Hospital, Jiangxi Medical CollegeNanchang UniversityNanchangChina
| | - Shu‐Jiao Yu
- Department of Rheumatology, The First Affiliated Hospital, Jiangxi Medical CollegeNanchang UniversityNanchangChina
| | - Rui Wu
- Department of Rheumatology, The First Affiliated Hospital, Jiangxi Medical CollegeNanchang UniversityNanchangChina
| |
Collapse
|
23
|
Lewińska A, Radoń A, Gil K, Błoniarz D, Ciuraszkiewicz A, Kubacki J, Kądziołka-Gaweł M, Łukowiec D, Gębara P, Krogul-Sobczak A, Piotrowski P, Fijałkowska O, Wybraniec S, Szmatoła T, Kolano-Burian A, Wnuk M. Carbon-Coated Iron Oxide Nanoparticles Promote Reductive Stress-Mediated Cytotoxic Autophagy in Drug-Induced Senescent Breast Cancer Cells. ACS APPLIED MATERIALS & INTERFACES 2024; 16:15457-15478. [PMID: 38483821 PMCID: PMC10982943 DOI: 10.1021/acsami.3c17418] [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: 11/20/2023] [Revised: 02/27/2024] [Accepted: 02/27/2024] [Indexed: 04/04/2024]
Abstract
The surface modification of magnetite nanoparticles (Fe3O4 NPs) is a promising approach to obtaining biocompatible and multifunctional nanoplatforms with numerous applications in biomedicine, for example, to fight cancer. However, little is known about the effects of Fe3O4 NP-associated reductive stress against cancer cells, especially against chemotherapy-induced drug-resistant senescent cancer cells. In the present study, Fe3O4 NPs in situ coated by dextran (Fe3O4@Dex) and glucosamine-based amorphous carbon coating (Fe3O4@aC) with potent reductive activity were characterized and tested against drug-induced senescent breast cancer cells (Hs 578T, BT-20, MDA-MB-468, and MDA-MB-175-VII cells). Fe3O4@aC caused a decrease in reactive oxygen species (ROS) production and an increase in the levels of antioxidant proteins FOXO3a, SOD1, and GPX4 that was accompanied by elevated levels of cell cycle inhibitors (p21, p27, and p57), proinflammatory (NFκB, IL-6, and IL-8) and autophagic (BECN1, LC3B) markers, nucleolar stress, and subsequent apoptotic cell death in etoposide-stimulated senescent breast cancer cells. Fe3O4@aC also promoted reductive stress-mediated cytotoxicity in nonsenescent breast cancer cells. We postulate that Fe3O4 NPs, in addition to their well-established hyperthermia and oxidative stress-mediated anticancer effects, can also be considered, if modified using amorphous carbon coating with reductive activity, as stimulators of reductive stress and cytotoxic effects in both senescent and nonsenescent breast cancer cells with different gene mutation statuses.
Collapse
Affiliation(s)
- Anna Lewińska
- Institute
of Biotechnology, College of Natural Sciences, University of Rzeszow, Pigonia 1, 35-310 Rzeszow, Poland
| | - Adrian Radoń
- Łukasiewicz
Research Network—Institute of Non-Ferrous Metals, Sowińskiego 5, 44-100 Gliwice, Poland
| | - Kacper Gil
- Institute
of Biotechnology, College of Natural Sciences, University of Rzeszow, Pigonia 1, 35-310 Rzeszow, Poland
| | - Dominika Błoniarz
- Institute
of Biotechnology, College of Natural Sciences, University of Rzeszow, Pigonia 1, 35-310 Rzeszow, Poland
| | - Agnieszka Ciuraszkiewicz
- Łukasiewicz
Research Network—Institute of Non-Ferrous Metals, Sowińskiego 5, 44-100 Gliwice, Poland
| | - Jerzy Kubacki
- Institute
of Physics, Faculty of Science and Technology, University of Silesia in Katowice, 75 Pułku Piechoty 1, 41-500 Chorzów, Poland
| | - Mariola Kądziołka-Gaweł
- Institute
of Physics, Faculty of Science and Technology, University of Silesia in Katowice, 75 Pułku Piechoty 1, 41-500 Chorzów, Poland
| | - Dariusz Łukowiec
- Faculty
of Mechanical Engineering, Silesian University of Technology, Konarskiego 18A, 44-100 Gliwice, Poland
| | - Piotr Gębara
- Department
of Physics, Częstochowa University
of Technology, Armii Krajowej 19, 42-200 Częstochowa, Poland
| | | | - Piotr Piotrowski
- Faculty
of
Chemistry, University of Warsaw, Pasteura 1, 02-093 Warsaw, Poland
| | - Oktawia Fijałkowska
- Institute
of Biotechnology, College of Natural Sciences, University of Rzeszow, Pigonia 1, 35-310 Rzeszow, Poland
| | - Sylwia Wybraniec
- Institute
of Biotechnology, College of Natural Sciences, University of Rzeszow, Pigonia 1, 35-310 Rzeszow, Poland
| | - Tomasz Szmatoła
- Center
of Experimental and Innovative Medicine, University of Agriculture in Krakow, Mickiewicza 24/28, 30-059 Krakow, Poland
| | - Aleksandra Kolano-Burian
- Łukasiewicz
Research Network—Institute of Non-Ferrous Metals, Sowińskiego 5, 44-100 Gliwice, Poland
| | - Maciej Wnuk
- Institute
of Biotechnology, College of Natural Sciences, University of Rzeszow, Pigonia 1, 35-310 Rzeszow, Poland
| |
Collapse
|
24
|
Volk Robertson K, Schleh MW, Harrison FE, Hasty AH. Microglial-specific knockdown of iron import gene, Slc11a2, blunts LPS-induced neuroinflammatory responses in a sex-specific manner. Brain Behav Immun 2024; 116:370-384. [PMID: 38141840 PMCID: PMC10874246 DOI: 10.1016/j.bbi.2023.12.020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Revised: 12/07/2023] [Accepted: 12/18/2023] [Indexed: 12/25/2023] Open
Abstract
Neuroinflammation and microglial iron load are significant hallmarks found in several neurodegenerative diseases. In in vitro systems, microglia preferentially upregulate the iron importer, divalent metal transporter 1 (DMT1, gene name Slc11a2) in response to inflammatory stimuli, and it has been shown that iron can augment cellular inflammation, suggesting a feed-forward loop between mechanisms involved in iron import and inflammatory signaling. However, it is not understood how microglial iron import mechanisms contribute to inflammation in vivo, or whether altering a microglial iron-related gene affects the inflammatory response. These studies aimed to determine the effect of knocking down microglial iron import gene Slc11a2 on the inflammatory response in vivo. We generated a novel model of tamoxifen-inducible, microglial-specific Slc11a2 knockdown using Cx3cr1Cre-ERT2 mice. Transgenic male and female mice were administered intraperitoneal saline or lipopolysaccharide (LPS) and assessed for sickness behavior post-injection. Plasma cytokines and microglial bulk RNA sequencing (RNASeq) analyses were performed at 4 h post-LPS, and microglia were collected for gene expression analysis after 24 h. A subset of mice was assessed in a behavioral test battery following LPS-induced sickness recovery. Control male, but not female, mice significantly upregulated microglial Slc11a2 at 4 and 24 h following LPS. In Slc11a2 knockdown mice, we observed an improvement in the acute behavioral sickness response post-LPS in male, but not female, animals. Microglia from male, but not female, knockdown animals exhibited a significant decrease in LPS-provoked pro-inflammatory cytokine expression after 24 h. RNASeq data from male knockdown microglia 4 h post-LPS revealed a robust downregulation in inflammatory genes including Il6, Tnfα, and Il1β, and an increase in anti-inflammatory and homeostatic markers (e.g., Tgfbr1, Cx3cr1, and Trem2). This corresponded with a profound decrease in plasma pro-inflammatory cytokines 4 h post-LPS. At 4 h, male knockdown microglia also upregulated expression of markers of iron export, iron recycling, and iron homeostasis and decreased iron storage and import genes, along with pro-oxidant markers such as Cybb, Nos2, and Hif1α. Overall, this work elucidates how manipulating a specific gene involved in iron import in microglia alters acute inflammatory signaling and overall cell activation state in male mice. These data highlight a sex-specific link between a microglial iron import gene and the pro-inflammatory response to LPS in vivo, providing further insight into the mechanisms driving neuroinflammatory disease.
Collapse
Affiliation(s)
- Katrina Volk Robertson
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, USA
| | - Michael W Schleh
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, USA
| | - Fiona E Harrison
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA.
| | - Alyssa H Hasty
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, USA; VA Tennessee Valley Healthcare System, Nashville, TN, USA.
| |
Collapse
|
25
|
Yang XY, An JR, Dong Q, Gou YJ, Jia CL, Song JX, Tan M, Sun MF, Li BL, Zhang Z, Ji ES, Zhao Y. Banxia-Houpu decoction inhibits iron overload and chronic intermittent hypoxia-induced neuroinflammation in mice. JOURNAL OF ETHNOPHARMACOLOGY 2024; 318:117078. [PMID: 37625604 DOI: 10.1016/j.jep.2023.117078] [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: 06/28/2023] [Revised: 08/18/2023] [Accepted: 08/22/2023] [Indexed: 08/27/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE The Banxia-Houpu decoction (BHD), a renowned prescription documented in the Chinese medical book "The Synopsis of the Golden Chamber," has been proven to effectively mitigate inflammation within the central nervous system. Previous studies have demonstrated the efficacy of BHD in ameliorating symptoms in patients with obstructive sleep apnea (OSA). Nevertheless, the precise mechanisms and comprehensive effects of BHD on central system injury in OSA models have not been fully investigated. AIM OF THE STUDY To investigate whether BHD could inhibit neuroinflammation to decrease iron-induced neurotoxic injury in CIH mice. MATERIALS AND METHODS C57BL/6N mice were divided into the Con, CIH, and BHD groups. Mice were exposed to CIH (21%-5% FiO2, 3 min/cycle, 8 h/d), and BHD was administered by gavage (3.51, 7.01, and 14.02 g/kg). The polarization of microglia, inflammatory factors, hepcidin, and brain iron levels were determined. RESULTS The administration of BHD at a dosage of 7.01 g/kg demonstrated a significant reduction in neurobehavioral abnormalities, neuronal damage, and degeneration caused by CIH. BHD exhibited the ability to inhibit the transition of microglial polarization from M2 to M1 by upregulating CD163 expression and downregulating iNOS levels. Furthermore, BHD decreased pro-inflammatory factor levels and increased anti-inflammatory factor levels. Additionally, BHD was found to decrease hepcidin expression in astrocytes through the TLR4/MyD88/NF-κB signaling pathway. BHD reduced the total and neuronal iron levels by elevating FPN1 and reducing TfR1 levels. BHD exhibited positive effects on synapse and synaptic spine abnormalities, as well as an increase in the Bcl-2/Bax ratio, thereby mitigating neuronal damage induced by CIH. CONCLUSIONS Based on these findings, BHD holds potential as a therapeutic intervention for neural damage injuries, which offers a theoretical foundation for the treatment of patients with OSA in clinical.
Collapse
Affiliation(s)
- Xin-Yue Yang
- Hebei Technology Innovation Center of TCM Combined Hydrogen Medicine, Hebei University of Chinese Medicine, Shijiazhuang, 050200, China
| | - Ji-Ren An
- Hebei Technology Innovation Center of TCM Combined Hydrogen Medicine, Hebei University of Chinese Medicine, Shijiazhuang, 050200, China; Liaoning University of Traditional Chinese Medicine, Shenyang, 110847, China
| | - QianBo Dong
- The Second Hospital of Hebei Medical University, Shijiazhuang, 050004, China
| | - Yu-Jing Gou
- Hebei Technology Innovation Center of TCM Combined Hydrogen Medicine, Hebei University of Chinese Medicine, Shijiazhuang, 050200, China
| | - Cui-Ling Jia
- Hebei Technology Innovation Center of TCM Combined Hydrogen Medicine, Hebei University of Chinese Medicine, Shijiazhuang, 050200, China
| | - Ji-Xian Song
- Hebei Technology Innovation Center of TCM Combined Hydrogen Medicine, Hebei University of Chinese Medicine, Shijiazhuang, 050200, China
| | - Miao Tan
- The Fourth Affiliated Hospital of Hebei Medical University, Shijiazhuang, 050011, China
| | - Meng-Fan Sun
- Hebei Technology Innovation Center of TCM Combined Hydrogen Medicine, Hebei University of Chinese Medicine, Shijiazhuang, 050200, China
| | - Bo-Liang Li
- Hebei Technology Innovation Center of TCM Combined Hydrogen Medicine, Hebei University of Chinese Medicine, Shijiazhuang, 050200, China
| | - Zhi Zhang
- Hebei Technology Innovation Center of TCM Combined Hydrogen Medicine, Hebei University of Chinese Medicine, Shijiazhuang, 050200, China
| | - En-Sheng Ji
- Hebei Technology Innovation Center of TCM Combined Hydrogen Medicine, Hebei University of Chinese Medicine, Shijiazhuang, 050200, China.
| | - Yashuo Zhao
- Hebei Technology Innovation Center of TCM Combined Hydrogen Medicine, Hebei University of Chinese Medicine, Shijiazhuang, 050200, China.
| |
Collapse
|
26
|
Khalil S, Cavagnero KJ, Williams MR, O'Neill A, Nakatsuji T, Gallo RL. Regulation of Epidermal Ferritin Expression Influences Systemic Iron Homeostasis. J Invest Dermatol 2024; 144:84-95.e3. [PMID: 37544587 DOI: 10.1016/j.jid.2023.07.009] [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: 10/12/2022] [Revised: 05/31/2023] [Accepted: 07/03/2023] [Indexed: 08/08/2023]
Abstract
Absorption of dietary iron is largely regulated by the liver hormone hepcidin, which is released under conditions of iron overload and inflammation. Although hepcidin-dependent regulation of iron uptake and circulation is well-characterized, recent studies have suggested that the skin may play an important role in iron homeostasis, including transferrin receptor-mediated epidermal iron uptake and direct hepcidin production by keratinocytes. In this study, we characterized direct keratinocyte responses to conditions of high and low iron. We observed potent iron storage capacity by keratinocytes in vitro and in vivo and the effects of iron on epidermal differentiation and gene expression associated with inflammation and barrier function. In mice, systemic iron was observed to be coupled to epidermal iron content. Furthermore, topical inflammation, as opposed to systemic inflammation, resulted in a primary iron-deficiency phenotype associated with low liver hepcidin. These studies suggest a role for keratinocytes and epidermal iron storage as regulators of iron homeostasis with direct contribution by the cutaneous inflammatory state.
Collapse
Affiliation(s)
- Shadi Khalil
- Department of Dermatology, University of California San Diego, La Jolla, California, USA
| | - Kellen J Cavagnero
- Department of Dermatology, University of California San Diego, La Jolla, California, USA
| | - Michael R Williams
- Department of Dermatology, University of California San Diego, La Jolla, California, USA
| | - Alan O'Neill
- Department of Dermatology, University of California San Diego, La Jolla, California, USA
| | - Teruaki Nakatsuji
- Department of Dermatology, University of California San Diego, La Jolla, California, USA
| | - Richard L Gallo
- Department of Dermatology, University of California San Diego, La Jolla, California, USA.
| |
Collapse
|
27
|
Muranov KO. Fenton Reaction in vivo and in vitro. Possibilities and Limitations. BIOCHEMISTRY. BIOKHIMIIA 2024; 89:S112-S126. [PMID: 38621747 DOI: 10.1134/s0006297924140074] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/03/2023] [Revised: 10/04/2023] [Accepted: 10/06/2023] [Indexed: 04/17/2024]
Abstract
The review considers the problem of hydrogen peroxide decomposition and hydroxyl radical formation in the presence of iron in vivo and in vitro. Analysis of the literature data allows us to conclude that, under physiological conditions, transport of iron, carried out with the help of carrier proteins, minimizes the possibility of appearance of free iron ions in cytoplasm of the cell. Under pathological conditions, when the process of transferring an iron ion from a donor protein to an acceptor protein can be disrupted due to modifications of the carrier proteins, iron ions can enter cytosol. However, at pH values close to neutral, which is typical for cytosol, iron ions are converted into water-insoluble hydroxides. This makes it impossible to decompose hydrogen peroxide according to the mechanism of the classical Fenton reaction. A similar situation is observed in vitro, since buffers with pH close to neutral are used to simulate free radical oxidation. At the same time, iron hydroxides are able to catalyze decomposition of hydrogen peroxide with formation of a hydroxyl radical. Decomposition of hydrogen peroxide with iron hydroxides is called Fenton-like reaction. Studying the features of Fenton-like reaction in biological systems is the subject of future research.
Collapse
Affiliation(s)
- Konstantin O Muranov
- Emanuel Institute of Biochemical Physics, Russian Academy of Sciences, Moscow, 119334, Russia.
| |
Collapse
|
28
|
Gu K, Wu A, Yu B, Zhang T, Lai X, Chen J, Yan H, Zheng P, Luo Y, Luo J, Pu J, Wang Q, Wang H, Chen D. Iron overload induces colitis by modulating ferroptosis and interfering gut microbiota in mice. THE SCIENCE OF THE TOTAL ENVIRONMENT 2023; 905:167043. [PMID: 37717771 DOI: 10.1016/j.scitotenv.2023.167043] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/22/2023] [Revised: 09/08/2023] [Accepted: 09/11/2023] [Indexed: 09/19/2023]
Abstract
BACKGROUND Iron plays a pivotal role in various physiological processes, including intestinal inflammation, ferroptosis, and the modulation of the gut microbiome. However, the way these factors interact with each other is unclear. METHODS Mice models were fed with low, normal and high iron diets to assess their impacts on colitis, ferroptosis and gut microbiota. Untargeted fecal metabolomics analysis, 16S rRNA sequencing, histopathology analysis, real-time quantitative PCR and western blot were performed to analyze the differences in the intestinal inflammatory response and understanding its regulatory mechanisms between low, normal and high iron groups. RESULTS The iron overload changed the serum iron, colon iron and fecal iron. In addition, the iron overload induced the colitis, induced the ferroptosis and altered the microbiome composition in the fecal of mice. By using untargeted fecal metabolomics analysis to screen of metabolites in the fecal, we found that different metabolomics profiles in the fecal samples between iron deficiency, normal iron and iron overload groups. The correlation analysis showed that both of iron deficiency and overload were closely related to Dubosiella. The relationship between microbial communities (e.g., Akkermansia, Alistipes, and Dubosiella) and colitis-related parameters was highly significant. Additionally, Alistipes and Bacteroides microbial communities displayed a close association with ferroptosis-related parameters. Iron overload reduced the concentration of metabolites, which exert the anti-inflammatory effects (e.g., (+)-.alpha.-tocopherol) in mice. The nucleotide metabolism, enzyme metabolism and metabolic diseases were decreased and the lipid metabolism was increased in iron deficiency and iron overload groups compared with normal iron group. CONCLUSION Iron overload exacerbated colitis in mice by modulating ferroptosis and perturbing the gut microbiota. Iron overload-induced ferroptosis was associated with NRF2/GPX-4 signaling pathway. Specific microbial taxa and their associated metabolites were closely intertwined with both colitis and ferroptosis markers.
Collapse
Affiliation(s)
- Ke Gu
- Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu 611130, China; Key Laboratory for Animal Disease-resistance Nutrition of China Ministry of Education, Sichuan Agricultural University, Chengdu 611130, China
| | - Aimin Wu
- Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu 611130, China; Key Laboratory for Animal Disease-resistance Nutrition of China Ministry of Education, Sichuan Agricultural University, Chengdu 611130, China
| | - Bing Yu
- Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu 611130, China; Key Laboratory for Animal Disease-resistance Nutrition of China Ministry of Education, Sichuan Agricultural University, Chengdu 611130, China
| | - Tingting Zhang
- Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu 611130, China; Key Laboratory for Animal Disease-resistance Nutrition of China Ministry of Education, Sichuan Agricultural University, Chengdu 611130, China
| | - Xin Lai
- Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu 611130, China; Key Laboratory for Animal Disease-resistance Nutrition of China Ministry of Education, Sichuan Agricultural University, Chengdu 611130, China
| | - Junzhou Chen
- Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu 611130, China; Key Laboratory for Animal Disease-resistance Nutrition of China Ministry of Education, Sichuan Agricultural University, Chengdu 611130, China
| | - Hui Yan
- Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu 611130, China; Key Laboratory for Animal Disease-resistance Nutrition of China Ministry of Education, Sichuan Agricultural University, Chengdu 611130, China
| | - Ping Zheng
- Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu 611130, China; Key Laboratory for Animal Disease-resistance Nutrition of China Ministry of Education, Sichuan Agricultural University, Chengdu 611130, China
| | - Yuheng Luo
- Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu 611130, China; Key Laboratory for Animal Disease-resistance Nutrition of China Ministry of Education, Sichuan Agricultural University, Chengdu 611130, China
| | - Junqiu Luo
- Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu 611130, China; Key Laboratory for Animal Disease-resistance Nutrition of China Ministry of Education, Sichuan Agricultural University, Chengdu 611130, China
| | - Junning Pu
- Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu 611130, China; Key Laboratory for Animal Disease-resistance Nutrition of China Ministry of Education, Sichuan Agricultural University, Chengdu 611130, China
| | - Quyuan Wang
- Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu 611130, China; Key Laboratory for Animal Disease-resistance Nutrition of China Ministry of Education, Sichuan Agricultural University, Chengdu 611130, China
| | - Huifen Wang
- Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu 611130, China; Key Laboratory for Animal Disease-resistance Nutrition of China Ministry of Education, Sichuan Agricultural University, Chengdu 611130, China
| | - Daiwen Chen
- Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu 611130, China; Key Laboratory for Animal Disease-resistance Nutrition of China Ministry of Education, Sichuan Agricultural University, Chengdu 611130, China.
| |
Collapse
|
29
|
Bagheri-Hosseinabadi Z, Pirsadeghi A, Ostadebrahimi H, Taghipour Khaje Sharifi G, Abbasifard M. Correlation of iron and related factors with disease severity and outcomes and mortality of patients with Coronavirus disease 2019. J Trace Elem Med Biol 2023; 80:127285. [PMID: 37660574 DOI: 10.1016/j.jtemb.2023.127285] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Revised: 08/06/2023] [Accepted: 08/18/2023] [Indexed: 09/05/2023]
Abstract
BACKGROUND Iron is a trace element that possesses immunomodulatory properties and modulates the proneness to the course and outcome of a diverse viral diseases. This study intended to investigate the correlation of different iron-related factors with disease severity and outcomes as well as the mortality of coronavirus disease 2019 (COVID-19) patients. METHODS Blood serum samples were obtained from 80 COVID-19 cases and 100 healthy controls. Concentrations of ferritin, transferrin, total iron binding capacity (TIBC) was measured by Enzyme-linked immunosorbent assay (ELISA) and iron level was measured by immunoturbidometric method. RESULTS Concentrations of iron, transferrin, and TIBC were low, while ferritin level was high in the COVID-19 cases in comparison to controls. In non-survivor (deceased) patients as well as severe subjects, the levels of iron, ferritin, transferrin, and TIBC were significantly different than survivors (discharged) and mild cases. Significant correlations were found between iron and related factors and the clinicopathological features of the patients. Based on ROC curve analysis, iron, ferritin, transferrin, and TIBC had potential to estimate disease severity in COVID-19 subjects. CONCLUSION Iron metabolism is involved in the pathogenesis of COVID-19. Iron and related factors correlate with disease outcomes and might serve as biomarker in diagnosis of the disease severity and estimation of mortality in the COVID-19 subjects.
Collapse
Affiliation(s)
- Zahra Bagheri-Hosseinabadi
- Immunology of Infectious Diseases Research Center, Research Institute of Basic Medical Sciences, Rafsanjan University of Medical Sciences, Rafsanjan, Iran; Department of Clinical Biochemistry, School of Medicine, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
| | - Ali Pirsadeghi
- Student Research Committee, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Hamid Ostadebrahimi
- Department of Pediatrics, Ali-Ibn Abi-Talib Hospital, School of Medicine, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
| | | | - Mitra Abbasifard
- Immunology of Infectious Diseases Research Center, Research Institute of Basic Medical Sciences, Rafsanjan University of Medical Sciences, Rafsanjan, Iran; Department of Internal Medicine, Ali-Ibn Abi-Talib Hospital, School of Medicine, Rafsanjan University of Medical Sciences, Rafsanjan, Iran.
| |
Collapse
|
30
|
Ianiro G, Niro A, Rosa L, Valenti P, Musci G, Cutone A. To Boost or to Reset: The Role of Lactoferrin in Energy Metabolism. Int J Mol Sci 2023; 24:15925. [PMID: 37958908 PMCID: PMC10650157 DOI: 10.3390/ijms242115925] [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: 10/03/2023] [Revised: 10/25/2023] [Accepted: 10/31/2023] [Indexed: 11/15/2023] Open
Abstract
Many pathological conditions, including obesity, diabetes, hypertension, heart disease, and cancer, are associated with abnormal metabolic states. The progressive loss of metabolic control is commonly characterized by insulin resistance, atherogenic dyslipidemia, inflammation, central obesity, and hypertension, a cluster of metabolic dysregulations usually referred to as the "metabolic syndrome". Recently, nutraceuticals have gained attention for the generalized perception that natural substances may be synonymous with health and balance, thus becoming favorable candidates for the adjuvant treatment of metabolic dysregulations. Among nutraceutical proteins, lactoferrin (Lf), an iron-binding glycoprotein of the innate immune system, has been widely recognized for its multifaceted activities and high tolerance. As this review shows, Lf can exert a dual role in human metabolism, either boosting or resetting it under physiological and pathological conditions, respectively. Lf consumption is safe and is associated with several benefits for human health, including the promotion of oral and gastrointestinal homeostasis, control of glucose and lipid metabolism, reduction of systemic inflammation, and regulation of iron absorption and balance. Overall, Lf can be recommended as a promising natural, completely non-toxic adjuvant for application as a long-term prophylaxis in the therapy for metabolic disorders, such as insulin resistance/type II diabetes and the metabolic syndrome.
Collapse
Affiliation(s)
- Giusi Ianiro
- Department of Biosciences and Territory, University of Molise, 86090 Pesche, Italy; (G.I.); (A.N.); (G.M.)
| | - Antonella Niro
- Department of Biosciences and Territory, University of Molise, 86090 Pesche, Italy; (G.I.); (A.N.); (G.M.)
| | - Luigi Rosa
- Department of Public Health and Infectious Diseases, University of Rome La Sapienza, 00185 Rome, Italy; (L.R.); (P.V.)
| | - Piera Valenti
- Department of Public Health and Infectious Diseases, University of Rome La Sapienza, 00185 Rome, Italy; (L.R.); (P.V.)
| | - Giovanni Musci
- Department of Biosciences and Territory, University of Molise, 86090 Pesche, Italy; (G.I.); (A.N.); (G.M.)
| | - Antimo Cutone
- Department of Biosciences and Territory, University of Molise, 86090 Pesche, Italy; (G.I.); (A.N.); (G.M.)
| |
Collapse
|
31
|
Mottaghi S, Abbaszadeh H. Grape seed extract in combination with deferasirox ameliorates iron overload, oxidative stress, inflammation, and liver dysfunction in beta thalassemia children. Complement Ther Clin Pract 2023; 53:101804. [PMID: 37832335 DOI: 10.1016/j.ctcp.2023.101804] [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: 08/13/2022] [Revised: 09/28/2023] [Accepted: 10/01/2023] [Indexed: 10/15/2023]
Abstract
BACKGROUND AND PURPOSE Iron overload in the body is associated with serious and irreversible tissue damage. This study aimed to investigate the iron-chelating, antioxidant, anti-inflammatory, and hepatoprotective activities of grape seed extract (GSE) supplement as well as its safety in β-thalassemia major (β-TM) pediatric patients receiving deferasirox as a standard iron-chelation therapy. MATERIALS AND METHODS The children were randomly allocated to either GSE group (n = 30) or control group (n = 30) to receive GSE (100 mg/day) or placebo capsules, respectively, for 4 weeks. The serum levels of iron, ferritin, total iron-binding capacity (TIBC), alanine transaminase (ALT), aspartate aminotransferase (AST), tumor necrosis factor alpha (TNF-α), high-sensitivity C-reactive protein (hs-CRP), malondialdehyde (MDA), and glutathione (GSH) as well as superoxide dismutase (SOD) activity and hemoglobin (Hb) concentration were measured pre-and post-intervention. RESULTS GSE supplement significantly attenuated the serum levels of iron (p = 0.030), ferritin (p = 0.017), ALT (p = 0.000), AST (p = 0.000), TNF-α (p = 0.000), and hs-CRP (p = 0.001). The TIBC level (p = 0.020) significantly enhanced in the GSE group compared with the placebo group. Moreover, GSE supplement remarkably improved the oxidative stress markers, MDA (p = 0.000) and GSH (p = 0.001). The changes in the SOD activity (p = 0.590) and Hb concentration (p = 0.670) were not statistically different between the groups. CONCLUSION GSE supplement possesses several health beneficial influences on children with β-TM by alleviating iron burden, oxidative stress, inflammation, and liver dysfunction.
Collapse
Affiliation(s)
- Sayeh Mottaghi
- Department of Pediatrics, Faculty of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Hassan Abbaszadeh
- Department of Pharmacology, Faculty of Pharmacy, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran.
| |
Collapse
|
32
|
Satué K, Fazio E, Medica P. Estrogen-iron axis in cyclic mares: Effect of age. Theriogenology 2023; 209:178-183. [PMID: 37418858 DOI: 10.1016/j.theriogenology.2023.06.027] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Revised: 06/16/2023] [Accepted: 06/17/2023] [Indexed: 07/09/2023]
Abstract
In woman and in animal models, estrogens are involved in iron (Fe) homeostasis supporting the hypothesis of the existence of an "estrogen-iron axis". Since advancing age leads to a decrease in estrogen levels, the mechanisms of Fe regulation could be compromised. In cyclic and pregnant mares, to date, there is evidence linking the iron state with estrogens pattern. Then, the objective of this study was to determine the relationship among Fe, ferritin (Ferr), hepcidin (Hepc) and estradiol-17β (E2) in cyclic mares with advancing age. A total of 40 Spanish Purebred mares of different ranges of age was analyzed: 4-6 years (n = 10), 7-9 years (n = 10), 10-12 years (n = 10), and >12 years (n = 10). Blood samples were obtained on days -5, 0, +5 and + 16 of the cycle. Compared to mares of 4-6 years, serum Ferr was significantly higher (P < 0.01) and Fe significantly lower (P < 0.01) in mares >12 years of age. Hepc was significantly higher in mares >12 years (P < 0.01) than in those 7-9 years of age. E2 levels were higher in mares of 7-9 years (P < 0.01) than in 4-6 and >12 years of age. Fe and Ferr were negatively correlated with Hepc (r = -0.71 and r = -0.02, respectively). E2 was negatively correlated with Ferr and Hepc (r = -0.28 and r = -0.50, respectively), and positively with Fe (r = 0.31). There is a direct relationship between E2 and Fe metabolism, mediated by the inhibition of Hepc in Spanish Purebred mares. The reduction of E2 decreases the inhibitory effects on Hepc, increasing the levels of stored Fe and mobilizing less the free Fe in circulation. Based on the fact that ovarian estrogens participate in changes in the parameters indicative of iron status with age, the existence of an "estrogen-iron axis" in the mares'estrous cycle could be considered. Future studies are required to clarify these hormonal and metabolic interrelationships in the mare.
Collapse
Affiliation(s)
- Katiuska Satué
- Department of Animal Medicine and Surgery, Faculty of Veterinary Medicine, CEU-Cardenal Herrera University, Tirant lo Blanc, 7, Alfara del Patriarca, 46115, Valencia, Spain.
| | - Esterina Fazio
- Department of Veterinary Sciences, Veterinary Physiology Unit, Polo Universitario Annunziata, Via Palatucci 13, 98168, Messina, Italy
| | - Pietro Medica
- Department of Veterinary Sciences, Veterinary Physiology Unit, Polo Universitario Annunziata, Via Palatucci 13, 98168, Messina, Italy
| |
Collapse
|
33
|
Liang Y, He J, Chen X, Yin L, Yuan Q, Zeng Q, Zu X, Shen Y. The emerging roles of metabolism in the crosstalk between breast cancer cells and tumor-associated macrophages. Int J Biol Sci 2023; 19:4915-4930. [PMID: 37781517 PMCID: PMC10539698 DOI: 10.7150/ijbs.86039] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Accepted: 09/11/2023] [Indexed: 10/03/2023] Open
Abstract
Breast cancer is the most common cancer affecting women worldwide. Investigating metabolism in breast cancer may accelerate the exploitation of new therapeutic options for immunotherapies. Metabolic reprogramming can confer breast cancer cells (BCCs) with a survival advantage in the tumor microenvironment (TME) and metabolic alterations in breast cancer, and the corresponding metabolic byproducts can affect the function of tumor-associated macrophages (TAMs). Additionally, TAMs undergo metabolic reprogramming in response to signals present in the TME, which can affect their function and breast cancer progression. Here, we review the metabolic crosstalk between BCCs and TAMs in terms of glucose, lipids, amino acids, iron, and adenosine metabolism. Summaries of inhibitors that target metabolism-related processes in BCCs or TAMs within breast cancer have also served as valuable inspiration for novel therapeutic approaches in the fight against this disease. This review provides new perspectives on targeted anticancer therapies for breast cancer that combine immunity with metabolism.
Collapse
Affiliation(s)
- Yuxin Liang
- Cancer Research Institute, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
- Department of Clinical Laboratory Medicine, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
| | - Jun He
- Department of Spine Surgery, The Nanhua Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, China
| | - Xiguang Chen
- Cancer Research Institute, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
| | - Liyang Yin
- Cancer Research Institute, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
| | - Qiong Yuan
- Cancer Research Institute, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
- Department of Clinical Laboratory Medicine, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
| | - Qiting Zeng
- Cancer Research Institute, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
- Department of Clinical Laboratory Medicine, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
| | - Xuyu Zu
- Cancer Research Institute, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
| | - Yingying Shen
- Cancer Research Institute, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
| |
Collapse
|
34
|
Ouyang J, Zhou L, Wang Q. Spotlight on iron and ferroptosis: research progress in diabetic retinopathy. Front Endocrinol (Lausanne) 2023; 14:1234824. [PMID: 37772084 PMCID: PMC10525335 DOI: 10.3389/fendo.2023.1234824] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Accepted: 08/21/2023] [Indexed: 09/30/2023] Open
Abstract
Iron, as the most abundant metallic element within the human organism, is an indispensable ion for sustaining life and assumes a pivotal role in governing glucose and lipid metabolism, along with orchestrating inflammatory responses. The presence of diabetes mellitus (DM) can induce aberrant iron accumulation within the corporeal system. Consequentially, iron overload precipitates a sequence of important adversities, subsequently setting in motion a domino effect wherein ferroptosis emerges as the utmost pernicious outcome. Ferroptosis, an emerging variant of non-apoptotic regulated cell death, operates independently of caspases and GSDMD. It distinguishes itself from alternative forms of controlled cell death through distinctive morphological and biochemical attributes. Its principal hallmark resides in the pathological accrual of intracellular iron and the concomitant generation of iron-driven lipid peroxides. Diabetic retinopathy (DR), established as the predominant cause of adult blindness, wields profound influence over the well-being and psychosocial strain experienced by afflicted individuals. Presently, an abundance of research endeavors has ascertained the pervasive engagement of iron and ferroptosis in the microangiopathy inherent to DR. Evidently, judicious management of iron overload and ferroptosis in the early stages of DR bears the potential to considerably decelerate disease progression. Within this discourse, we undertake a comprehensive exploration of the regulatory mechanisms governing iron homeostasis and ferroptosis. Furthermore, we expound upon the subsequent detriments induced by their dysregulation. Concurrently, we elucidate the intricate interplay linking iron overload, ferroptosis, and DR. Delving deeper, we engage in a comprehensive deliberation regarding strategies to modulate their influence, thereby effecting prospective interventions in the trajectory of DR's advancement or employing them as therapeutic modalities.
Collapse
Affiliation(s)
- Junlin Ouyang
- Department of Endocrinology, China–Japan Union Hospital of Jilin University, Changchun, Jilin, China
| | - Ling Zhou
- Department of Obstetrics and Gynecology, China–Japan Union Hospital of Jilin University, Changchun, Jilin, China
| | - Qing Wang
- Department of Endocrinology, China–Japan Union Hospital of Jilin University, Changchun, Jilin, China
| |
Collapse
|
35
|
Shastri M, Kotru M, Raizada A, Mahajan B, Jain R, Sikka M. Inflammatory markers in geriatric anemia: A study from North India. J Family Med Prim Care 2023; 12:1663-1668. [PMID: 37767440 PMCID: PMC10521830 DOI: 10.4103/jfmpc.jfmpc_2443_22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Revised: 06/03/2023] [Accepted: 06/12/2023] [Indexed: 09/29/2023] Open
Abstract
Background Inflammation has several effects in the geriatrics with reference to iron deficiency anemia (IDA), anemia of chronic disease (ACD), and unexplained anemia (UA). Whether hyperinflammation is part of their pathogenesis or just incidental is unknown. Data are limited regarding inflammatory patterns in IDA, ACD, and UA in anemic geriatrics and inflammation as a component of UA. There is little known about the overlap of inflammation between ACD and UA. Objective The study was undertaken to find the proportion of anemic geriatric patients, aged ≥60 years with raised serum levels of inflammatory markers and their study within IDA, ACD, and UA. Materials and Methods Seventy-five anemic geriatric patients were evaluated for raised serum levels of inflammatory markers: high sensitive C-reactive protein (hsCRP), tumor necrosis factor-α (TNF-α), interleukin-6 (IL-6), and interleukin-8 (IL-8) along with serum ferritin (SF). Results Raised markers were seen in 94.7% of anemic geriatric patients.IL-8 was raised most frequently followed by TNF-α, IL-6, hsCRP, and SF. No distinct inflammatory profile could be elicited between ACD and UA. The hyperinflammatory profile irrespective of the underlying etiology of geriatric anemia suggests that aging per se is pro-inflammatory state. Conclusion Geriatric anemia can be thought to develop on background of subclinical low-grade inflammation along with superimposed nutritional deficiencies or chronic diseases.
Collapse
Affiliation(s)
- Malvika Shastri
- Department of Pathology, University College of Medical Sciences and Guru Teg Bahadur Hospital, New Delhi, Delhi, India
| | - Mrinalini Kotru
- Department of Pathology, University College of Medical Sciences and Guru Teg Bahadur Hospital, New Delhi, Delhi, India
| | - Alpana Raizada
- Department of Medicine, University College of Medical Sciences and Guru Teg Bahadur Hospital, New Delhi, Delhi, India
| | - Bhawna Mahajan
- Department of Biochemistry, GB Pant Institute of Post Graduate Medical Education and Research, New Delhi, Delhi, India
| | - Rajat Jain
- Department of Pathology, University College of Medical Sciences and Guru Teg Bahadur Hospital, New Delhi, Delhi, India
| | - Meera Sikka
- Department of Pathology, University College of Medical Sciences and Guru Teg Bahadur Hospital, New Delhi, Delhi, India
| |
Collapse
|
36
|
Ceron JJ, Pardo-Marin L, Wdowiak A, Zoia A, Wochnik M, Szczubiał M, Bochniarz M, Tecles F, Martinez-Subiela S, Tvarijonaviciute A, Dąbrowski R. Divergences between serum C-reactive protein and ferritin concentrations in canine pyometra. BMC Vet Res 2023; 19:78. [PMID: 37344860 DOI: 10.1186/s12917-023-03630-3] [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: 12/03/2022] [Accepted: 06/05/2023] [Indexed: 06/23/2023] Open
Abstract
The main aim of this report was to investigate and compare the response of serum C-reactive protein (CRP) and ferritin, two positive acute phase proteins (APPs) which usually show an increase in inflammatory processes, in dogs with pyometra. For this purpose, two different studies were made. In the first one , both proteins were measured together in an APPs profile in 25 dogs with pyometra, 25 dogs with pancreatitis (as an example of a positive inflammatory control group), and in 25 healthy dogs. In the second study, to advance the knowledge of the changes and evolution of serum ferritin and CRP in dogs with pyometra after treatment, the concentrations of both APPs were analyzed in 30 dogs with pyometra at diagnosis and after ovariohysterectomy and in 10 clinically healthy female dogs before and after elective spaying. In both studies, bitches with pyometra showed significant increases in serum CRP, indicating an inflammatory condition, but not in serum ferritin despite being a moderate positive APP. This divergence between the dynamics of these APPs could be a useful tool for the suspicion of cases of canine pyometra.
Collapse
Affiliation(s)
- Jose J Ceron
- Interdisciplinary Laboratory of Clinical Pathology (Interlab-UMU), Campus of Excellence Mare Nostrum, University of Murcia, Espinardo, Murcia, 30100, Spain
| | - Luis Pardo-Marin
- Interdisciplinary Laboratory of Clinical Pathology (Interlab-UMU), Campus of Excellence Mare Nostrum, University of Murcia, Espinardo, Murcia, 30100, Spain
| | - Anna Wdowiak
- Department and Clinic of Animal Reproduction, Faculty of Veterinary Medicine, University of Life Sciences, Głeboka 30st, Lublin, 20-612, Poland
| | - Andrea Zoia
- Division of Internal Medicine, San Marco Veterinary Clinic, Viale dell'Industria 3, Veggiano, Padua, 35030, Italy
| | - Marco Wochnik
- Department and Clinic of Animal Reproduction, Faculty of Veterinary Medicine, University of Life Sciences, Głeboka 30st, Lublin, 20-612, Poland
| | - Marek Szczubiał
- Department and Clinic of Animal Reproduction, Faculty of Veterinary Medicine, University of Life Sciences, Głeboka 30st, Lublin, 20-612, Poland
| | - Mariola Bochniarz
- Department and Clinic of Animal Reproduction, Faculty of Veterinary Medicine, University of Life Sciences, Głeboka 30st, Lublin, 20-612, Poland
| | - Fernando Tecles
- Interdisciplinary Laboratory of Clinical Pathology (Interlab-UMU), Campus of Excellence Mare Nostrum, University of Murcia, Espinardo, Murcia, 30100, Spain
| | - Silvia Martinez-Subiela
- Interdisciplinary Laboratory of Clinical Pathology (Interlab-UMU), Campus of Excellence Mare Nostrum, University of Murcia, Espinardo, Murcia, 30100, Spain
| | - Asta Tvarijonaviciute
- Interdisciplinary Laboratory of Clinical Pathology (Interlab-UMU), Campus of Excellence Mare Nostrum, University of Murcia, Espinardo, Murcia, 30100, Spain.
| | - Roman Dąbrowski
- Department and Clinic of Animal Reproduction, Faculty of Veterinary Medicine, University of Life Sciences, Głeboka 30st, Lublin, 20-612, Poland
| |
Collapse
|
37
|
Park JJ, Yoon M, Cho HW, Lee SE, Choi JO, Yoo BS, Kang SM, Choi DJ. Iron Deficiency in Korean Patients With Heart Failure. J Korean Med Sci 2023; 38:e177. [PMID: 37309696 PMCID: PMC10261703 DOI: 10.3346/jkms.2023.38.e177] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Accepted: 02/24/2023] [Indexed: 06/14/2023] Open
Abstract
BACKGROUND Although iron deficiency (ID) is an important and treatable risk factor for heart failure (HF), data on ID are scarce in Asian patients with HF. Therefore, we sought to determine the prevalence and clinical characteristics of ID in hospitalized Korean patients with HF. METHODS In this prospective, multicenter cohort study, 461 patients with acute HF seen at five tertiary centers from January to November 2019 in Korea were enrolled. ID was defined as serum ferritin < 100 μg/L or ferritin 100-299 μg/L in combination with transferrin saturation < 20%. RESULTS The patients' mean age was 67.6 ± 14.9 years, and 61.8% were male. Among total 461 patients, ID was present in 248 patients (53.8%). The prevalence of ID was significantly higher in women than in men (65.3% vs. 47.3%, P < 0.001). In a multivariable logistic regression analysis, the independent predictors of ID were female sex (odds ratio [OR], 2.19; 95% confidence interval [CI], 1.47-3.30), valvular heart disease (OR, 2.10; 95% CI, 1.10-4.17), higher heart rate (OR, 1.10; 95% CI, 1.01-1.21), anemia (OR, 1.60; 95% CI, 1.07-2.40), and the use of clopidogrel (OR, 1.56; 95% CI, 1.00-2.45). Among women, the prevalence of ID did not significantly differ between younger and older women (< 65 years: 73.7% vs. ≥ 65 years: 63.0%, P = 0.222), those with low and high body mass index (BMI < 25 kg/m²: 66.2% vs. BMI ≥ 25 kg/m²: 69.6%, P = 0.703), or those with low and high natriuretic peptide (NP) levels (NP < median: 69.8% vs. NP ≥ median: 61.1%, P = 0.295). Only 0.2% patients with acute HF received intravenous iron supplementation in Korea. CONCLUSION The prevalence of ID is high in hospitalized Korean patients with HF. Because ID cannot be diagnosed by clinical parameters, routine laboratory examinations are necessary to identify patients with ID. TRIAL REGISTRATION ClinicalTrials.gov Identifier: NCT04812873.
Collapse
Affiliation(s)
- Jin Joo Park
- Division of Cardiology, Cardiovascular Center, Department of Internal Medicine, Seoul National University Bundang Hospital, Seoul National University College of Medicine, Seongnam, Korea
| | - Minjae Yoon
- Division of Cardiology, Cardiovascular Center, Department of Internal Medicine, Seoul National University Bundang Hospital, Seoul National University College of Medicine, Seongnam, Korea
| | - Hyoung-Won Cho
- Division of Cardiology, Cardiovascular Center, Department of Internal Medicine, Seoul National University Bundang Hospital, Seoul National University College of Medicine, Seongnam, Korea
| | - Sang-Eun Lee
- Division of Cardiology, Department of Internal Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Jin-Oh Choi
- Department of Internal Medicine, Sungkyunkwan University College of Medicine, Seoul, Korea
| | - Byung-Su Yoo
- Department of Internal Medicine, Yonsei University Wonju College of Medicine, Wonju, Korea
| | - Seok-Min Kang
- Division of Cardiology, Department of Internal Medicine, Severance Hospital, Yonsei University College of Medicine, Seoul, Korea
| | - Dong-Ju Choi
- Division of Cardiology, Cardiovascular Center, Department of Internal Medicine, Seoul National University Bundang Hospital, Seoul National University College of Medicine, Seongnam, Korea.
| |
Collapse
|
38
|
Yuan T, Jia Q, Zhu B, Chen D, Long H. Synergistic immunotherapy targeting cancer-associated anemia: prospects of a combination strategy. Cell Commun Signal 2023; 21:117. [PMID: 37208766 DOI: 10.1186/s12964-023-01145-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Accepted: 04/23/2023] [Indexed: 05/21/2023] Open
Abstract
Cancer-associated anemia promotes tumor progression, leads to poor quality of life in patients with cancer, and even obstructs the efficacy of immune checkpoint inhibitors therapy. However, the precise mechanism for cancer-associated anemia remains unknown and the feasible strategy to target cancer-associated anemia synergizing immunotherapy needs to be clarified. Here, we review the possible mechanisms of cancer-induced anemia regarding decreased erythropoiesis and increased erythrocyte destruction, and cancer treatment-induced anemia. Moreover, we summarize the current paradigm for cancer-associated anemia treatment. Finally, we propose some prospective paradigms to slow down cancer-associated anemia and synergistic the efficacy of immunotherapy. Video Abstract.
Collapse
Affiliation(s)
- Ting Yuan
- Institute of Cancer, Xinqiao Hospital, Third Military Medical University, Chongqing, 400037, China
| | - Qingzhu Jia
- Institute of Cancer, Xinqiao Hospital, Third Military Medical University, Chongqing, 400037, China
- Chongqing Key Laboratory of Immunotherapy, Xinqiao Hospital, Third Military Medical University, Chongqing, 400037, China
| | - Bo Zhu
- Institute of Cancer, Xinqiao Hospital, Third Military Medical University, Chongqing, 400037, China.
- Chongqing Key Laboratory of Immunotherapy, Xinqiao Hospital, Third Military Medical University, Chongqing, 400037, China.
| | - Degao Chen
- Institute of Cancer, Xinqiao Hospital, Third Military Medical University, Chongqing, 400037, China.
- Chongqing Key Laboratory of Immunotherapy, Xinqiao Hospital, Third Military Medical University, Chongqing, 400037, China.
| | - Haixia Long
- Institute of Cancer, Xinqiao Hospital, Third Military Medical University, Chongqing, 400037, China.
- Chongqing Key Laboratory of Immunotherapy, Xinqiao Hospital, Third Military Medical University, Chongqing, 400037, China.
| |
Collapse
|
39
|
Hamlett ED, Flores-Aguilar L, Handen B, Potier MC, Granholm AC, Sherman S, Puig V, Santoro JD, Carmona-Iragui M, Rebillat AS, Head E, Strydom A, Busciglio J. Innovating Therapies for Down Syndrome: An International Virtual Conference of the T21 Research Society. Mol Syndromol 2023; 14:89-100. [PMID: 37064334 PMCID: PMC10090974 DOI: 10.1159/000526021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Accepted: 07/11/2022] [Indexed: 11/13/2022] Open
Abstract
Research focused on Down syndrome continued to gain momentum in the last several years and is advancing our understanding of how trisomy 21 (T21) modifies molecular and cellular processes. The Trisomy 21 Research Society (T21RS) is the premier scientific organization for researchers and clinicians studying Down syndrome. During the COVID pandemic, T21RS held its first virtual conference program, sponsored by the University of California at Irvine, on June 8-10, 2021 and brought together 342 scientists, families, and industry representatives from over 25 countries to share the latest discoveries on underlying cellular and molecular mechanisms of T21, cognitive and behavioral changes, and comorbidities associated with Down syndrome, including Alzheimer's disease and Regression Disorder. Presentations of 91 cutting-edge abstracts reflecting neuroscience, neurology, model systems, psychology, biomarkers, and molecular and pharmacological therapeutic approaches demonstrate the compelling interest and continuing advancement toward innovating biomarkers and therapies aimed at ameliorating health conditions associated with T21.
Collapse
Affiliation(s)
- Eric D. Hamlett
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Lisi Flores-Aguilar
- Department of Anatomy and Cell Biology, McGill University, Montreal, Québec, Canada
- Department of Pathology and Laboratory Medicine, University of California, Irvine, California, USA
| | - Benjamin Handen
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | | | - Ann-Charlotte Granholm
- Department of Neurosurgery, University of Colorado Anschutz Medical Campus, Denver, Colorado, USA
| | - Stephanie Sherman
- Department of Human Genetics, Emory University, Atlanta, Georgia, USA
| | - Victoria Puig
- Catalan Institute of Nanoscience and Nanotechnology, Barcelona, Spain
| | - Jonathan D. Santoro
- Neurological Institute, University of Southern California Keck School of Medicine, Los Angeles, California, USA
| | - María Carmona-Iragui
- Hospital de la Santa Crue I Sant Pau, Universitat Autònoma de Barcelona, Barcelona, Spain
| | | | - Elizabeth Head
- Department of Pathology and Laboratory Medicine, University of California, Irvine, California, USA
| | - André Strydom
- Institute of Psychiatry, King's College London, London, UK
| | - Jorge Busciglio
- Neurobiology and Behavior School of Biological Sciences, University of California, Irvine, California, USA
| |
Collapse
|
40
|
Barania Adabi S, Daneghian S, Khalkhali H, Nejadrahim R, Shivappa N. The association between inflammatory and immune system biomarkers and the dietary inflammatory index in patients with COVID-19. Front Nutr 2023; 10:1075061. [PMID: 37063325 PMCID: PMC10103612 DOI: 10.3389/fnut.2023.1075061] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Accepted: 02/07/2023] [Indexed: 04/03/2023] Open
Abstract
BackgroundInflammation and cytokine storm have been reported to be the main cause of acute symptoms of coronavirus disease (COVID-19). Diet-induced inflammation may affect the condition of patients with COVID-19. Therefore, this study aimed to investigate the relationship between disease severity, inflammatory and immune system biomarkers, and the dietary inflammatory index (DII) in patients with COVID-19.MethodsThis cross-sectional study was conducted on 500 adult patients with COVID-19. Patients were divided into mild, moderate, and severe conditions based on clinical and laboratory evidence. A validated food frequency questionnaire (FFQ) was used to determine DII and energy-adjusted DII (E-DII) scores. The serum C-reactive protein (CRP) level and blood cell count were measured for all patients. Multiple linear regression was used to explore the association between DII and E-DII and CRP, blood cell counts, and hospitalization in patients with COVID-19.ResultsCoronavirus disease (COVID-19) patients with higher DII had higher consumption of fat and carbohydrate and lower intakes of protein, anti-inflammatory nutrients, garlic, caffeine, tea, onion, and fiber (P < 0.05). There was a positive association between DII and CRP (β = 1.024, P < 0.001), hospitalization (β = 1.062, P < 0.001), WBC count (β = 0.486, P < 0.009), neutrophil count (β = 0.565, P < 0.001), and neutrophil-to-lymphocyte ratio (β = 0.538, P < 0.001) and a negative association between DII and the lymphocyte count (β = −0.569, P < 0.001). There was a positive association between E-DII and hospitalization (β = 1.645, P < 0.001), WBC count (β = 0.417, P < 0.02), and neutrophil-to-lymphocyte ratio (β = 0.35, P < 0.03).ConclusionThere is a positive correlation between DII and inflammation, immune hyperactivation, and length of hospital stay in patients with COVID-19. Further longitudinal studies are necessary.
Collapse
Affiliation(s)
- Somayyeh Barania Adabi
- Maternal and Childhood Obesity Research Center, Urmia University of Medical Sciences, Urmia, Iran
- Department of Nutrition, School of Medicine, Urmia University of Medical Sciences, Urmia, Iran
| | - Sevana Daneghian
- Department of Nutrition, School of Medicine, Urmia University of Medical Sciences, Urmia, Iran
- Food and Beverages Safety Research Center, Urmia University of Medical Sciences, Urmia, Iran
- *Correspondence: Sevana Daneghian
| | | | - Rahim Nejadrahim
- Department of Infectious Diseases and Dermatology, Urmia University of Medical Sciences, Urmia, Iran
| | - Nitin Shivappa
- Cancer Prevention and Control Program, Department of Epidemiology and Biostatistics, Arnold School of Public Health, University of South Carolina, Columbia, SC, United States
| |
Collapse
|
41
|
Halcrow PW, Kumar N, Hao E, Khan N, Meucci O, Geiger JD. Mu opioid receptor-mediated release of endolysosome iron increases levels of mitochondrial iron, reactive oxygen species, and cell death. NEUROIMMUNE PHARMACOLOGY AND THERAPEUTICS 2023; 2:19-35. [PMID: 37027339 PMCID: PMC10070011 DOI: 10.1515/nipt-2022-0013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Accepted: 09/01/2022] [Indexed: 11/15/2022]
Abstract
Objectives Opioids including morphine and DAMGO activate mu-opioid receptors (MOR), increase intracellular reactive oxygen species (ROS) levels, and induce cell death. Ferrous iron (Fe2+) through Fenton-like chemistry increases ROS levels and endolysosomes are "master regulators of iron metabolism" and contain readily-releasable Fe2+ stores. However, mechanisms underlying opioid-induced changes in endolysosome iron homeostasis and downstream-signaling events remain unclear. Methods We used SH-SY5Y neuroblastoma cells, flow cytometry, and confocal microscopy to measure Fe2+ and ROS levels and cell death. Results Morphine and DAMGO de-acidified endolysosomes, decreased endolysosome Fe2+ levels, increased cytosol and mitochondria Fe2+ and ROS levels, depolarized mitochondrial membrane potential, and induced cell death; effects blocked by the nonselective MOR antagonist naloxone and the selective MOR antagonist β-funaltrexamine (β-FNA). Deferoxamine, an endolysosome-iron chelator, inhibited opioid agonist-induced increases in cytosolic and mitochondrial Fe2+ and ROS. Opioid-induced efflux of endolysosome Fe2+ and subsequent Fe2+ accumulation in mitochondria were blocked by the endolysosome-resident two-pore channel inhibitor NED-19 and the mitochondrial permeability transition pore inhibitor TRO. Conclusions Opioid agonist-induced increases in cytosolic and mitochondrial Fe2+ and ROS as well as cell death appear downstream of endolysosome de-acidification and Fe2+ efflux from the endolysosome iron pool that is sufficient to affect other organelles.
Collapse
Affiliation(s)
- Peter W. Halcrow
- Department of Biomedical Sciences, University of North Dakota School of Medicine and Health Sciences, Grand Forks, ND, USA
| | - Nirmal Kumar
- Department of Biomedical Sciences, University of North Dakota School of Medicine and Health Sciences, Grand Forks, ND, USA
| | - Emily Hao
- Department of Biomedical Sciences, University of North Dakota School of Medicine and Health Sciences, Grand Forks, ND, USA
| | - Nabab Khan
- Department of Biomedical Sciences, University of North Dakota School of Medicine and Health Sciences, Grand Forks, ND, USA
| | - Olimpia Meucci
- Department of Physiology and Pharmacology, Drexel University School of Medicine, Philadelphia, PA, USA
| | - Jonathan D. Geiger
- Department of Biomedical Sciences, University of North Dakota School of Medicine and Health Sciences, Grand Forks, ND, USA
| |
Collapse
|
42
|
Ayton S, Janelidze S, Kalinowski P, Palmqvist S, Belaidi AA, Stomrud E, Roberts A, Roberts B, Hansson O, Bush AI. CSF ferritin in the clinicopathological progression of Alzheimer's disease and associations with APOE and inflammation biomarkers. J Neurol Neurosurg Psychiatry 2023; 94:211-219. [PMID: 36357168 PMCID: PMC9992756 DOI: 10.1136/jnnp-2022-330052] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Accepted: 10/23/2022] [Indexed: 11/12/2022]
Abstract
BACKGROUND A putative role for iron in driving Alzheimer's disease (AD) progression is complicated by previously reported associations with neuroinflammation, apolipoprotein E and AD proteinopathy. To establish how iron interacts with clinicopathological features of AD and at what disease stage iron influences cognitive outcomes, we investigated the association of cerebrospinal fluid (CSF) biomarkers of iron (ferritin), inflammation (acute phase response proteins) and apolipoproteins with pathological biomarkers (CSF Aβ42/t-tau, p-tau181), clinical staging and longitudinal cognitive deterioration in subjects from the BioFINDER cohort, with replication of key results in the Alzheimer's Disease Neuroimaging Initiative (ADNI) cohort. METHODS Ferritin, acute phase response proteins (n=9) and apolipoproteins (n=6) were measured in CSF samples from BioFINDER (n=1239; 4 years cognitive follow-up) participants stratified by cognitive status (cognitively unimpaired, mild cognitive impairment, AD) and for the presence of amyloid and tangle pathology using CSF Aβ42/t-tau (A+) and p-tau181 (T+). The ferritin and apolipoprotein E associations were replicated in the ADNI (n=264) cohort. RESULTS In both cohorts, ferritin and apoE were elevated in A-T+ and A+T+ subjects (16%-40%), but not clinical diagnosis. Other apolipoproteins and acute phase response proteins increased with clinical diagnosis, not pathology. CSF ferritin was positively associated with p-tau181, which was mediated by apolipoprotein E. An optimised threshold of ferritin predicted cognitive deterioration in mild cognitive impairment subjects in the BioFINDER cohort, especially those people classified as A-T- and A+T-. CONCLUSIONS CSF markers of iron and neuroinflammation have distinct associations with disease stages, while iron may be more intimately associated with apolipoprotein E and tau pathology.
Collapse
Affiliation(s)
- Scott Ayton
- Melbourne Dementia Research Centre, Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, Australia
| | - Shorena Janelidze
- Clinical Memory Research Unit, Department of Clinical Sciences, Malmö, Lund University, Sweden
| | - Pawel Kalinowski
- Melbourne Dementia Research Centre, Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, Australia
| | - Sebastian Palmqvist
- Clinical Memory Research Unit, Department of Clinical Sciences, Malmö, Lund University, Sweden
- Memory Clinic, Skåne University Hospital, Malmö, Sweden
| | - Abdel A. Belaidi
- Melbourne Dementia Research Centre, Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, Australia
| | - Erik Stomrud
- Clinical Memory Research Unit, Department of Clinical Sciences, Malmö, Lund University, Sweden
- Memory Clinic, Skåne University Hospital, Malmö, Sweden
| | - Anne Roberts
- Department of Biochemistry, Department of Neurology, Emory University School of Medicine, Atlanta, GA, USA
| | - Blaine Roberts
- Department of Biochemistry, Department of Neurology, Emory University School of Medicine, Atlanta, GA, USA
| | - Oskar Hansson
- Clinical Memory Research Unit, Department of Clinical Sciences, Malmö, Lund University, Sweden
- Memory Clinic, Skåne University Hospital, Malmö, Sweden
| | - Ashley I. Bush
- Melbourne Dementia Research Centre, Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, Australia
| | | | | |
Collapse
|
43
|
Stair MI, Winn CB, Burns MA, Holcombe H, Artim SC, Ge Z, Shen Z, Wang TC, Muthupalani S, Franco-Mahecho O, Ennis K, Georgieff MK, Fox JG. Effects of chronic Helicobacter pylori strain PMSS1 infection on whole brain and gastric iron homeostasis in male INS-GAS mice. Microbes Infect 2023; 25:105045. [PMID: 36162750 DOI: 10.1016/j.micinf.2022.105045] [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: 04/15/2022] [Revised: 09/09/2022] [Accepted: 09/18/2022] [Indexed: 02/03/2023]
Abstract
Iron deficiency, the most common micronutrient deficiency in humans, is associated with long-term deficits in cognition and memory if left untreated. Infection with the gastric pathogen Helicobacter pylori has been linked to iron deficiency anemia (IDA). The H. pylori virulence factor cytotoxin-associated gene A (cagA) is proposed to be especially pertinent in iron deficiency. Male INS-GAS/FVB mice were infected with the CagA+ strain pre-murine Sydney strain 1 (PMSS1) for 12-13 or 27-29 weeks to investigate the role of chronic H. pylori infection in iron deficiency and neurological sequelae. Mice at both timepoints demonstrated significantly elevated gastric histopathology scores and inflammatory cytokines compared to sham-dosed controls. However, only mice at 27-29 weeks post infection had changes in hematological parameters, with significantly decreased erythrocyte count, hematocrit, serum hemoglobin, and increased serum total iron binding capacity. Gastric transcription of iron-regulatory genes Hamp and Bmp4 were significantly downregulated at both timepoints. In the brain, iron-dependent myelingergic and synaptic markers were significantly downregulated at 27-29 weeks. These results indicated that long-term infection of the CagA + PMSS1 strain of H. pylori in this study caused anemia, altered gastric iron homeostasis, and neurological changes similar to those reported in other rodent H. pylori CagA- strain infection models.
Collapse
Affiliation(s)
- Melissa I Stair
- Division of Comparative Medicine, Massachusetts Institute of Technology, Cambridge, MA, United States; Animal Resource Program, Atrium Health Wake Forest Baptist, Winston Salem, NC, United States
| | - Caroline Bodi Winn
- Division of Comparative Medicine, Massachusetts Institute of Technology, Cambridge, MA, United States; Department of Comparative Medicine, Worldwide Research, Development, and Medical, Pfizer, Cambridge, MA, United States
| | - Monika A Burns
- Division of Comparative Medicine, Massachusetts Institute of Technology, Cambridge, MA, United States; Novartis Institutes for BioMedical Research, Cambridge, MA, United States
| | - Hilda Holcombe
- Division of Comparative Medicine, Massachusetts Institute of Technology, Cambridge, MA, United States
| | - Stephen C Artim
- Division of Comparative Medicine, Massachusetts Institute of Technology, Cambridge, MA, United States; Merck Research Laboratories, Merck, South San Francisco, CA, United States
| | - Zhongming Ge
- Division of Comparative Medicine, Massachusetts Institute of Technology, Cambridge, MA, United States
| | - Zeli Shen
- Division of Comparative Medicine, Massachusetts Institute of Technology, Cambridge, MA, United States
| | - Timothy C Wang
- Department of Medicine, Columbia University, New York, NY, United States
| | - Sureshkumar Muthupalani
- Division of Comparative Medicine, Massachusetts Institute of Technology, Cambridge, MA, United States
| | - Olga Franco-Mahecho
- Division of Comparative Medicine, Massachusetts Institute of Technology, Cambridge, MA, United States
| | - Kathleen Ennis
- Division of Neonatology, Department of Pediatrics, University of Minnesota Medical School, Minneapolis, MN, United States
| | - Michael K Georgieff
- Division of Neonatology, Department of Pediatrics, University of Minnesota Medical School, Minneapolis, MN, United States
| | - James G Fox
- Division of Comparative Medicine, Massachusetts Institute of Technology, Cambridge, MA, United States; Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, United States.
| |
Collapse
|
44
|
Kunasegaran T, Balasubramaniam VRMT, Arasoo VJT, Palanisamy UD, Ramadas A. Diet Gut Microbiota Axis in Pregnancy: A Systematic Review of Recent Evidence. Curr Nutr Rep 2023; 12:203-214. [PMID: 36810808 PMCID: PMC9974723 DOI: 10.1007/s13668-023-00453-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/27/2022] [Indexed: 02/23/2023]
Abstract
PURPOSE OF REVIEW Although gut microbiota have been associated with the etiology of some diseases, the influence of foods on gut microbiota, especially among pregnant women, remains unclear. Hence, a systematic review was performed to investigate the association between diet and gut microbiota and their influence on metabolic health in pregnant women. RECENT FINDINGS We performed the systematic review using the Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) 2020 protocol to investigate the association between diet and gut microbiota and their influence on metabolic role in pregnant women. Five databases were searched for relevant peer-reviewed articles published in English since 2011. Two-staged screening of 659 retrieved records resulted in the inclusion of 10 studies. The collated findings suggested associations between nutrient intakes and four key microbes: Collinsella, Lachnospira, Sutterella, Faecalibacterium, and the Firmicutes/Bacteroidetes ratio in pregnant women. Dietary intakes in pregnancy were found to modify the gut microbiota and positively influence the cell metabolism in pregnant women. This review, however, emphasizes the importance of conducting well-designed prospective cohorts to investigate the role of changes in dietary intakes within the pregnancy and the influence of such changes on gut microbiota.
Collapse
Affiliation(s)
- Thubasni Kunasegaran
- Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, 47500 Bandar Sunway, Malaysia
| | | | | | - Uma Devi Palanisamy
- Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, 47500 Bandar Sunway, Malaysia
| | - Amutha Ramadas
- Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, 47500 Bandar Sunway, Malaysia
| |
Collapse
|
45
|
Ledesma-Colunga MG, Baschant U, Weidner H, Alves TC, Mirtschink P, Hofbauer LC, Rauner M. Transferrin receptor 2 deficiency promotes macrophage polarization and inflammatory arthritis. Redox Biol 2023; 60:102616. [PMID: 36746004 PMCID: PMC9932570 DOI: 10.1016/j.redox.2023.102616] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Revised: 01/19/2023] [Accepted: 01/24/2023] [Indexed: 02/04/2023] Open
Abstract
OBJECTIVE Rheumatoid arthritis is an inflammatory joint disease in which synovial iron deposition has been described. Transferrin receptor 2 (Tfr2) represents a critical regulator of systemic iron levels. Loss of Tfr2 function in humans and mice results in iron overload. As iron contributes to inflammatory processes, we investigated whether Tfr2-deletion affects the pathogenesis of inflammatory arthritis in an iron-dependent manner. METHODS Using a global and conditional genetic disruption of Tfr2, we assessed the relevance of Tfr2 in K/BxN serum-transfer arthritis (STA) and macrophage polarization. RESULTS Male Tfr2-/- mice subjected to STA developed pronounced joint swelling, and bone erosion as compared to Tfr2+/+ littermate-controls (P < 0.01). Furthermore, an increase of neutrophils and macrophages/monocytes was observed in the inflammatory infiltrate within the paws of Tfr2-/- mice. To elucidate whether Tfr2 in myeloid cells has a direct role in the pathogenesis of arthritis or whether the effects were mediated via the systemic iron overload, we induced STA in Tfr2fl/fl-LysMCre + mice, which showed normal iron-loading. Cre + female mice displayed increased disease development compared to Cre-controls. As macrophages regulate iron availability and innate immunity, we hypothesized that Tfr2-deficiency would polarize macrophages toward a pro-inflammatory state (M1) that contributes to arthritis progression. In response to IFN-γ stimulation, Tfr2-/- macrophages showed increased expression of M1-like cytokines, IFN-γ-target genes, nitric-oxide production, and prolonged STAT1 activation compared to Tfr2+/+ macrophages (P < 0.01), while pre-treatment with ruxolitinib abolished Tfr2-driven M1-like polarization. CONCLUSION Taken together, these findings suggest a protective role of Tfr2 in macrophages on the progression of arthritis via suppression of M1-like polarization.
Collapse
Affiliation(s)
- Maria G Ledesma-Colunga
- Department of Medicine III and Center for Healthy Aging, Technische Universität Dresden, Dresden, Germany
| | - Ulrike Baschant
- Department of Medicine III and Center for Healthy Aging, Technische Universität Dresden, Dresden, Germany
| | - Heike Weidner
- Department of Medicine III and Center for Healthy Aging, Technische Universität Dresden, Dresden, Germany
| | - Tiago C Alves
- Institute of Clinical Chemistry and Laboratory Medicine, Technische Universität Dresden, Dresden, Germany
| | - Peter Mirtschink
- Institute of Clinical Chemistry and Laboratory Medicine, Technische Universität Dresden, Dresden, Germany
| | - Lorenz C Hofbauer
- Department of Medicine III and Center for Healthy Aging, Technische Universität Dresden, Dresden, Germany
| | - Martina Rauner
- Department of Medicine III and Center for Healthy Aging, Technische Universität Dresden, Dresden, Germany.
| |
Collapse
|
46
|
Seo H, Yoon SY, ul-Haq A, Jo S, Kim S, Rahim MA, Park HA, Ghorbanian F, Kim MJ, Lee MY, Kim KH, Lee N, Won JH, Song HY. The Effects of Iron Deficiency on the Gut Microbiota in Women of Childbearing Age. Nutrients 2023; 15:nu15030691. [PMID: 36771397 PMCID: PMC9919165 DOI: 10.3390/nu15030691] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2023] [Revised: 01/26/2023] [Accepted: 01/27/2023] [Indexed: 01/31/2023] Open
Abstract
Iron deficiency anemia (IDA) is the most prevalent and common nutritional deficiency worldwide and is a global health problem with significant risk, particularly among women of reproductive age. Oral iron supplementation is the most widely used and cost-effective treatment for iron deficiency and IDA. However, there are limitations regarding side effects such as enteritis, treatment compliance, and bioavailability. Intestinal microbiome characteristic research has been recently conducted to overcome these issues, but more is needed. Against this background, a metagenomics study on the 16S gene in the feces of young women vulnerable to IDA was conducted. As a result of analyzing 16 normal subjects and 15 IDA patients, significant differences in bacterial community distribution were identified. In particular, a significant decrease in Faecalibacterium was characteristic in IDA patients compared with normal subjects. Furthermore, in the case of patients who recovered from IDA following iron supplementation treatment, it was confirmed that Faecalibacterium significantly recovered to normal levels. However, no significance in beta diversity was seen compared with before treatment. There were also no differences in the beta diversity results between the recovered and normal subjects. Therefore, intestinal dysbiosis during the disease state was considered to be restored as IDA improved. Although the results were derived from a limited number of subjects and additional research is needed, the results of this study are expected to be the basis for developing treatment and prevention strategies based on host-microbiome crosstalk in IDA.
Collapse
Affiliation(s)
- Hoonhee Seo
- Probiotics Microbiome Convergence Center, Soonchunhyang University, Asan-si 31538, Republic of Korea
| | - Seug Yun Yoon
- Division of Hematology & Medical Oncology, Department of Internal Medicine, Soonchunhyang University Seoul Hospital, Seoul 04401, Republic of Korea
| | - Asad ul-Haq
- Probiotics Microbiome Convergence Center, Soonchunhyang University, Asan-si 31538, Republic of Korea
| | - Sujin Jo
- Department of Microbiology and Immunology, School of Medicine, Soonchunhyang University, Cheonan-si 31151, Republic of Korea
| | - Sukyung Kim
- Probiotics Microbiome Convergence Center, Soonchunhyang University, Asan-si 31538, Republic of Korea
| | - Md Abdur Rahim
- Department of Microbiology and Immunology, School of Medicine, Soonchunhyang University, Cheonan-si 31151, Republic of Korea
| | - Hyun-A Park
- Department of Microbiology and Immunology, School of Medicine, Soonchunhyang University, Cheonan-si 31151, Republic of Korea
| | - Fatemeh Ghorbanian
- Department of Microbiology and Immunology, School of Medicine, Soonchunhyang University, Cheonan-si 31151, Republic of Korea
| | - Min Jung Kim
- Division of Hematology & Medical Oncology, Department of Internal Medicine, Soonchunhyang University Seoul Hospital, Seoul 04401, Republic of Korea
| | - Min-Young Lee
- Division of Hematology & Medical Oncology, Department of Internal Medicine, Soonchunhyang University Seoul Hospital, Seoul 04401, Republic of Korea
| | - Kyoung Ha Kim
- Division of Hematology & Medical Oncology, Department of Internal Medicine, Soonchunhyang University Seoul Hospital, Seoul 04401, Republic of Korea
| | - Namsu Lee
- Division of Hematology & Medical Oncology, Department of Internal Medicine, Soonchunhyang University Seoul Hospital, Seoul 04401, Republic of Korea
| | - Jong-Ho Won
- Division of Hematology & Medical Oncology, Department of Internal Medicine, Soonchunhyang University Seoul Hospital, Seoul 04401, Republic of Korea
- Correspondence: (J.-H.W.); (H.-Y.S.)
| | - Ho-Yeon Song
- Probiotics Microbiome Convergence Center, Soonchunhyang University, Asan-si 31538, Republic of Korea
- Department of Microbiology and Immunology, School of Medicine, Soonchunhyang University, Cheonan-si 31151, Republic of Korea
- Correspondence: (J.-H.W.); (H.-Y.S.)
| |
Collapse
|
47
|
Hortová-Kohoutková M, Skotáková M, Onyango IG, Slezáková M, Panovský R, Opatřil L, Slanina P, De Zuani M, Mrkva O, Andrejčinová I, Lázničková P, Dvončová M, Mýtniková A, Ostland V, Šitina M, Stokin GB, Šrámek V, Vlková M, Helán M, Frič J. Hepcidin and ferritin levels as markers of immune cell activation during septic shock, severe COVID-19 and sterile inflammation. Front Immunol 2023; 14:1110540. [PMID: 36776891 PMCID: PMC9911830 DOI: 10.3389/fimmu.2023.1110540] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2022] [Accepted: 01/16/2023] [Indexed: 01/28/2023] Open
Abstract
Introduction Major clinically relevant inflammatory events such as septic shock and severe COVID-19 trigger dynamic changes in the host immune system, presenting promising candidates for new biomarkers to improve precision diagnostics and patient stratification. Hepcidin, a master regulator of iron metabolism, has been intensively studied in many pathologies associated with immune system activation, however these data have never been compared to other clinical settings. Thus, we aimed to reveal the dynamics of iron regulation in various clinical settings and to determine the suitability of hepcidin and/or ferritin levels as biomarkers of inflammatory disease severity. Cohorts To investigate the overall predictive ability of hepcidin and ferritin, we enrolled the patients suffering with three different diagnoses - in detail 40 patients with COVID-19, 29 patients in septic shock and eight orthopedic patients who were compared to nine healthy donors and all cohorts to each other. Results We showed that increased hepcidin levels reflect overall immune cell activation driven by intrinsic stimuli, without requiring direct involvement of infection vectors. Contrary to hepcidin, ferritin levels were more strongly boosted by pathogen-induced inflammation - in septic shock more than four-fold and in COVID-19 six-fold in comparison to sterile inflammation. We also defined the predictive capacity of hepcidin-to-ferritin ratio with AUC=0.79 and P = 0.03. Discussion Our findings confirm that hepcidin is a potent marker of septic shock and other acute inflammation-associated pathologies and demonstrate the utility of the hepcidin-to-ferritin ratio as a predictor of mortality in septic shock, but not in COVID-19.
Collapse
Affiliation(s)
| | - Monika Skotáková
- International Clinical Research Center, St. Anne’s University Hospital, Brno, Czechia
| | - Isaac G. Onyango
- International Clinical Research Center, St. Anne’s University Hospital, Brno, Czechia
| | - Miriam Slezáková
- International Clinical Research Center, St. Anne’s University Hospital, Brno, Czechia
| | - Roman Panovský
- International Clinical Research Center, St. Anne’s University Hospital, Brno, Czechia,1st Department of Internal Medicine/Cardioangiology, Faculty of Medicine, Masaryk University, Brno, Czechia
| | - Lukáš Opatřil
- International Clinical Research Center, St. Anne’s University Hospital, Brno, Czechia,1st Department of Internal Medicine/Cardioangiology, Faculty of Medicine, Masaryk University, Brno, Czechia
| | - Peter Slanina
- Institute of Clinical Immunology and Allergology, Faculty of Medicine, Masaryk University, Brno, Czechia
| | - Marco De Zuani
- International Clinical Research Center, St. Anne’s University Hospital, Brno, Czechia
| | - Ondřej Mrkva
- International Clinical Research Center, St. Anne’s University Hospital, Brno, Czechia
| | - Ivana Andrejčinová
- International Clinical Research Center, St. Anne’s University Hospital, Brno, Czechia,Department of Biology, Faculty of Medicine, Masaryk University, Brno, Czechia
| | - Petra Lázničková
- International Clinical Research Center, St. Anne’s University Hospital, Brno, Czechia
| | - Martina Dvončová
- Department of Anesthesiology and Intensive Care, Faculty of Medicine, Masaryk University, Brno, Czechia
| | - Alexandra Mýtniková
- International Clinical Research Center, St. Anne’s University Hospital, Brno, Czechia,Department of Anesthesiology and Intensive Care, Faculty of Medicine, Masaryk University, Brno, Czechia
| | | | - Michal Šitina
- International Clinical Research Center, St. Anne’s University Hospital, Brno, Czechia,Department of Anesthesiology and Intensive Care, Faculty of Medicine, Masaryk University, Brno, Czechia
| | - Gorazd B. Stokin
- International Clinical Research Center, St. Anne’s University Hospital, Brno, Czechia,Celica BIOMEDICAL, Ljubljana, Slovenia,Division of Neurology, University Medical Centre, Ljubljana, Slovenia
| | - Vladimír Šrámek
- Department of Anesthesiology and Intensive Care, Faculty of Medicine, Masaryk University, Brno, Czechia
| | - Marcela Vlková
- Institute of Clinical Immunology and Allergology, Faculty of Medicine, Masaryk University, Brno, Czechia
| | - Martin Helán
- International Clinical Research Center, St. Anne’s University Hospital, Brno, Czechia,Department of Anesthesiology and Intensive Care, Faculty of Medicine, Masaryk University, Brno, Czechia
| | - Jan Frič
- International Clinical Research Center, St. Anne’s University Hospital, Brno, Czechia,Department of Modern Immunotherapy, Institute of Hematology and Blood Transfusion, Prague, Czechia,*Correspondence: Jan Frič,
| |
Collapse
|
48
|
Xia Y, Luo Q, Huang C, Shi L, Jahangir A, Pan T, Wei X, He J, Liu W, Shi R, Geng Y, Fang J, Tang L, Guo H, Ouyang P, Chen Z. Ferric citrate-induced colonic mucosal damage associated with oxidative stress, inflammation responses, apoptosis, and the changes of gut microbial composition. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2023; 249:114364. [PMID: 36508806 DOI: 10.1016/j.ecoenv.2022.114364] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/28/2021] [Revised: 09/05/2022] [Accepted: 11/28/2022] [Indexed: 06/17/2023]
Abstract
Ferric citrate (FC) has been used as an iron fortifier and nutritional supplement, which is reported to induce colitis in rats, however the underlying mechanism remains to be elucidated. We performed a 16-week study of FC in male healthy C57BL/6 mice (nine-month-old) with oral administration of Ctr (0.9 % NaCl), 1.25 % FC (71 mg/kg/bw), 2.5 % FC (143 mg/kg/bw) and 5 % FC (286 mg/kg/bw). FC-exposure resulted in colon iron accumulation, histological alteration and reduce antioxidant enzyme activities, such as glutathione (GSH), glutathione peroxidase (GSH-Px), superoxide dismutase (SOD) and total antioxidant capacity (T-AOC), together with enhanced lipid peroxidation level, including malondialdehyde (MDA) level and 4-Hydroxynonenal (4-HNE) protein expression. Exposure to FC was associated with upregulated levels of the interleukin (IL)- 6, IL-1β, IL-18, IL-8 and tumor necrosis factor α (TNF-α), while down-regulated levels of IL-4 and IL-10. Exposure to FC was positively associated with the mRNA and protein expressions of cysteine-aspartic proteases (Caspase)- 9, Caspase-3, Bcl-2-associated X protein (Bax), while negatively associated with B-cell lymphoma 2 (Bcl2) in mitochondrial apoptosis signaling pathway. FC-exposure changed the diversity and composition of gut microbes. Additionally, the serum lipopolysaccharide (LPS) contents increased in FC-exposed groups when compared with the control group, while the expression of colonic tight junction proteins (TJPs), such as Claudin-1 and Occludin were decreased. These findings indicate that the colonic mucosal injury induced by FC-exposure are associated with oxidative stress generation, inflammation response and cell apoptosis, as well as the changes in gut microbes diversity and composition.
Collapse
Affiliation(s)
- Yu Xia
- Laboratory of Experimental Animal Disease Model, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China
| | - Qihui Luo
- Laboratory of Experimental Animal Disease Model, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China
| | - Chao Huang
- Laboratory of Experimental Animal Disease Model, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China
| | - Liangqin Shi
- Chengdu University of Traditional Chinese Medicine, College of Basic Medicine, Chengdu 611130, China
| | - Asad Jahangir
- Laboratory of Experimental Animal Disease Model, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China
| | - Ting Pan
- Laboratory of Experimental Animal Disease Model, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China
| | - Xiaoli Wei
- Laboratory of Experimental Animal Disease Model, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China
| | - Junbo He
- Laboratory of Experimental Animal Disease Model, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China
| | - Wentao Liu
- Laboratory of Experimental Animal Disease Model, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China
| | - Riyi Shi
- Center for Paralysis Research & Department of Basic Medical Sciences, College of Veterinary Medicine, Purdue University, West Lafayette, IN 47907, USA
| | - Yi Geng
- Laboratory of Experimental Animal Disease Model, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China
| | - Jing Fang
- Laboratory of Experimental Animal Disease Model, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China
| | - Li Tang
- Laboratory of Experimental Animal Disease Model, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China
| | - Hongrui Guo
- Laboratory of Experimental Animal Disease Model, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China
| | - Ping Ouyang
- Laboratory of Experimental Animal Disease Model, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China
| | - Zhengli Chen
- Laboratory of Experimental Animal Disease Model, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China.
| |
Collapse
|
49
|
Melittin regulates iron homeostasis and mediates macrophage polarization in rats with lumbar spinal stenosis. Biomed Pharmacother 2022; 156:113776. [DOI: 10.1016/j.biopha.2022.113776] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Revised: 09/20/2022] [Accepted: 09/28/2022] [Indexed: 11/21/2022] Open
|
50
|
Pap R, Pandur E, Jánosa G, Sipos K, Nagy T, Agócs A, Deli J. Lutein Decreases Inflammation and Oxidative Stress and Prevents Iron Accumulation and Lipid Peroxidation at Glutamate-Induced Neurotoxicity. Antioxidants (Basel) 2022; 11:2269. [PMID: 36421455 PMCID: PMC9687421 DOI: 10.3390/antiox11112269] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Revised: 11/11/2022] [Accepted: 11/15/2022] [Indexed: 07/30/2023] Open
Abstract
The xanthophyll carotenoid lutein has been widely used as supplementation due to its protective effects in light-induced oxidative stress. Its antioxidant and anti-inflammatory features suggest that it has a neuroprotective role as well. Glutamate is a major excitatory neurotransmitter in the central nervous system (CNS), which plays a key role in regulating brain function. Excess accumulation of intracellular glutamate accelerates an increase in the concentration of reactive oxygen species (ROS) in neurons leading to glutamate neurotoxicity. In this study, we focused on the effects of glutamate on SH-SY5Y neuroblastoma cells to identify the possible alterations in oxidative stress, inflammation, and iron metabolism that affect the neurological function itself and in the presence of antioxidant lutein. First, ROS measurements were performed, and then catalase (CAT) and Superoxide Dismutase (SOD) enzyme activity were determined by enzyme activity assay kits. The ELISA technique was used to detect proinflammatory TNFα, IL-6, and IL-8 cytokine secretions. Alterations in iron uptake, storage, and release were followed by gene expression measurements and Western blotting. Total iron level detections were performed by a ferrozine-based iron detection method, and a heme assay kit was used for heme measurements. The gene expression toward lipid-peroxidation was determined by RT-PCR. Our results show glutamate changes ROS, inflammation, and antioxidant enzyme activity, modulate iron accumulation, and may initiate lipid peroxidation in SH-SY5Y cells. Meanwhile, lutein attenuates the glutamate-induced effects on ROS, inflammation, iron metabolism, and lipid peroxidation. According to our findings, lutein could be a beneficial, supportive treatment in neurodegenerative disorders.
Collapse
Affiliation(s)
- Ramóna Pap
- Department of Pharmaceutical Biology, Faculty of Pharmacy, University of Pécs, Rókus u. 2, H-7624 Pécs, Hungary
| | - Edina Pandur
- Department of Pharmaceutical Biology, Faculty of Pharmacy, University of Pécs, Rókus u. 2, H-7624 Pécs, Hungary
| | - Gergely Jánosa
- Department of Pharmaceutical Biology, Faculty of Pharmacy, University of Pécs, Rókus u. 2, H-7624 Pécs, Hungary
| | - Katalin Sipos
- Department of Pharmaceutical Biology, Faculty of Pharmacy, University of Pécs, Rókus u. 2, H-7624 Pécs, Hungary
| | - 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
|