1
|
Bangolo A, Amoozgar B, Habibi M, Simms E, Nagesh VK, Wadhwani S, Wadhwani N, Auda A, Elias D, Mansour C, Abbott R, Jebara N, Zhang L, Gill S, Ahmed K, Ip A, Goy A, Cho C. Exploring the gut microbiome’s influence on cancer-associated anemia: Mechanisms, clinical challenges, and innovative therapies. World J Gastrointest Pharmacol Ther 2025; 16:105375. [DOI: 10.4292/wjgpt.v16.i2.105375] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/22/2025] [Revised: 03/19/2025] [Accepted: 03/20/2025] [Indexed: 06/03/2025] Open
Abstract
BACKGROUND Anemia is a prevalent and challenging complication in patients with hematologic and solid malignancies, which stems from the direct effects of malignancy, treatment-induced toxicities, and systemic inflammation. It affects patients’ survival, functional status, and quality of life profoundly. Recent literature has highlighted the emerging role of the gut microbiome in the pathogenesis of cancer-associated anemia. The gut microbiota, through its intricate interplay with iron metabolism, inflammatory pathways, and immune modulation, may either exacerbate or ameliorate anemia depending on its composition, and functional integrity. Dysbiosis, characterized by disruption in the gut microbial ecosystem, is very common in cancer patients. This microbial imbalance is implicated in anemia causation through diminished iron absorption, persistent low-grade inflammation, and suppression of erythropoiesis.
AIM To consolidate current evidence regarding the interplay between gut microbiome and anemia in the setting of malignancies. It aims to provide a detailed exploration of the mechanistic links between dysbiosis and anemia, identifies unique challenges associated with various cancer types, and evaluates the efficacy of microbiome-focused therapies. Through this integrative approach, the review seeks to establish a foundation for innovative clinical strategies aimed at mitigating anemia and improving patient outcomes in oncology.
METHODS A literature search was performed using multiple databases, including Google Scholar, PubMed, Scopus, and Web of Science, using a combination of keywords and Boolean operators to refine results. Keywords included “cancer-associated anemia”, “gut microbiome”, “intestinal microbiota”, “iron metabolism”, “gut dysbiosis”, “short-chain fatty acids”, “hematopoiesis”, “probiotics”, “prebiotics”, and “fecal microbiota transplantation”. Articles published in English between 2000 and December 2024 were included, with a focus on contemporary and relevant findings.
RESULTS Therapeutic strategies aimed at restoration of gut microbial homeostasis, such as probiotics, prebiotics, dietary interventions, and fecal microbiota transplantation (FMT), can inhibit anemia-causing pathways by enhancing microbial diversity, suppressing detrimental flora, reducing systemic inflammation and optimizing nutrient absorption.
CONCLUSION Gut dysbiosis causes anemia and impairs response to chemotherapy in cancer patients. Microbiome-centered interventions, such as probiotics, prebiotics, dietary modifications, and FMT, have shown efficacy in restoring microbial balance, reducing inflammation, and enhancing nutrient bioavailability. Emerging approaches, including engineered probiotics and bacteriophage therapies, are promising precision-based, customizable solutions for various microbiome compositions and imbalances. Future research should focus on integrating microbiome-targeted strategies with established anemia therapies.
Collapse
Affiliation(s)
- Ayrton Bangolo
- Department of Hematology and Oncology, John Theurer Cancer Center, Hackensack, NJ 07601, United States
| | - Behzad Amoozgar
- Department of Hematology and Oncology, John Theurer Cancer Center, Hackensack, NJ 07601, United States
| | - Maryam Habibi
- Department of Research, Tulane National Primate Research Center, Covington, LA 70433, United States
| | - Elizabeth Simms
- Department of Medicine, Wake Forest University School of Medicine, Winston-Salem, NC 27101, United States
| | - Vignesh K Nagesh
- Department of Internal Medicine, Hackensack Palisades Medical Center, North Bergen, NJ 07047, United States
| | - Shruti Wadhwani
- Department of Internal Medicine, Hackensack Palisades Medical Center, North Bergen, NJ 07047, United States
| | - Nikita Wadhwani
- Department of Internal Medicine, Hackensack Palisades Medical Center, North Bergen, NJ 07047, United States
| | - Auda Auda
- Department of Family Medicine, Palisades Medical Center, North Bergen, NJ 07047, United States
| | - Daniel Elias
- Department of Internal Medicine, Palisades Medical Center, North Bergen, NJ 07047, United States
| | - Charlene Mansour
- Department of Internal Medicine, Palisades Medical Center, North Bergen, NJ 07047, United States
| | - Robert Abbott
- Rutgers New Jersey Medical School, Newark, NJ 07103, United States
| | - Nisrene Jebara
- Columbia University School of Nursing, New York, NY 10032, United States
| | - Lili Zhang
- Department of Hematology and Oncology, John Theurer Cancer Center, Hackensack, NJ 07601, United States
| | - Sarvarinder Gill
- Department of Hematology and Oncology, John Theurer Cancer Center, Hackensack, NJ 07601, United States
| | - Kareem Ahmed
- Department of Medicine, University of Washington, Seattle, WA 98195, United States
| | - Andrew Ip
- Division of Lymphoma, John Theurer Cancer Center, Hackensack, NJ 07601, United States
| | - Andre Goy
- Division of Lymphoma, John Theurer Cancer Center, Hackensack, NJ 07601, United States
| | - Christina Cho
- Division of Stem Cell Transplant and Cellular Therapy, John Theurer Cancer Center, Hackensack, NJ 07601, United States
| |
Collapse
|
2
|
Wang H, Xu Q, Zhao W, Chan BKW, Chen K, Xie M, Yang X, Ni H, Chan EWC, Yang G, Chen S. Simultaneous functional disruption of the iron acquisition system and type VI secretion system results in complete suppression of virulence in Acinetobacter baumannii. Microbiol Res 2025; 295:128105. [PMID: 40023109 DOI: 10.1016/j.micres.2025.128105] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2024] [Revised: 02/03/2025] [Accepted: 02/14/2025] [Indexed: 03/04/2025]
Abstract
Acinetobacter baumannii (Ab) is one of the most significant bacterial pathogens inducing hospital-acquired infections worldwide, with a high mortality rate. The continuous emergence of multidrug-resistant (MDR) phenotypes presents a significant challenge in combating Ab infections with antimicrobial drugs. In this study, we found that the type VI secretion system and the iron transportation system synergistically enhance siderophore production and further contribute to the virulence of Ab. The double knockout mutant strain, ΔhcpΔbasE, exhibited further reductions in growth rate, siderophore production under iron-deficient conditions, biofilm formation, serum resistance, cell adhesion and invasion, and cytotoxicity compared to the single knockout strains, knockout of T6SS, Δhcp or iron transportation system, ΔbasE. In vitro experiments demonstrated that these two systems work synergistically to enhance virulence, with their combined effect exceeding the additive contributions of each individual system. Consistently, the ΔhcpΔbasE strain failed to cause mortality in the mouse model, even at very high inoculum levels. Further studies revealed that, compared to ATCC17978, ΔhcpΔbasE strain infection resulted in lower levels of extracellular hepcidin and intracellular iron in host cells, which correlate well with the significantly reduced ability to produce siderophores in the double knockout strain. Due to impaired iron acquisition, ΔhcpΔbasE strain became more susceptible to macrophage phagocytosis and exhibited lower survival rates in the host, leading to an inability to trigger a cytokine storm and subsequent host death. The findings of this study provide insights into the Ab pathogenesis and contribute to the development of intervention measures to control clinical Ab infections and mortality.
Collapse
Affiliation(s)
- Han Wang
- Department of Infectious Diseases and Public Health, Jockey Club College of Veterinary Medicine and Life Sciences, City University of Hong Kong, Kowloon, Hong Kong; State Key Lab of Chemical Biology and Drug Discovery and The Department of Food Science and Nutrition, The Hong Kong Polytechnic University, Kowloon, Hong Kong
| | - Qi Xu
- State Key Lab of Chemical Biology and Drug Discovery and The Department of Food Science and Nutrition, The Hong Kong Polytechnic University, Kowloon, Hong Kong
| | - Wenxing Zhao
- State Key Lab of Chemical Biology and Drug Discovery and The Department of Food Science and Nutrition, The Hong Kong Polytechnic University, Kowloon, Hong Kong
| | - Bill Kwan Wai Chan
- State Key Lab of Chemical Biology and Drug Discovery and The Department of Food Science and Nutrition, The Hong Kong Polytechnic University, Kowloon, Hong Kong
| | - Kaichao Chen
- State Key Lab of Chemical Biology and Drug Discovery and The Department of Food Science and Nutrition, The Hong Kong Polytechnic University, Kowloon, Hong Kong
| | - Miaomiao Xie
- State Key Lab of Chemical Biology and Drug Discovery and The Department of Food Science and Nutrition, The Hong Kong Polytechnic University, Kowloon, Hong Kong
| | - Xuemei Yang
- State Key Lab of Chemical Biology and Drug Discovery and The Department of Food Science and Nutrition, The Hong Kong Polytechnic University, Kowloon, Hong Kong
| | - Hongyuhang Ni
- Department of Infectious Diseases and Public Health, Jockey Club College of Veterinary Medicine and Life Sciences, City University of Hong Kong, Kowloon, Hong Kong; State Key Lab of Chemical Biology and Drug Discovery and The Department of Food Science and Nutrition, The Hong Kong Polytechnic University, Kowloon, Hong Kong
| | - Edward Wai Chi Chan
- State Key Lab of Chemical Biology and Drug Discovery and The Department of Food Science and Nutrition, The Hong Kong Polytechnic University, Kowloon, Hong Kong
| | - Guan Yang
- Department of Infectious Diseases and Public Health, Jockey Club College of Veterinary Medicine and Life Sciences, City University of Hong Kong, Kowloon, Hong Kong.
| | - Sheng Chen
- State Key Lab of Chemical Biology and Drug Discovery and The Department of Food Science and Nutrition, The Hong Kong Polytechnic University, Kowloon, Hong Kong; Shenzhen Key Lab for Food Biological Safety Control, The Hong Kong Polytechnic University Shenzhen Research Institute, Shenzhen 518057, China.
| |
Collapse
|
3
|
Givian A, Azizan A, Jamshidi A, Mahmoudi M, Farhadi E. Iron metabolism in rheumatic diseases. J Transl Autoimmun 2025; 10:100267. [PMID: 39867458 PMCID: PMC11763848 DOI: 10.1016/j.jtauto.2025.100267] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2024] [Revised: 12/24/2024] [Accepted: 01/03/2025] [Indexed: 01/28/2025] Open
Abstract
Iron is a crucial element for living organism in terms of oxygen transport, hematopoiesis, enzymatic activity, mitochondrial respiratory chain function and also immune system function. The human being has evolved a mechanism to regulate body iron. In some rheumatic diseases such as rheumatoid arthritis (RA), systemic lupus erythematous (SLE), systemic sclerosis (SSc), ankylosing spondylitis (AS), and gout, this balanced iron regulation is impaired. Altered iron homeostasis can contribute to disease progression through ROS production, fibrosis, inflammation, abnormal bone homeostasis, NETosis and cell senescence. In this review, we have focused on the iron metabolism in rheumatic disease and its role in disease progression.
Collapse
Affiliation(s)
- Aliakbar Givian
- Rheumatology Research Center, Tehran University of Medical Science, Tehran, Iran
- Department of Immunology, School of Medicine, Semnan University of Medical Science, Semnan, Iran
| | - Amin Azizan
- Rheumatology Research Center, Tehran University of Medical Science, Tehran, Iran
- Research Center for Chronic Inflammatory Diseases, Tehran University of Medical Sciences, Tehran, Iran
| | - Ahmadreza Jamshidi
- Rheumatology Research Center, Tehran University of Medical Science, Tehran, Iran
| | - Mahdi Mahmoudi
- Rheumatology Research Center, Tehran University of Medical Science, Tehran, Iran
- Research Center for Chronic Inflammatory Diseases, Tehran University of Medical Sciences, Tehran, Iran
| | - Elham Farhadi
- Rheumatology Research Center, Tehran University of Medical Science, Tehran, Iran
- Research Center for Chronic Inflammatory Diseases, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
4
|
Perry M, Hamza I. Heme and immunity: The heme oxygenase dichotomy. J Inorg Biochem 2025; 267:112844. [PMID: 39978176 DOI: 10.1016/j.jinorgbio.2025.112844] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2024] [Revised: 01/12/2025] [Accepted: 02/02/2025] [Indexed: 02/22/2025]
Abstract
Heme, an iron containing organic ring, is required for a diverse range of biological processes across all forms of life. Although this nutrient is essential, its pro-inflammatory and cytotoxic properties can lead to cellular damage. Heme oxygenase 1 (HO-1) is an endoplasmic reticulum (ER)-anchored enzyme that degrades heme, releasing equimolar amounts of carbon monoxide (CO), biliverdin (BV), and iron. The induction of HO-1 by heme presents an interesting dichotomy in the cell: CO and BV possess anti-inflammatory and antioxidant properties while free iron can be detrimental as it can generate hydroxyl radicals through the Fenton reaction. The heme/HO-1 axis is tightly regulated, and can influence cell fate, local tissue environments, and disease outcomes during pathogen infection. In this review we explore the role of heme during macrophage polarization and its ability to act as an immune activator while also examining the contribution of HO-1 and heme during infections with intracellular and extracellular pathogens. We highlight work from the emerging field of nutritional immunity of heme and iron, and how the substrates and byproducts of heme metabolism via HO-1 can be beneficial to the host or the pathogen depending on the context.
Collapse
Affiliation(s)
- Melissa Perry
- Graduate Program in Biological Sciences, University of Maryland, College Park, MD 20742, USA; Center for Blood Oxygen Transport and Hemostasis, Department of Pediatrics, School of Medicine, University of Maryland, Baltimore, MD 21201, USA
| | - Iqbal Hamza
- Center for Blood Oxygen Transport and Hemostasis, Department of Pediatrics, School of Medicine, University of Maryland, Baltimore, MD 21201, USA; Department of Animal and Avian Sciences, University of Maryland, College Park, MD 20742, USA.
| |
Collapse
|
5
|
Takemura K, Kolasinski V, Del Poeta M, Vieira de Sa NF, Garg A, Ojima I, Del Poeta M, Pereira de Sa N. Iron acquisition strategies in pathogenic fungi. mBio 2025:e0121125. [PMID: 40391928 DOI: 10.1128/mbio.01211-25] [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: 05/22/2025] Open
Abstract
Iron plays a crucial role in various biological processes, including enzyme function, DNA replication, energy production, oxygen transport, lipid, and carbon metabolism. Although it is abundant in the Earth's crust, its bioavailability is restricted by the insolubility of ferric iron (Fe³+) and the auto-oxidation of ferrous iron (Fe²+) in oxygen-rich environments. This limitation poses significant challenges for all organisms, including fungi, which have developed intricate mechanisms for iron acquisition and utilization. These mechanisms include reductive iron uptake, siderophore production/transport, and heme utilization. Fungi employ a variety of enzymes-such as ferric reductases, ferroxidases, permeases, and transporters-to regulate intracellular iron levels effectively. The challenge is heightened for pathogenic fungi during infection, as they must compete with the host's iron-binding proteins like transferrin and lactoferrin, which sequester iron to restrict pathogen growth. This review delves into the iron acquisition strategies of medically important fungi, emphasizing the roles of reductive iron uptake and siderophore pathways. Understanding these mechanisms is vital for enhancing our knowledge of fungal pathogenesis and developing effective treatments. By targeting these iron acquisition processes, new antifungal therapies can be formulated more effectively to combat fungal infections.
Collapse
Affiliation(s)
- Kathryn Takemura
- Institute of Chemical Biology and Drug Discovery, Stony Brook, New York, USA
| | - Vanessa Kolasinski
- Department of Microbiology and Immunology, Stony Brook University, Stony Brook, New York, USA
| | - Matteo Del Poeta
- Department of Microbiology and Immunology, Stony Brook University, Stony Brook, New York, USA
| | | | - Ashna Garg
- Institute of Chemical Biology and Drug Discovery, Stony Brook, New York, USA
| | - Iwao Ojima
- Institute of Chemical Biology and Drug Discovery, Stony Brook, New York, USA
| | - Maurizio Del Poeta
- Institute of Chemical Biology and Drug Discovery, Stony Brook, New York, USA
- Department of Microbiology and Immunology, Stony Brook University, Stony Brook, New York, USA
- Division of Infectious Diseases, School of Medicine, Stony Brook University, Stony Brook, New York, USA
- Veterans Affairs Medical Center, Northport, New York, USA
| | - Nivea Pereira de Sa
- Department of Microbiology and Immunology, Stony Brook University, Stony Brook, New York, USA
| |
Collapse
|
6
|
Zhao J, Liang T, Song M, Ye Y, Guo B, Yan X, Qi P. McFerritin cooperates with McNrf2 to attenuate benzo[a]pyrene-induced oxidative stress in the thick-shell mussel Mytilus coruscus. Int J Biol Macromol 2025; 311:144065. [PMID: 40348216 DOI: 10.1016/j.ijbiomac.2025.144065] [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: 02/05/2025] [Revised: 04/28/2025] [Accepted: 05/07/2025] [Indexed: 05/14/2025]
Abstract
Benzo[a]pyrene (B[a]P), a pervasive polycyclic aromatic hydrocarbon (PAH), induces oxidative stress and cellular damage in marine organisms, posing significant ecological risks. Nuclear factor erythroid 2-related factor 2 (Nrf2) is a key regulator of antioxidant defenses, yet its interaction with ferritin-a critical iron storage protein-remains poorly understood in mollusks. Here, we identified a novel ferritin homolog, McFerritin, in the thick-shelled mussel Mytilus coruscus and investigated its role in B[a]P-induced oxidative stress. Quantitative PCR revealed that McFerritin expression in the adductor muscle was 4.37-fold higher than in the gills, with 5.58 to 9.11-fold upregulation observed in the digestive glands and mantles post-B[a]P exposure. GST pull-down assays confirmed the direct interaction of McFerritin with McNrf2. RNAi experiments demonstrated reciprocal regulation: knockdown of McFerritin increased McNrf2 expression by 1.55-fold and 1.70-fold, while silencing McNrf2 elevated McFerritin levels by 1.30-fold and 1.34-fold in DMSO-treated samples. Silencing either gene resulted in increased apoptosis rates (5.1 % and 13.2 %), enhanced reactive oxygen species (ROS) accumulation (1.26- and 1.79-fold), and decreased activities of antioxidant enzymes (T-AOC, SOD, CAT), underscoring their synergistic role in oxidative stress mitigation. This study elucidates the protective role of Ferritin in M. coruscus, confirming the interaction between McFerritin and McNrf2 in mediating defense mechanisms against oxidative stress, thereby advancing understanding of oxidative stress regulation in marine invertebrates and emphasizing their potential as biomarkers for assessing PAH toxicity in coastal ecosystems.
Collapse
Affiliation(s)
- Jiemei Zhao
- National Engineering Research Center of Marine Facilities Aquaculture, Marine Science and Technology College, Zhejiang Ocean University, Zhoushan, Zhejiang 316004, PR China; Zhoushan Fishery Breeding and Hatching Innovation Center, Zhoushan, Zhejiang 316004, PR China
| | - Tianyi Liang
- National Engineering Research Center of Marine Facilities Aquaculture, Marine Science and Technology College, Zhejiang Ocean University, Zhoushan, Zhejiang 316004, PR China; Zhoushan Fishery Breeding and Hatching Innovation Center, Zhoushan, Zhejiang 316004, PR China
| | - Mingshan Song
- National Engineering Research Center of Marine Facilities Aquaculture, Marine Science and Technology College, Zhejiang Ocean University, Zhoushan, Zhejiang 316004, PR China; Zhoushan Fishery Breeding and Hatching Innovation Center, Zhoushan, Zhejiang 316004, PR China
| | - Yingying Ye
- National Engineering Research Center of Marine Facilities Aquaculture, Marine Science and Technology College, Zhejiang Ocean University, Zhoushan, Zhejiang 316004, PR China; Zhoushan Fishery Breeding and Hatching Innovation Center, Zhoushan, Zhejiang 316004, PR China
| | - Baoying Guo
- National Engineering Research Center of Marine Facilities Aquaculture, Marine Science and Technology College, Zhejiang Ocean University, Zhoushan, Zhejiang 316004, PR China; Zhoushan Fishery Breeding and Hatching Innovation Center, Zhoushan, Zhejiang 316004, PR China
| | - Xiaojun Yan
- National Engineering Research Center of Marine Facilities Aquaculture, Marine Science and Technology College, Zhejiang Ocean University, Zhoushan, Zhejiang 316004, PR China; Zhoushan Fishery Breeding and Hatching Innovation Center, Zhoushan, Zhejiang 316004, PR China
| | - Pengzhi Qi
- National Engineering Research Center of Marine Facilities Aquaculture, Marine Science and Technology College, Zhejiang Ocean University, Zhoushan, Zhejiang 316004, PR China; Zhoushan Fishery Breeding and Hatching Innovation Center, Zhoushan, Zhejiang 316004, PR China.
| |
Collapse
|
7
|
Jawed R, Bhatti H, Khan A. Genetic profile of ferroptosis in non-small cell lung carcinoma and pharmaceutical options for ferroptosis induction. Clin Transl Oncol 2025; 27:1867-1886. [PMID: 39460894 DOI: 10.1007/s12094-024-03754-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Accepted: 10/04/2024] [Indexed: 10/28/2024]
Abstract
Lung cancer (LC) is the leading cause of cancer-related deaths and the second most commonly diagnosed malignancy worldwide. Lung adenocarcinoma (LUAD) and lung squamous cell LC (LUSCC) are the most common subtypes of non-small cell LC (NSCLC). Early diagnosis of LC can be challenging due to a lack of biomarkers. The overall survival (OS) of patients with NSCLC is still poor despite the enormous efforts that have been made to develop novel treatments. Understanding fundamental molecular and genetic mechanisms is necessary to develop new therapeutic approaches for NSCLC. A recently identified type of programmed cell death known as ferroptosis is one potential approach. Ferroptosis causes oxidative damage and the death of cancerous cells by peroxidizing unsaturated phospholipids and accumulating reactive oxygen species (ROS) in an iron-dependent manner. Ferroptosis-related gene (FRG) signatures have recently been evaluated for their ability to predict patient OS and prognosis. These analyses show FRGs are involved in cancer progression, and may serve as promising biomarkers for tumor diagnosis and therapy. Moreover, we summarize the current pharmaceutical options of ferroptosis induction and their underlying molecular mechanism in LC. Therefore, this review aims to provide a comprehensive summary of FRG-based prognostic models, their associated metabolic and signaling pathways, and promising therapeutic options for ferroptosis induction in NSCLC.
Collapse
Affiliation(s)
- Rohil Jawed
- Department of Biomedical Engineering, College of Engineering and Applied Sciences, Nanjing University, Nanjing, 210023, China.
| | - Huma Bhatti
- School of Chemistry and Chemical Engineering, State Key Laboratory of Analytical Chemistry for Life Science, Nanjing University, Nanjing, 210023, China
| | - Adnan Khan
- Clinical and Molecular Labs, Karachi Institute of Radiotherapy and Nuclear Medicine (KIRAN), KDA Scheme 33 Near Safoora Chowk, Karachi, Pakistan
| |
Collapse
|
8
|
Prachayakul A, Sucharitakul J, Chadsiri J, Prapinjumrune C. Salivary iron and Candida colonization in oral lichen planus patients undergoing topical steroid therapy. BMC Oral Health 2025; 25:589. [PMID: 40251548 PMCID: PMC12008899 DOI: 10.1186/s12903-025-05950-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2024] [Accepted: 04/04/2025] [Indexed: 04/20/2025] Open
Abstract
BACKGROUND The most common adverse event in oral lichen planus (OLP) patients undergoing topical corticosteroid therapy is opportunistic infection with oral Candida. Oral candidiasis superimposed on OLP can worsen the patient's symptoms and obscure the true clinical appearance making the treatment of OLP more complex and difficult. The aim of this study was to investigate the salivary iron level in OLP patients undergoing topical steroid therapy with a history of oral candidiasis compared with OLP patients undergoing topical steroid therapy without a history of oral candidiasis and healthy individuals. MATERIALS AND METHODS The 68 OLP patients treated with a topical steroid were divided into 34 OLP patients with a history of oral candidiasis, 34 OLP patients without a history of oral candidiasis and 34 healthy individuals were enrolled in the study. Unstimulated whole saliva was collected from the participants to determine the salivary iron level and to investigate the presence of oral Candida colonization. RESULTS The salivary iron level was significantly higher in OLP patients undergoing topical steroid therapy with a history of oral candidiasis compared with OLP patients undergoing topical steroid therapy without a history of oral candidiasis and healthy individuals (P = 0.04 and P < 0.001, respectively). Moreover, the Candida colony count in OLP patients undergoing topical steroid therapy with a history of oral candidiasis was significantly higher compared with OLP patients undergoing topical steroid therapy without a history of oral candidiasis and healthy Individuals (P < 0.001 and P < 0.001, respectively). CONCLUSION High salivary iron level associates with a high amount of oral Candida colonization in OLP patients undergoing topical steroid therapy with a history of oral candidiasis. TRIAL REGISTRATION This study was registered at the Thai Clinical Trials Registry on 2nd June 2022 (TCTR identifier: TCTR20220602006).
Collapse
Affiliation(s)
- Auranat Prachayakul
- Department of Oral Medicine, Faculty of Dentistry, Chulalongkorn University, Bangkok, Thailand
- Phakhao Hospital, Loei, Thailand
| | - Jeerus Sucharitakul
- Department of Biochemistry and Center of Excellence in Integrative Immuno-Microbial Biochemistry and Bioresponsive Nanomaterials, Faculty of Dentistry, Chulalongkorn University, Bangkok, Thailand
| | - Jaranya Chadsiri
- Department of Oral Medicine, Faculty of Dentistry, Chulalongkorn University, Bangkok, Thailand
| | - Chanwit Prapinjumrune
- Department of Oral Medicine, Faculty of Dentistry, Chulalongkorn University, Bangkok, Thailand.
| |
Collapse
|
9
|
Yang Y, Deng X, Li W, Leng Y, Xiong Y, Wang B, Gong S, Wang Y, Yang B, Li W. Targeting the epigenetic regulation of ferroptosis: a potential therapeutic approach for sepsis-associated acute kidney injury. Clin Epigenetics 2025; 17:57. [PMID: 40189571 PMCID: PMC11974148 DOI: 10.1186/s13148-025-01861-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2024] [Accepted: 03/13/2025] [Indexed: 04/09/2025] Open
Abstract
Sepsis is a syndrome of organ dysfunction caused by the invasion of pathogenic microorganisms. In clinical practice, patients with sepsis are prone to concurrent acute kidney injury, which has high morbidity and mortality rates. Thus, understanding the pathogenesis of sepsis-associated acute kidney injury is of significant clinical importance. Ferroptosis is an iron-dependent programmed cell death pathway, which is proved to play a critical role in the process of sepsis-associated acute kidney injury through various mechanisms. Epigenetic regulation modulates the content and function of nucleic acids and proteins within cells through various modifications. Its impact on ferroptosis has garnered increasing attention; however, the role of epigenetic regulation targeting ferroptosis in sepsis-associated acute kidney injury has not been fully elucidated. Growing evidence suggests that epigenetic regulation can modulate ferroptosis through complex pathway networks, thereby affecting the development and prognosis of sepsis-associated acute kidney injury. This paper summarizes the impact of ferroptosis on sepsis-associated acute kidney injury and the regulatory mechanisms of epigenetic regulation on ferroptosis, providing new insights for the targeted therapy of sepsis-associated acute kidney injury.
Collapse
Affiliation(s)
- Yuhang Yang
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei, China
| | - Xinqi Deng
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei, China
| | - Wenyuan Li
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei, China
| | - Yan Leng
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei, China
| | - Yonghong Xiong
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei, China
| | - Bihan Wang
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei, China
| | - Siyuan Gong
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei, China
| | - Yunhao Wang
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei, China
| | - Baichuan Yang
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei, China
| | - Wei Li
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei, China.
| |
Collapse
|
10
|
Wang R, Huang Y, Shi Y, Zhao Z. Transferrin from obscure puffer Takifugu obscurus promotes antimicrobial immune response. FISH & SHELLFISH IMMUNOLOGY 2025; 159:110184. [PMID: 39947339 DOI: 10.1016/j.fsi.2025.110184] [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: 11/28/2024] [Revised: 02/03/2025] [Accepted: 02/08/2025] [Indexed: 02/17/2025]
Abstract
Transferrin (TF) is an iron-binding glycoprotein that plays a crucial role in host defense by restricting iron availability to pathogens and activating antimicrobial immune responses. However, its antimicrobial role in fish remains largely unexplored. In this study, we identified and characterized transferrin from obscure puffer Takifugu obscurus, named ToTF, and investigated its functions in bacterial infections. The coding region of the ToTF gene consists of 2295 bp, encoding a polypeptide of 764 amino acids. Tissue distribution analysis revealed that ToTF is primarily expressed in the liver. The mRNA expression of ToTF was significantly upregulated in both the liver and kidney following challenge with Staphylococcus aureus and Vibrio harveyi. Moreover, recombinant ToTF (rToTF) markedly inhibits iron-dependent bacteria growth, including S. aureus and V. harveyi. The iron-binding capacity of rToTF peaked at pH 8 and decreased significantly under acidic conditions. Furthermore, the antibacterial activity of rToTF peaked at near-neutral pH but decreased significantly at pH below 5. This dual mechanism deprives bacteria of essential iron through high-affinity binding and directly inhibits their growth under optimal conditions. These findings highlight the dual role of ToTF in the antibacterial immunity of T. obscurus, mediated by iron sequestration and direct bacteriostatic effects. This study supports the development of TF-based antibacterial agents and enhances strategies for aquatic disease prevention and fish disease resistance.
Collapse
Affiliation(s)
- Ruixia Wang
- Jiangsu Province Engineering Research Center for Marine Bio-resources Sustainable Utilization, College of Oceanography, Hohai University, Nanjing, 210024, China
| | - Ying Huang
- Jiangsu Province Engineering Research Center for Marine Bio-resources Sustainable Utilization, College of Oceanography, Hohai University, Nanjing, 210024, China
| | - Yan Shi
- Jiangsu Province Engineering Research Center for Marine Bio-resources Sustainable Utilization, College of Oceanography, Hohai University, Nanjing, 210024, China
| | - Zhe Zhao
- Jiangsu Province Engineering Research Center for Marine Bio-resources Sustainable Utilization, College of Oceanography, Hohai University, Nanjing, 210024, China.
| |
Collapse
|
11
|
Izuka S, Komai T, Tsuchida Y, Tsuchiya H, Okamura T, Fujio K. The role of monocytes and macrophages in idiopathic inflammatory myopathies: insights into pathogenesis and potential targets. Front Immunol 2025; 16:1567833. [PMID: 40181992 PMCID: PMC11965591 DOI: 10.3389/fimmu.2025.1567833] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2025] [Accepted: 03/03/2025] [Indexed: 04/05/2025] Open
Abstract
Idiopathic inflammatory myopathies (IIMs) are heterogeneous autoimmune disorders characterized by muscle inflammation, weakness, and extramuscular manifestations such as interstitial lung disease, skin rash, arthritis, dysphagia, myocarditis and other systemic organ involvement. Although T and B cells have historically been central to the understanding of IIM immunopathology, monocytes and their differentiated progenitor cells, macrophages, are increasingly being recognized as critical mediators of both tissue damage and repair. In subtypes such as dermatomyositis, immune-mediated necrotizing myopathy and antisynthetase syndrome, macrophages infiltrate skeletal muscle and other affected tissues, contributing to inflammation via production of pro-inflammatory cytokines, chemokines, and reactive oxygen species. Dysregulated interferon signaling, mitochondrial stress, and aberrant metabolic states in these cells further perpetuate tissue injury in IIMs. Conversely, certain macrophage subsets can support muscle fiber regeneration and dampen inflammation, underscoring the dual roles these cells can play. Future research into the heterogeneity of monocytes and macrophages, including single-cell transcriptomic and metabolomic approaches, will help clarify disease mechanisms, identify biomarkers of disease activity and prognosis, and guide novel therapeutic strategies targeting these innate immune cells in IIM.
Collapse
Affiliation(s)
- Shinji Izuka
- Department of Allergy and Rheumatology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Toshihiko Komai
- Department of Allergy and Rheumatology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Yumi Tsuchida
- Department of Allergy and Rheumatology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Haruka Tsuchiya
- Department of Allergy and Rheumatology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Tomohisa Okamura
- Department of Allergy and Rheumatology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
- Department of Functional Genomics and Immunological Diseases, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Keishi Fujio
- Department of Allergy and Rheumatology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| |
Collapse
|
12
|
Wang L, Ju C, Han C, Yu Z, Bai MY, Wang C. The interaction of nutrient uptake with biotic and abiotic stresses in plants. JOURNAL OF INTEGRATIVE PLANT BIOLOGY 2025; 67:455-487. [PMID: 39783785 DOI: 10.1111/jipb.13827] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/15/2024] [Accepted: 11/21/2024] [Indexed: 01/12/2025]
Abstract
Plants depend heavily on efficient nutrient uptake and utilization for optimal growth and development. However, plants are constantly subjected to a diverse array of biotic stresses, such as pathogen infections, insect pests, and herbivory, as well as abiotic stress like drought, salinity, extreme temperatures, and nutrient imbalances. These stresses significantly impact the plant's ability to take up nutrient and use it efficiency. Understanding how plants maintain nutrient uptake and use efficiency under biotic and abiotic stress conditions is crucial for improving crop resilience and sustainability. This review explores the recent advancements in elucidating the mechanisms underlying nutrient uptake and utilization efficiency in plants under such stress conditions. Our aim is to offer a comprehensive perspective that can guide the breeding of stress-tolerant and nutrition-efficient crop varieties, ultimately contributing to the advancement of sustainable agriculture.
Collapse
Affiliation(s)
- Lingyan Wang
- The Key Laboratory of Plant Development and Environmental Adaptation Biology, Ministry of Education, School of Life Sciences, Shandong University, Qingdao, 266237, China
| | - Chuanfeng Ju
- State Key Laboratory for Crop Stress Resistance and High-Efficiency Production, College of Life Sciences, Northwest A&F University, Yangling, 712100, Shaanxi, China
| | - Chao Han
- The Key Laboratory of Plant Development and Environmental Adaptation Biology, Ministry of Education, School of Life Sciences, Shandong University, Qingdao, 266237, China
| | - Zhenghao Yu
- State Key Laboratory for Crop Stress Resistance and High-Efficiency Production, College of Life Sciences, Northwest A&F University, Yangling, 712100, Shaanxi, China
| | - Ming-Yi Bai
- The Key Laboratory of Plant Development and Environmental Adaptation Biology, Ministry of Education, School of Life Sciences, Shandong University, Qingdao, 266237, China
| | - Cun Wang
- State Key Laboratory for Crop Stress Resistance and High-Efficiency Production, College of Life Sciences, Northwest A&F University, Yangling, 712100, Shaanxi, China
| |
Collapse
|
13
|
Ren Z, Zhang L, Li H, Yang M, Wu X, Hu R, Lu J, Wang H, Wu X, Wang Z, Li X. The BRUTUS iron sensor and E3 ligase facilitates soybean root nodulation by monoubiquitination of NSP1. NATURE PLANTS 2025; 11:595-611. [PMID: 39900829 DOI: 10.1038/s41477-024-01896-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/16/2024] [Accepted: 12/05/2024] [Indexed: 02/05/2025]
Abstract
Legumes form root nodules with symbiotic nitrogen-fixing rhizobacteria, which require ample iron to ensure symbiosis establishment and efficient nitrogen fixation. The functions and mechanisms of iron in nitrogen-fixing nodules are well established. However, the role of iron and the mechanisms by which legumes sense iron and incorporate this cue into nodulation signalling pathways remain unclear. Here we show that iron is a key driver of nodulation because symbiotic nodules cannot form without iron, even under conditions of sufficient light and low nitrogen. We further identify an iron optimum for soybean nodulation and the iron sensor BRUTUS A (BTSa) which acts as a hub for integrating iron and nodulation cues. BTSa is induced by rhizobia, binds to and is stabilized by iron. In turn, BTSa stabilizes and enhances the transcriptional activation activity of pro-nodulation transcription factor NSP1a by monoubiquitination from its RING domain and consequently activates nodulation signalling. Monoubiquitination of NSP1 by BTS is conserved in legumes to trigger nodulation under iron sufficiency. Thus, iron status is an essential cue to trigger nodulation and BTSa integrates cues from rhizobial infection and iron status to orchestrate host responses towards establishing symbiotic nitrogen fixation.
Collapse
Affiliation(s)
- Ziyin Ren
- National Key Laboratory of Crop Genetic Improvement, College of Plant Science and Technology, Huazhong Agricultural University, Wuhan, China
| | - Ling Zhang
- Jilin Provincial Key Laboratory of Agricultural Biotechnology, Jilin Academy of Agricultural Sciences, Changchun, China
| | - Haizhen Li
- National Key Laboratory of Crop Genetic Improvement, College of Plant Science and Technology, Huazhong Agricultural University, Wuhan, China
| | - Mi Yang
- National Key Laboratory of Crop Genetic Improvement, College of Plant Science and Technology, Huazhong Agricultural University, Wuhan, China
| | - Xuesong Wu
- National Key Laboratory of Crop Genetic Improvement, College of Plant Science and Technology, Huazhong Agricultural University, Wuhan, China
| | - Runxu Hu
- National Key Laboratory of Crop Genetic Improvement, College of Plant Science and Technology, Huazhong Agricultural University, Wuhan, China
| | - Jingjing Lu
- National Key Laboratory of Crop Genetic Improvement, College of Plant Science and Technology, Huazhong Agricultural University, Wuhan, China
| | - Hui Wang
- National Key Laboratory of Crop Genetic Improvement, College of Plant Science and Technology, Huazhong Agricultural University, Wuhan, China
| | - Xinying Wu
- National Key Laboratory of Crop Genetic Improvement, College of Plant Science and Technology, Huazhong Agricultural University, Wuhan, China
| | - Zhijuan Wang
- National Key Laboratory of Crop Genetic Improvement, College of Plant Science and Technology, Huazhong Agricultural University, Wuhan, China.
| | - Xia Li
- National Key Laboratory of Crop Genetic Improvement, College of Plant Science and Technology, Huazhong Agricultural University, Wuhan, China.
- Hubei Hongshan Laboratory, Wuhan, China.
| |
Collapse
|
14
|
Viel S, Vivier E, Walzer T, Marçais A. Targeting metabolic dysfunction of CD8 T cells and natural killer cells in cancer. Nat Rev Drug Discov 2025; 24:190-208. [PMID: 39668206 DOI: 10.1038/s41573-024-01098-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/06/2024] [Indexed: 12/14/2024]
Abstract
The importance of metabolic pathways in regulating immune responses is now well established, and a mapping of the bioenergetic metabolism of different immune cell types is under way. CD8 T cells and natural killer (NK) cells contribute to cancer immunosurveillance through their cytotoxic functions and secretion of cytokines and chemokines, complementing each other in target recognition mechanisms. Several immunotherapies leverage these cell types by either stimulating their activity or redirecting their specificity against tumour cells. However, the anticancer activity of CD8 T cells and NK cells is rapidly diminished in the tumour microenvironment, closely linked to a decline in their metabolic capacities. Various strategies have been developed to restore cancer immunosurveillance, including targeting bioenergetic metabolism or genetic engineering. This Review provides an overview of metabolic dysfunction in CD8 T cells and NK cells within the tumour microenvironment, highlighting current therapies aiming to overcome these issues.
Collapse
Affiliation(s)
- Sébastien Viel
- Plateforme de Biothérapie et de Production de Médicaments de Thérapie Innovante, Hôpital Edouard Herriot, Hospices Civils de Lyon, Lyon, France
| | - Eric Vivier
- Innate Pharma Research Laboratories, Innate Pharma, Marseille, France
- Aix Marseille University, CNRS, INSERM, CIML, Marseille, France
- APHM, Hôpital de la Timone, Marseille, France
- Paris Saclay Cancer Cluster, Villejuif, France
- Université Paris-Saclay, Gustave Roussy, Inserm, Prédicteurs moléculaires et nouvelles cibles en oncologie, Villejuif, France
| | - Thierry Walzer
- CIRI, Centre International de Recherche en Infectiologie, (Team Lyacts), Univ Lyon, Inserm, U1111, Université Claude Bernard Lyon 1, CNRS UMR5308 ENS de Lyon, Lyon, France
| | - Antoine Marçais
- CIRI, Centre International de Recherche en Infectiologie, (Team Lyacts), Univ Lyon, Inserm, U1111, Université Claude Bernard Lyon 1, CNRS UMR5308 ENS de Lyon, Lyon, France.
| |
Collapse
|
15
|
Duarte-Silva E, Maes M, Alves Peixoto C. Iron metabolism dysfunction in neuropsychiatric disorders: Implications for therapeutic intervention. Behav Brain Res 2025; 479:115343. [PMID: 39557130 DOI: 10.1016/j.bbr.2024.115343] [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: 09/19/2024] [Revised: 11/13/2024] [Accepted: 11/15/2024] [Indexed: 11/20/2024]
Abstract
Iron is a trace metal that takes part in the maintenance of body homeostasis by, for instance, aiding in energy production and immunity. A body of evidence now demonstrates that dysfunction in iron metabolism can have detrimental effects and is intricately associated with the development of neuropsychiatric disorders, including Major Depressive Disorder (MDD), anxiety, and schizophrenia. For instance, changes in serum and central nervous system (CNS) levels of iron and in proteins mediating iron metabolism have been documented in patients grappling with the aforementioned diseases. By contrast, targeting iron metabolism by using iron chelators, for instance, has proven to be effective in alleviating disease burden. Therefore, here we review the state-of-the-art regarding the role of iron metabolism and its dysfunction in the context of neuropsychiatric disorders. Furthermore, we discuss how targeting iron metabolism can be an effective therapeutic option to tackle this class of diseases. Finally, we discuss the mechanisms linking this dysfunction to behavioral changes in these disorders. Harnessing the knowledge of iron metabolism is not only key to the characterization of novel molecular targets and disease biomarkers but also crucial to drug repurposing and drug design.
Collapse
Affiliation(s)
- Eduardo Duarte-Silva
- Center for Research in Inflammatory Diseases (CRID), Ribeirão Preto Medical School, Department of Pharmacology, University of São Paulo, São Paulo, Brazil; Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), Ribeirão Preto, SP, Brazil.
| | - Michael Maes
- Mental Health Center, University of Electronic Science and Technology of China, Chengdu 611731, China; Deakin University, IMPACT, the Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Barwon Health, Geelong, VIC, Australia; Research Institute, Medical University of Plovdiv, Plovdiv 4002, Bulgaria; Department of Psychiatry, Medical University of Plovdiv, Plovdiv 4002, Bulgaria; Cognitive Impairment and Dementia Research Unit, Faculty of Medicine, Chulalongkorn University, Bangkok 10330, Thailand; Department of Psychiatry, Faculty of Medicine, Chulalongkorn University, Bangkok 10330, Thailand
| | - Christina Alves Peixoto
- Laboratory of Ultrastructure, Aggeu Magalhães Institute (IAM), PE, Brazil; National Institute of Science and Technology on Neuroimmunomodulation (INCT-NIM), Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil.
| |
Collapse
|
16
|
Li Y, Pham T, Hipsher K, Lee CWJ, Jiao J, Penninger JM, Kronstad JW, Fan Y, Zhao Y, Ambati S, Meagher RB, Xie X, Lin X. Identification of a protective antigen reveals the trade-off between iron acquisition and antigen exposure in a global fungal pathogen. Proc Natl Acad Sci U S A 2025; 122:e2420898122. [PMID: 39946532 PMCID: PMC11848283 DOI: 10.1073/pnas.2420898122] [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: 10/17/2024] [Accepted: 01/09/2025] [Indexed: 02/26/2025] Open
Abstract
Systemic infections caused by Cryptococcus claim over 161,000 lives annually, with global mortality rate close to 70% despite antifungal therapies. Currently, no vaccine is available. To develop an effective multivalent vaccine against this free-living opportunistic eukaryotic pathogen, it is critical to identify protective antigens. We previously discovered ZNF2oe strains elicit protective host immune responses and increase the abundance of antigens present in the capsule, which is required for its immunoprotection. Capsule is a defining feature of Cryptococcus species and composed of polysaccharides and mannoproteins. Here, we found increased levels of exposed mannoproteins in ZNF2oe cells. As mannoproteins are the primary components recognized by anticryptococcal cell-mediated immune responses and few have been characterized, we systemically screened all 49 predicted GPI-mannoproteins in Cryptococcus neoformans for enhanced host recognition. We identified those highly present in ZNF2oe cells and found Cig1 to be a protective antigen against cryptococcosis either as a recombinant protein vaccine or an mRNA vaccine. Cig1 is induced by iron limitation and is highly expressed by this fungus in infected mice and in patients with cryptococcal meningitis. Remarkably, iron restriction by the host induces cryptococcal cells to express iron-uptake proteins including Cig1, which act as cryptococcal antigens and in turn enhance host detection. Our results highlight an arms race between the pathogen and the host centered on iron competition, and the trade-off between cryptococcal iron acquisition and antigen exposure. These findings demonstrate the potential of leveraging this host-pathogen interaction for vaccine development.
Collapse
Affiliation(s)
- Yeqi Li
- Department of Microbiology, University of Georgia, Athens, GA30602
| | - Tuyetnhu Pham
- Department of Plant Biology, University of Georgia, Athens, GA30602
| | - Kenton Hipsher
- Department of Microbiology, University of Georgia, Athens, GA30602
| | - Christopher W. J. Lee
- Michael Smith Laboratories, Department of Microbiology and Immunology, University of British Columbia, Vancouver, BCV6T 1Z4, Canada
| | - Jie Jiao
- Department of Medical Genetics, Life Sciences Institute, University of British Columbia, Vancouver, BCV6T 1Z3, Canada
| | - Josef M. Penninger
- Department of Medical Genetics, Life Sciences Institute, University of British Columbia, Vancouver, BCV6T 1Z3, Canada
- Eric Kandel Institute, Department of Laboratory Medicine, Medical University of Vienna, Vienna1090, Austria
- Helmholtz Centre for Infection Research, Braunschweig38124, Germany
- Institute of Molecular Biotechnology of the Austrian Academy of Sciences, Vienna1030, Austria
| | - James W. Kronstad
- Michael Smith Laboratories, Department of Microbiology and Immunology, University of British Columbia, Vancouver, BCV6T 1Z4, Canada
| | - Yumeng Fan
- Department of Microbiology, University of Georgia, Athens, GA30602
| | - Youbao Zhao
- Department of Microbiology, University of Georgia, Athens, GA30602
| | - Suresh Ambati
- Department of Genetics, University of Georgia, Athens, GA30602
| | | | - Xiaofeng Xie
- Department of Microbiology, University of Georgia, Athens, GA30602
| | - Xiaorong Lin
- Department of Microbiology, University of Georgia, Athens, GA30602
- Department of Plant Biology, University of Georgia, Athens, GA30602
| |
Collapse
|
17
|
Chao ZF, Chao DY. Barriers and carriers for transition metal homeostasis in plants. PLANT COMMUNICATIONS 2025; 6:101235. [PMID: 39731291 PMCID: PMC11897463 DOI: 10.1016/j.xplc.2024.101235] [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: 09/29/2024] [Revised: 12/02/2024] [Accepted: 12/25/2024] [Indexed: 12/29/2024]
Abstract
Transition metals are types of metals with high chemical activity. They play critical roles in plant growth, development, reproduction, and environmental adaptation, as well as in human health. However, the acquisition, transport, and storage of these metals pose specific challenges due to their high reactivity and poor solubility. In addition, distinct yet interconnected apoplastic and symplastic diffusion barriers impede their movement throughout plants. To overcome these obstacles, plants have evolved sophisticated carrier systems to facilitate metal transport, relying on the tight coordination of vesicles, enzymes, metallochaperones, low-molecular-weight metal ligands, and membrane transporters for metals, ligands, and metal-ligand complexes. This review highlights recent advances in the homeostasis of transition metals in plants, focusing on the barriers to transition metal transport and the carriers that facilitate their passage through these barriers.
Collapse
Affiliation(s)
- Zhen-Fei Chao
- National Key Laboratory of Plant Molecular Genetics, CAS Center for Excellence in Molecular Plant Sciences, Shanghai Institute of Plant Physiology and Ecology, Chinese Academy of Sciences, Shanghai 200032, China; Leibniz Institute of Plant Genetics & Crop Plant Research (IPK) OT Gatersleben, Corrensstr 3, 06466 Seeland, Germany
| | - Dai-Yin Chao
- National Key Laboratory of Plant Molecular Genetics, CAS Center for Excellence in Molecular Plant Sciences, Shanghai Institute of Plant Physiology and Ecology, Chinese Academy of Sciences, Shanghai 200032, China.
| |
Collapse
|
18
|
Qian X, Zhou Q, Ouyang Y, Wu X, Sun X, Wang S, Duan Y, Hu Z, Hou Y, Wang Z, Chen X, Wang KL, Shen Y, Dong B, Lin Y, Wen T, Tian Q, Guo Z, Li M, Xiao L, Wu Q, Meng Y, Liu G, Ying H, Zhou Y, Zhang W, Duan S, Bai X, Liu T, Zhan P, Lu Z, Xu D. Transferrin promotes fatty acid oxidation and liver tumor growth through PHD2-mediated PPARα hydroxylation in an iron-dependent manner. Proc Natl Acad Sci U S A 2025; 122:e2412473122. [PMID: 39888917 PMCID: PMC11804496 DOI: 10.1073/pnas.2412473122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2024] [Accepted: 01/02/2025] [Indexed: 02/02/2025] Open
Abstract
Tumor cells reshape iron and lipid metabolism for their rapid proliferation. However, how tumor cells coordinate the interplay between tumor cell-specific iron homeostasis and lipid metabolism reprogramming to counteract energy shortages remains unclear. Here, we demonstrated that glucose deprivation in hepatocellular carcinoma (HCC) cells induced AMPK-dependent Transferrin S685 phosphorylation, which exposed Transferrin nuclear localization signal (NLS) for binding to importin α7 and subsequent nuclear translocation. Nucleus-translocated Transferrin interacts with PPARα and enhance its protein stability to increase fatty acid oxidation (FAO) upon glucose deprivation. Mechanistically, PPARα-associated Transferrin upregulates iron-dependent PHD2-mediated PPARα P87 hydroxylation and subsequently disrupts the binding of MDM2 to PPARα, therefore inhibiting MDM2-mediated PPARα ubiquitination and degradation. Reconstitution of Transferrin S685A and NLS mutation or knock-in expression of PPARα P87A inhibited PPARα-mediated FAO upon energy stress, enhanced HCC cell apoptosis, and impeded liver tumor growth in mice. Importantly, combined treatment with Transferrin pS685 blocking peptide suppressing AMPK-Transferrin-PPARα axis could synergize with a well-established AMPK activator Metformin to inhibit tumor growth. Additionally, Transferrin pS685-mediated PPARα P87 hydroxylation is positively correlated with PPARα expression levels in human HCC specimens and poor patient prognosis. These findings revealed a mechanism by which Transferrin can sense energy stress to promote the hydroxylation and protein stability of PPARα through iron-dependent activation of PHD2 and underscore the moonlighting function of Transferrin in lipid catabolism and liver tumor development.
Collapse
Affiliation(s)
- Xu Qian
- Department of Clinical Laboratory, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine, Chinese Academy of Sciences, Hangzhou310022, China
- Zhejiang Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Zhejiang Key Laboratory of Frontier Medical Research on Cancer Metabolism, and Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, Zhejiang310029, China
- Institute of Fundamental and Transdisciplinary Research, Cancer Center, Zhejiang University, Hangzhou, Zhejiang310029, China
| | - Qimin Zhou
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai200011, China
| | - Yuan Ouyang
- Laboratory of Oral Microbiota and Systemic Diseases, Shanghai Ninth People’s Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai200125, China
- National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Shanghai, China
| | - Xiaohong Wu
- National Health Commission (NHC) Key Laboratory of Molecular Probe and Targeted Theranostics, Harbin Medical University, Harbin, Heilongjiang150081, China
| | - Xue Sun
- Department of Surgical Oncology, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang150081, China
| | - Shuo Wang
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology, Ministry of Education, School of Pharmaceutical Sciences, Shandong University, Jinan, Shandong250012, China
| | - Yuran Duan
- Zhejiang Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Zhejiang Key Laboratory of Frontier Medical Research on Cancer Metabolism, and Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, Zhejiang310029, China
- Institute of Fundamental and Transdisciplinary Research, Cancer Center, Zhejiang University, Hangzhou, Zhejiang310029, China
| | - Zhiqiang Hu
- Zhejiang Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Zhejiang Key Laboratory of Frontier Medical Research on Cancer Metabolism, and Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, Zhejiang310029, China
- Institute of Fundamental and Transdisciplinary Research, Cancer Center, Zhejiang University, Hangzhou, Zhejiang310029, China
| | - Yueru Hou
- Zhejiang Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Zhejiang Key Laboratory of Frontier Medical Research on Cancer Metabolism, and Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, Zhejiang310029, China
- Institute of Fundamental and Transdisciplinary Research, Cancer Center, Zhejiang University, Hangzhou, Zhejiang310029, China
| | - Zheng Wang
- Zhejiang Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Zhejiang Key Laboratory of Frontier Medical Research on Cancer Metabolism, and Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, Zhejiang310029, China
- Institute of Fundamental and Transdisciplinary Research, Cancer Center, Zhejiang University, Hangzhou, Zhejiang310029, China
| | - Xiaohan Chen
- Department of Surgical Oncology, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang150081, China
| | | | - Yuli Shen
- Zhejiang Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Zhejiang Key Laboratory of Frontier Medical Research on Cancer Metabolism, and Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, Zhejiang310029, China
- Institute of Fundamental and Transdisciplinary Research, Cancer Center, Zhejiang University, Hangzhou, Zhejiang310029, China
| | - Bofei Dong
- Zhejiang Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Zhejiang Key Laboratory of Frontier Medical Research on Cancer Metabolism, and Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, Zhejiang310029, China
- Institute of Fundamental and Transdisciplinary Research, Cancer Center, Zhejiang University, Hangzhou, Zhejiang310029, China
| | - Yanni Lin
- Zhejiang Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Zhejiang Key Laboratory of Frontier Medical Research on Cancer Metabolism, and Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, Zhejiang310029, China
- Institute of Fundamental and Transdisciplinary Research, Cancer Center, Zhejiang University, Hangzhou, Zhejiang310029, China
| | - Ting Wen
- Zhejiang Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Zhejiang Key Laboratory of Frontier Medical Research on Cancer Metabolism, and Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, Zhejiang310029, China
- Institute of Fundamental and Transdisciplinary Research, Cancer Center, Zhejiang University, Hangzhou, Zhejiang310029, China
| | - Qi Tian
- Zhejiang Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Zhejiang Key Laboratory of Frontier Medical Research on Cancer Metabolism, and Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, Zhejiang310029, China
- Institute of Fundamental and Transdisciplinary Research, Cancer Center, Zhejiang University, Hangzhou, Zhejiang310029, China
| | - Zhanpeng Guo
- Zhejiang Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Zhejiang Key Laboratory of Frontier Medical Research on Cancer Metabolism, and Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, Zhejiang310029, China
- Institute of Fundamental and Transdisciplinary Research, Cancer Center, Zhejiang University, Hangzhou, Zhejiang310029, China
| | - Min Li
- Zhejiang Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Zhejiang Key Laboratory of Frontier Medical Research on Cancer Metabolism, and Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, Zhejiang310029, China
- Institute of Fundamental and Transdisciplinary Research, Cancer Center, Zhejiang University, Hangzhou, Zhejiang310029, China
| | - Liwei Xiao
- Zhejiang Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Zhejiang Key Laboratory of Frontier Medical Research on Cancer Metabolism, and Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, Zhejiang310029, China
- Institute of Fundamental and Transdisciplinary Research, Cancer Center, Zhejiang University, Hangzhou, Zhejiang310029, China
| | - Qingang Wu
- Zhejiang Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Zhejiang Key Laboratory of Frontier Medical Research on Cancer Metabolism, and Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, Zhejiang310029, China
- Institute of Fundamental and Transdisciplinary Research, Cancer Center, Zhejiang University, Hangzhou, Zhejiang310029, China
| | - Ying Meng
- Zhejiang Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Zhejiang Key Laboratory of Frontier Medical Research on Cancer Metabolism, and Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, Zhejiang310029, China
- Institute of Fundamental and Transdisciplinary Research, Cancer Center, Zhejiang University, Hangzhou, Zhejiang310029, China
| | - Guijun Liu
- Zhejiang Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Zhejiang Key Laboratory of Frontier Medical Research on Cancer Metabolism, and Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, Zhejiang310029, China
- Institute of Fundamental and Transdisciplinary Research, Cancer Center, Zhejiang University, Hangzhou, Zhejiang310029, China
| | - Hangjie Ying
- Department of Clinical Laboratory, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine, Chinese Academy of Sciences, Hangzhou310022, China
| | - Yahui Zhou
- Department of Clinical Laboratory, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine, Chinese Academy of Sciences, Hangzhou310022, China
| | - Wuchang Zhang
- Laboratory of Oral Microbiota and Systemic Diseases, Shanghai Ninth People’s Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai200125, China
- National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Shanghai, China
| | - Shengzhong Duan
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Engineering Research Center of Oral Biomaterials and Devices of Zhejiang Province, Hangzhou310000, China
| | - Xueli Bai
- Zhejiang Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Zhejiang Key Laboratory of Frontier Medical Research on Cancer Metabolism, and Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, Zhejiang310029, China
| | - Tong Liu
- Department of Surgical Oncology, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang150081, China
- National Health Commission (NHC) Key Laboratory of Cell Transplantation, Harbin Medical University, Harbin, Heilongjiang150081, China
| | - Peng Zhan
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology, Ministry of Education, School of Pharmaceutical Sciences, Shandong University, Jinan, Shandong250012, China
| | - Zhimin Lu
- Zhejiang Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Zhejiang Key Laboratory of Frontier Medical Research on Cancer Metabolism, and Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, Zhejiang310029, China
- Institute of Fundamental and Transdisciplinary Research, Cancer Center, Zhejiang University, Hangzhou, Zhejiang310029, China
| | - Daqian Xu
- Zhejiang Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Zhejiang Key Laboratory of Frontier Medical Research on Cancer Metabolism, and Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, Zhejiang310029, China
- Institute of Fundamental and Transdisciplinary Research, Cancer Center, Zhejiang University, Hangzhou, Zhejiang310029, China
- National Health Commission (NHC) Key Laboratory of Cell Transplantation, Harbin Medical University, Harbin, Heilongjiang150081, China
| |
Collapse
|
19
|
Kunitomo Y, Putcha N, Fawzy A, Raju S, McCormack MC, Wise RA, Hansel NN, Balasubramanian A. Iron Deficiency and All-Cause Hospitalization Risk in a Clinical Cohort of COPD. CHRONIC OBSTRUCTIVE PULMONARY DISEASES (MIAMI, FLA.) 2025; 12:72-81. [PMID: 39879352 PMCID: PMC11925066 DOI: 10.15326/jcopdf.2024.0550] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/31/2025]
Abstract
Background The impact of iron deficiency on COPD morbidity independent of anemia status is unknown. Understanding the association between iron deficiency, anemia status, and risk of hospitalization in COPD may inform an approach to these comorbidities. Study Design and Methods Adults ≥40 years from the Johns Hopkins COPD Precision Medicine Center of Excellence data repository with an outpatient iron profile and 1 year of subsequent follow-up time were included in the study. Baseline characteristics were compared across iron status, defined by transferrin saturation (TSAT), using t-tests and Chi-squared tests. The association between continuous TSAT and all-cause hospitalization over the 1-year follow-up period was assessed by logistic regression. Models were adjusted by covariates with an interaction term for anemia and stratified by sex. Results There were 6532 individuals included with an average age of 65±12 years, 59% were female, and 56% White. Fifty-two percent of the cohort were iron deficient (TSAT≤20%), among whom 27% were non-anemic. Iron-deficient individuals had lower lung function and a higher prevalence of heart failure and diabetes. Iron deficiency was more prevalent among females (57%) compared to males (44%). In adjusted models, a decrease in TSAT by 10% was associated with 14.3% higher odds of all-cause hospitalization for females (95%CI:1.0-1.3), but not among males (OR:1.08, 95%CI:0.9-1.3). There was effect modification by anemia such that the association between TSAT and all-cause hospitalization was greater in non-anemic women (p-value interaction=0.08). Interpretation Iron deficiency may be associated with adverse outcomes in the absence of anemia, with non-anemic women being a COPD sub-population particularly sensitive to iron deficiency.
Collapse
Affiliation(s)
- Yukiko Kunitomo
- Division of Pulmonary and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States
| | - Nirupama Putcha
- Division of Pulmonary and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States
| | - Ashraf Fawzy
- Division of Pulmonary and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States
| | - Sarath Raju
- Division of Pulmonary and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States
| | - Meredith C McCormack
- Division of Pulmonary and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States
| | - Robert A Wise
- Division of Pulmonary and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States
| | - Nadia N Hansel
- Division of Pulmonary and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States
| | - Aparna Balasubramanian
- Division of Pulmonary and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States
| |
Collapse
|
20
|
Zhou P, Yang L, Li H, Zeng L, Zhang Y, Zhong Z, Li R, Yin Y, Tao K, Zhang P. IRG1/Itaconate inhibits hepatic stellate cells ferroptosis and attenuates TAA-induced liver fibrosis by regulating SLC39A14 expression. Int Immunopharmacol 2025; 146:113945. [PMID: 39724735 DOI: 10.1016/j.intimp.2024.113945] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2024] [Revised: 12/13/2024] [Accepted: 12/22/2024] [Indexed: 12/28/2024]
Abstract
This study aimed to elucidate the protective roles of Immune Response Gene-1 (IRG1) and exogenous itaconate in murine models of hepatic fibrosis and to delineate the underlying mechanistic pathways using both wild-type and IRG1-deficient (IRG1-/-) mice. Primary murine stellate cells (mHSC) and bone marrow-derived macrophages (BMDM) were isolated and cocultured. Hepatocellular fibrosis was induced in vitro using Transforming Growth Factor-beta (TGF-β) to evaluate the protective efficacy of IRG1/itaconate. Histopathological damage in the hepatic tissues was assessed using Hematoxylin and Eosin (H&E), Masson's trichrome, and Sirius red staining, followed by hepatic fibrosis scoring. The levels of released inflammatory cytokines were quantified using enzyme-linked immunosorbent assay (ELISA) kits. Immunohistochemistry was used to detect 4-Hydroxynonenal (4-HNE) levels and Perls staining was used to assess ferroptosis. RNA sequencing and gene enrichment analyses were performed to identify implicated molecular entities and signaling pathways. IRG1 and SLC39A14 knockdown and overexpression cell lines were generated. Quantitative real-time PCR (qRT-PCR) and western blotting (WB) were used to measure the mRNA and protein expression levels in hepatic tissues and cells. Kits were used to assess reactive oxygen species (ROS) and malondialdehyde (MDA) levels, and the concentrations of liver enzymes, iron, GSH, and GSSG within hepatic tissues and cells.4-octyl itaconate (4-OI) significantly attenuated the histopathological damage in hepatic tissues, preserved the normal hepatic function, effectively reduced the release of inflammatory cytokines, and mitigated oxidative stress markers such as ROS and MDA in Thioacetamide (TAA)-induced fibrotic mice. Notably, this study is the first to reveal the pivotal role of SLC39A14 in the pathogenesis of hepatic fibrosis in murine models and elucidate how IRG1/itaconate mediates downstream ferroptosis-related signaling pathways by targeting SLC39A14, thereby inhibiting ferroptosis-induced hepatic fibrosis. IRG1/itaconate can alleviate the TAA-induced hepatic fibrosis in mice by regulating the expression of SLC39A14, consequently suppressing hepatic stellate cell ferroptosis.
Collapse
Affiliation(s)
- Pei Zhou
- Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Lei Yang
- Department of General Surgery, The Second Hospital of Shanxi Medical University, Taiyuan 030001, China
| | - Hang Li
- Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Liwu Zeng
- Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Yizhuo Zhang
- Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Ziyou Zhong
- Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Ruidong Li
- Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Yuping Yin
- Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Kaixiong Tao
- Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China.
| | - Peng Zhang
- Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China.
| |
Collapse
|
21
|
Costantini C, Brancorsini S, Grignani F, Romani L, Bellet MM. Circadian metabolic adaptations to infections. Philos Trans R Soc Lond B Biol Sci 2025; 380:20230473. [PMID: 39842481 PMCID: PMC11753887 DOI: 10.1098/rstb.2023.0473] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2024] [Revised: 04/22/2024] [Accepted: 05/16/2024] [Indexed: 01/24/2025] Open
Abstract
Circadian clocks are biological oscillators that evolved to coordinate rhythms in behaviour and physiology around the 24-hour day. In mammalian tissues, circadian rhythms and metabolism are highly intertwined. The clock machinery controls rhythmic levels of circulating hormones and metabolites, as well as rate-limiting enzymes catalysing biosynthesis or degradation of macromolecules in metabolic tissues, such control being exerted both at the transcriptional and post-transcriptional level. During infections, major metabolic adaptation occurs in mammalian hosts, at the level of both the single immune cell and the whole organism. Under these circumstances, the rhythmic metabolic needs of the host intersect with those of two other players: the pathogen and the microbiota. These three components cooperate or compete to meet their own metabolic demands across the 24 hours. Here, we review findings describing the circadian regulation of the host response to infection, the circadian metabolic adaptations occurring during host-microbiota-pathogen interactions and how such regulation can influence the immune response of the host and, ultimately, its own survival.This article is part of the Theo Murphy meeting issue 'Circadian rhythms in infection and immunity'.
Collapse
Affiliation(s)
- Claudio Costantini
- Department of Medicine and Surgery, University of Perugia, P.le L. Severi 1, Perugia06132, Italy
| | - Stefano Brancorsini
- Department of Medicine and Surgery, University of Perugia, P.le L. Severi 1, Perugia06132, Italy
| | - Francesco Grignani
- Department of Medicine and Surgery, University of Perugia, P.le L. Severi 1, Perugia06132, Italy
| | - Luigina Romani
- Department of Medicine and Surgery, University of Perugia, P.le L. Severi 1, Perugia06132, Italy
| | - Marina Maria Bellet
- Department of Medicine and Surgery, University of Perugia, P.le L. Severi 1, Perugia06132, Italy
| |
Collapse
|
22
|
de Man AM, Stoppe C, Koekkoek KW, Briassoulis G, Subasinghe LS, Cobilinschi C, Deane AM, Manzanares W, Grințescu I, Mirea L, Roshdy A, Cotoia A, Bear DE, Boraso S, Fraipont V, Christopher KB, Casaer MP, Gunst J, Pantet O, Elhadi M, Bolondi G, Forceville X, Angstwurm MW, Gurjar M, Biondi R, van Zanten AR, Berger MM, ESICM/FREM MN group. What do we know about micronutrients in critically ill patients? A narrative review. JPEN J Parenter Enteral Nutr 2025; 49:33-58. [PMID: 39555865 PMCID: PMC11717498 DOI: 10.1002/jpen.2700] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Revised: 09/03/2024] [Accepted: 10/07/2024] [Indexed: 11/19/2024]
Abstract
Micronutrient (MN) status alterations (both depletion and deficiency) are associated with several complications and worse outcomes in critically ill patients. On the other side of the spectrum, improving MN status has been shown to be a potential co-adjuvant therapy. This review aims to collect existing data to better guide research in the critical care setting. This narrative review was conducted by the European Society of Intensive Care Medicine Feeding, Rehabilitation, Endocrinology, and Metabolism MN group. The primary objective was to identify studies focusing on individual MNs in critically ill patients, selecting the MNs that appear to be most relevant and most frequently investigated in the last decade: A, B1, B2, B3, B6, folate, C, D, E, copper, iron, selenium, zinc, and carnitine. Given the limited number of interventional studies for most MNs, observational studies were included. For each selected MN, the review summarizes the main form and functions, special needs and risk factors, optimal treatment strategies, pharmacological dosing, and clinical implications all specific to critically ill patients. A rigorous rebalancing of research strategies and priorities is needed to improve clinical practice. An important finding is that high-dose monotherapy of MNs is not recommended. Basal daily needs must be provided, with higher doses in diseases with known higher needs, and identified deficiencies treated. Finally, the review provides a list of ongoing trials on MNs in critically ill patients and identifies a priority list of future research topics.
Collapse
Affiliation(s)
- Angelique M.E. de Man
- Department of Intensive Care; Amsterdam Cardiovascular Sciences, Amsterdam UMClocation Vrije UniversiteitAmsterdamthe Netherlands
| | - Christian Stoppe
- University Hospital Wuerzburg, Department of Anaesthesiology, Intensive Care, Emergency, and Pain MedicineWuerzburgGermany
| | | | - George Briassoulis
- Postgraduate Program, Emergency and Intensive Care in Children Adolescents and Young Adults, School of MedicineUniversity of CreteHeraklionGreece
| | - Lilanthi S.D.P. Subasinghe
- Head of the Department ‐ Division of Intensive Care, University HospitalGeneral Sir John Kotelawala Defence UniversityColomboSri Lanka
| | - Cristian Cobilinschi
- Department of Anesthesiology and Intensive Care II“Carol Davila” University of Medicine and PharmacyBucharestRomania
- Department of Anesthesiology and Intensive Care I, Clinical Emergency Hospital of BucharestBucharestRomania
| | - Adam M. Deane
- Department of Critical Care, Melbourne Medical SchoolUniversity of MelbourneParkvilleVicAustralia
| | - William Manzanares
- Department of Critical Care, Hospital de Clínicas (University Hospital)Faculty of MedicineUdelaRMontevideoUruguay
| | - Ioana Grințescu
- Department of Anesthesiology and Intensive Care II“Carol Davila” University of Medicine and PharmacyBucharestRomania
- Department of Anesthesiology and Intensive Care I, Clinical Emergency Hospital of BucharestBucharestRomania
| | - Liliana Mirea
- Department of Anesthesiology and Intensive Care II“Carol Davila” University of Medicine and PharmacyBucharestRomania
- Department of Anesthesiology and Intensive Care I, Clinical Emergency Hospital of BucharestBucharestRomania
| | - Ashraf Roshdy
- Critical Care Medicine Department, Faculty of MedicineAlexandria UniversityAlexandriaEgypt
| | - Antonella Cotoia
- Department of Critical CareUniversity Hospital of FoggiaFoggiaItaly
| | - Danielle E. Bear
- Department of Nutritional Sciences, School of Life Course and Population SciencesKing's College LondonLondonUK
- Department of Nutrition and Dietetics and Department of Critical CareGuy's and St Thomas’ NHS Foundation TrustLondonUK
| | - Sabrina Boraso
- General and Neurosurgical Intensive Care Unit, Ospedale dell'AngeloMestre‐VeneziaItaly
| | | | - Kenneth B. Christopher
- Channing Division of Network Medicine, Brigham and Women's HospitalBostonUSA
- Division of Renal Medicine, Brigham and Women's HospitalBostonUSA
| | - Michael P. Casaer
- Department of Cellular and Molecular Medicine, Laboratory of Intensive Care MedicineKU LeuvenLeuvenBelgium
- Intensive Care MedicineUZ LeuvenBelgium
| | - Jan Gunst
- Department of Cellular and Molecular Medicine, Laboratory of Intensive Care MedicineKU LeuvenLeuvenBelgium
- Intensive Care MedicineUZ LeuvenBelgium
| | - Olivier Pantet
- Department of Intensive Care MedicineUniversity Hospital of LausanneLausanneSwitzerland
| | | | - Giuliano Bolondi
- Anesthesia and Intensive Care Unit, Ospedale BufaliniCesena (FC)Italy
| | - Xavier Forceville
- Inserm, CIC 1414 (Centre d′ Investigation Clinique de Rennes)Univ Rennes, CHU RennesRennesF‐35000France
| | | | - Mohan Gurjar
- Department of Critical Care MedicineSanjay Gandhi Post Graduate Institute of Medical SciencesIndia
| | | | - Arthur R.H. van Zanten
- Department of Intensive Care Medicine, Gelderse Vallei Hospital, Ede, the Netherlands; Wageningen University & Research, Division of Human Nutrition and HealthWageningenthe Netherlands
| | - Mette M. Berger
- Faculty of Biology and MedicineLausanne UniversityLausanneSwitzerland
| | | |
Collapse
|
23
|
Polani R, De Francesco A, Tomolillo D, Artuso I, Equestre M, Trirocco R, Arcari G, Antonelli G, Villa L, Prosseda G, Visca P, Carattoli A. Cefiderocol Resistance Conferred by Plasmid-Located Ferric Citrate Transport System in KPC-Producing Klebsiella pneumoniae. Emerg Infect Dis 2025; 31:123-124. [PMID: 39714320 PMCID: PMC11682805 DOI: 10.3201/eid3101.241426] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2024] Open
Abstract
Cefiderocol (FDC), a siderophore-cephalosporin conjugate, is the newest option for treating infection with carbapenem-resistant gram-negative bacteria. We identified a novel mechanism contributing to decreased FDC susceptibility in Klebsiella pneumoniae clinical isolates. The mechanism involves 2 coresident plasmids: pKpQIL, carrying variants of blaKPC carbapenemase gene, and pKPN, carrying the ferric citrate transport (FEC) system. We observed increasing FDC MICs in an Escherichia coli model system carrying different natural pKpQIL plasmids, encoding different K. pneumoniae carbapenemase (KPC) variants, in combination with a conjugative low copy number vector carrying the fec gene cluster from pKPN. We observed transcriptional repression of fiu, cirA, fepA, and fhuA siderophore receptor genes in blaKPC-fec-E. coli cells treated with ferric citrate. Screening of 27,793 K. pneumoniae whole-genome sequences revealed that the fec cluster occurs frequently in some globally distributed different KPC-producing K. pneumoniae clones (sequence types 258, 14, 45, and 512), contributing to reduced FDC susceptibility.
Collapse
|
24
|
Wang B, Xi F, Jin C, Zhu HL, Tu M, Li Z. A ferrous fluorescence lifetime response probe for monitoring changes in lipid droplets during ferroptosis and imaging in liver disease model. Biosens Bioelectron 2025; 267:116742. [PMID: 39243450 DOI: 10.1016/j.bios.2024.116742] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Revised: 09/03/2024] [Accepted: 09/03/2024] [Indexed: 09/09/2024]
Abstract
Ferrous ions (Fe2⁺) accumulation and abnormal alterations in lipid droplets (LDs) are closely associated with ferroptosis. In the liver, excessive iron accumulation promotes oxidative stress and exacerbates lipid droplet accumulation, while the disruption of iron homeostasis may also affect the formation and size of lipid droplets, their increased number and size can exacerbate the severity of disease under fatty liver conditions. The leads to hepatocyte damage, further triggering liver inflammation, fibrosis, and ultimately resulting in cirrhosis and hepatocellular carcinoma. Therefore, real-time monitoring of iron ion and lipid droplet changes is crucial for assessing the severity of liver disease, disease progression, and understanding the mechanisms of ferroptosis. We have developed a fluorescent probe, NRFep, for real-time monitoring of iron ion fluctuations and visualization of lipid droplet changes in ferroptosis and liver disease models. NRFep is specific and sensitive to iron ions and exhibits excellent stability in both cells and animal models. In addition, NRFep can be used to monitor changes in iron ions and lipid droplets in mouse liver injury and fatty liver models. Through fluorescence lifetime imaging technology, NRFep can also study the dynamic changes of intracellular iron ion content. NRFep provides a powerful tool for studying ferroptosis and related diseases, and its unique dual-monitoring function opens up new possibilities for developing new diagnostic and therapeutic strategies.
Collapse
Affiliation(s)
- Bin Wang
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, No.163 Xianlin Road, Nanjing, 210023, China
| | - Fangmin Xi
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, No.163 Xianlin Road, Nanjing, 210023, China
| | - Chen Jin
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, No.163 Xianlin Road, Nanjing, 210023, China
| | - Hai-Liang Zhu
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, No.163 Xianlin Road, Nanjing, 210023, China
| | - Min Tu
- Pancreas Center, The First Affiliated Hospital of Nanjing Medical University, Jiangsu Province Hospital, Nanjing, 210029, China
| | - Zhen Li
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, No.163 Xianlin Road, Nanjing, 210023, China.
| |
Collapse
|
25
|
Sato R, Koziolek MJ, von Haehling S. Translating evidence into practice: Managing electrolyte imbalances and iron deficiency in heart failure. Eur J Intern Med 2025; 131:15-26. [PMID: 39521682 DOI: 10.1016/j.ejim.2024.10.024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/21/2024] [Revised: 10/23/2024] [Accepted: 10/27/2024] [Indexed: 11/16/2024]
Abstract
Mineral abnormalities are a common complication of heart failure (HF). In particular, dyskalaemia, hyponatraemia, and hypomagnesaemia are prevalent, with hypo- and hyperkalaemia observed in over 40 % of HF patients, hyponatraemia in 18-27 %, hypomagnesaemia in 7-52 %, and phosphate imbalance in 13 %. These abnormalities serve as indicators of the severity of HF and are strongly associated with an increased risk of morbidity and mortality. The neurohumoral activation, including the renin-angiotensin-aldosterone system (RAAS), the sympathetic nervous system, and vasopressin, HF medications such as diuretics and RAAS inhibitors, amd concomitant diseases such as chronic kidney disease, can disrupt mineral homeostasis. Iron deficiency (ID) is another of the most common mineral abnormalities, affecting up to 60 % of HF patients. ID is significantly associated with adverse clinical outcomes such as reduced quality of life and exercise capacity, HF re-hospitalization, and all-cause mortality. Various pathways contribute to the development of ID in HF, including reduced iron intake due to anorexia, increased hepcidin levels associated with chronic inflammation and hepatic congestion, and occult gastrointestinal bleeding due to the concomitant use of antithrombotic agents. The efficacy of iron replacement therapy has been demonstrated in clinical trials, particularly in heart failure with reduced ejection fraction (HFrEF), whilst more recently, it has also been shown to improve exercise capacity in patients with heart failure with preserved ejection fraction (HFpEF). This review focuses on potassium and phosphate abnormalities, hyponatraemia, hypomagnesaemia, and ID in HF, providing a comprehensive overview of the mechanisms, clinical significance, and intervention strategies with the latest findings.
Collapse
Affiliation(s)
- Ryosuke Sato
- Department of Cardiology and Pneumology, University Medical Center Göttingen, Göttingen, Germany; DZHK (German Center for Cardiovascular Research), Partner Site Lower Saxony, Germany
| | - Michael J Koziolek
- DZHK (German Center for Cardiovascular Research), Partner Site Lower Saxony, Germany; Department of Nephrology and Rheumatology, University Medical Centre, Göttingen, Germany
| | - Stephan von Haehling
- Department of Cardiology and Pneumology, University Medical Center Göttingen, Göttingen, Germany; DZHK (German Center for Cardiovascular Research), Partner Site Lower Saxony, Germany.
| |
Collapse
|
26
|
Ou H, Lin J, Ji L, Ye L, Ling M, Liao X, Lin F, Wang Y, Luo B, Hu Z, Pan L. Ferritinophagy mediated by the AMPK/ULK1 pathway is involved in ferroptosis subsequent to ventilator-induced lung injury. Respir Res 2024; 25:440. [PMID: 39719634 DOI: 10.1186/s12931-024-03076-7] [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: 10/04/2024] [Accepted: 12/18/2024] [Indexed: 12/26/2024] Open
Abstract
Mechanical ventilation (MV) remains a cornerstone of critical care; however, its prolonged application can exacerbate lung injury, leading to ventilator-induced lung injury (VILI). Although previous studies have implicated ferroptosis in the pathogenesis of VILI, the underlying mechanisms remain unclear. This study investigated the roles of ferritinophagy in ferroptosis subsequent to VILI. Using C57BL/6J mice and MLE-12 cells, we established both in vivo and in vitro models of VILI and cyclic stretching (CS)-induced cellular injury. We assessed lung injury and the biomarkers of ferroptosis and ferritinophagy, after appropriate pretreatments. This study demonstrated that high tidal volumes (HTV) for 4 h enhanced the sensitivity to ferroptosis in both models, evidenced by increased intracellular iron levels, lipid peroxidation and cell death, which can be mitigated by ferrostatin-1 treatment. Notably, nuclear receptor coactivator 4 (NCOA4)-mediated ferritinophagy contributed to ferroptosis in VILI. Inhibition of autophagy with 3-methyladenine or NCOA4 knockdown decreased intracellular Fe2+ levels and inhibited lipid peroxidation, thereby attenuating CS-induced lung injury. Furthermore, it has also been observed that the AMPK/ULK1 axis can trigger ferritinophagy in VILI. Collectively, our study indicated that MV can induce ferroptosis by promoting NCOA4-dependent ferritinophagy, which could be a novel therapeutic target for the prevention and treatment of VILI.
Collapse
Affiliation(s)
- Huajin Ou
- Department of Anesthesiology, Guangxi Medical University Cancer Hospital, He Di Rd No.71, Nanning, 530021, P. R. China
- Guangxi Engineering Research Center for Tissue & Organ Injury and Repair Medicine, Nanning, China
- Guangxi Health Commission Key Laboratory of Basic Science and Prevention of Perioperative Organ Disfunction, Nanning, China
- Guangxi Clinical Research Center for Anesthesiology, Nanning, China
| | - Jinyuan Lin
- Department of Anesthesiology, Guangxi Medical University Cancer Hospital, He Di Rd No.71, Nanning, 530021, P. R. China
- Guangxi Engineering Research Center for Tissue & Organ Injury and Repair Medicine, Nanning, China
- Guangxi Health Commission Key Laboratory of Basic Science and Prevention of Perioperative Organ Disfunction, Nanning, China
- Guangxi Clinical Research Center for Anesthesiology, Nanning, China
| | - Liu Ji
- Department of Anesthesiology, Guangxi Medical University Cancer Hospital, He Di Rd No.71, Nanning, 530021, P. R. China
- Guangxi Engineering Research Center for Tissue & Organ Injury and Repair Medicine, Nanning, China
- Guangxi Health Commission Key Laboratory of Basic Science and Prevention of Perioperative Organ Disfunction, Nanning, China
- Guangxi Clinical Research Center for Anesthesiology, Nanning, China
| | - Liu Ye
- Department of Anesthesiology, Guangxi Medical University Cancer Hospital, He Di Rd No.71, Nanning, 530021, P. R. China
- Guangxi Engineering Research Center for Tissue & Organ Injury and Repair Medicine, Nanning, China
- Guangxi Health Commission Key Laboratory of Basic Science and Prevention of Perioperative Organ Disfunction, Nanning, China
- Guangxi Clinical Research Center for Anesthesiology, Nanning, China
| | - Maoyao Ling
- Department of Anesthesiology, Guangxi Medical University Cancer Hospital, He Di Rd No.71, Nanning, 530021, P. R. China
- Guangxi Engineering Research Center for Tissue & Organ Injury and Repair Medicine, Nanning, China
- Guangxi Health Commission Key Laboratory of Basic Science and Prevention of Perioperative Organ Disfunction, Nanning, China
- Guangxi Clinical Research Center for Anesthesiology, Nanning, China
| | - Xiaoting Liao
- Department of Anesthesiology, Guangxi Medical University Cancer Hospital, He Di Rd No.71, Nanning, 530021, P. R. China
- Guangxi Engineering Research Center for Tissue & Organ Injury and Repair Medicine, Nanning, China
- Guangxi Health Commission Key Laboratory of Basic Science and Prevention of Perioperative Organ Disfunction, Nanning, China
- Guangxi Clinical Research Center for Anesthesiology, Nanning, China
| | - Fei Lin
- Department of Anesthesiology, Guangxi Medical University Cancer Hospital, He Di Rd No.71, Nanning, 530021, P. R. China
- Guangxi Engineering Research Center for Tissue & Organ Injury and Repair Medicine, Nanning, China
- Guangxi Health Commission Key Laboratory of Basic Science and Prevention of Perioperative Organ Disfunction, Nanning, China
- Guangxi Clinical Research Center for Anesthesiology, Nanning, China
| | - Yuqing Wang
- Department of Anesthesiology, Guangxi Medical University Cancer Hospital, He Di Rd No.71, Nanning, 530021, P. R. China
- Guangxi Engineering Research Center for Tissue & Organ Injury and Repair Medicine, Nanning, China
- Guangxi Health Commission Key Laboratory of Basic Science and Prevention of Perioperative Organ Disfunction, Nanning, China
- Guangxi Clinical Research Center for Anesthesiology, Nanning, China
| | - Bijun Luo
- Guangxi Engineering Research Center for Tissue & Organ Injury and Repair Medicine, Nanning, China
- Guangxi Health Commission Key Laboratory of Basic Science and Prevention of Perioperative Organ Disfunction, Nanning, China
- Guangxi Clinical Research Center for Anesthesiology, Nanning, China
- Department of Anesthesiology, The Maternal and Child Health Care Hospital of Guangxi Zhuang Autonomous Region, Nanning, Guangxi, China
| | - Zhaokun Hu
- Department of Anesthesiology, Guangxi Medical University Cancer Hospital, He Di Rd No.71, Nanning, 530021, P. R. China
- Guangxi Engineering Research Center for Tissue & Organ Injury and Repair Medicine, Nanning, China
- Guangxi Health Commission Key Laboratory of Basic Science and Prevention of Perioperative Organ Disfunction, Nanning, China
- Guangxi Clinical Research Center for Anesthesiology, Nanning, China
| | - Linghui Pan
- Department of Anesthesiology, Guangxi Medical University Cancer Hospital, He Di Rd No.71, Nanning, 530021, P. R. China.
- Guangxi Engineering Research Center for Tissue & Organ Injury and Repair Medicine, Nanning, China.
- Guangxi Health Commission Key Laboratory of Basic Science and Prevention of Perioperative Organ Disfunction, Nanning, China.
- Guangxi Clinical Research Center for Anesthesiology, Nanning, China.
| |
Collapse
|
27
|
Wander K, Ogunleye OO, Nwagu EN, Unigwe US, Odo AN, Chukwubike CM, Omilabu SA, Salu OB, Owolabi BS, Osikomaiya BI, Ebede SO, Bowale A, Olaitan AO, Chukwu CU, Ndiokwelu CO, Edu-Alamba C, Azubuike C, Odubiyi OA, Hassan YA, Oloniniyi N, Muyiwa Kelvin A, Rashidat Abiola R, Saliu A, Fadipe OO, Anyanwu RA, Orenolu MR, Abdullah MA, Ishaya OD, Agulefo CJ, Akase IE, Gauck ME, Huang Z, Chen MH, Okoror TA, Fujita M. Iron nutrition and COVID-19 among Nigerian healthcare workers. Evol Med Public Health 2024; 12:287-297. [PMID: 39759428 PMCID: PMC11697216 DOI: 10.1093/emph/eoae034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Revised: 10/07/2024] [Indexed: 01/07/2025] Open
Abstract
Background and objectives The optimal iron hypothesis (OIH) posits that risk for infection is lowest at a mild level of iron deficiency. The extent to which this protection results from arms race dynamics in the evolution of iron acquisition and sequestration mechanisms is unclear. We evaluated the OIH with regard to severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), an emerging infectious agent. Methodology We tested 304 healthcare workers at baseline for iron deficiency (zinc protoporphyrin:heme), anemia (hemoglobin), and SARS-CoV-2 (salivary PCR), and followed them for ~3 months with biweekly SARS-CoV-2 tests. We fit logistic regression models based on Akaike Information Criterion. Results Adequate data were available for 199 participants. Iron replete (OR: 2.87, 95% CI: 0.85, 9.75) and anemia (OR: 2.48; 95% CI: 0.82, 7.85) were associated with higher risk for SARS-CoV-2 infection after control for covariates. Logistic regression and Cox proportional hazards models of the SARS-CoV-2 outcome were similar. Anemia (OR: 1.81; 95% CI: 0.88, 3.71) was associated with respiratory symptoms regardless of SARS-CoV-2 infection. Conclusions and implications These findings provide partial support for the OIH: SARS-CoV-2 infection risk was elevated at the high end of the range of iron availability; however, the elevated risk among those with anemia was not, as expected, specific to severe iron deficiency. Narrowly, for COVID-19 epidemiology, these findings accord with evidence that SARS-CoV-2's ability to establish infection is enhanced by access to iron. More broadly, these findings suggest that the OIH does not hinge on a long history of evolutionary arms race dynamics in access to host iron.
Collapse
Affiliation(s)
- Katherine Wander
- Department of Anthropology, Binghamton University, Binghamton, NY, USA
| | - Olayinka O Ogunleye
- Department of Medicine, Lagos State University Teaching Hospital, Ikeja, Lagos, Nigeria
- Department of Pharmacology, Therapeutics and Toxicology, Lagos State University College of Medicine, Ikeja, Lagos, Nigeria
| | - Evelyn N Nwagu
- Department of Human Kinetics and Health Education, University of Nigeria, Nsukka, Enugu, Nigeria
| | - Uche S Unigwe
- Department of Medicine, University of Nigeria Teaching Hospital, Ituku-Ozalla, Enugu, Nigeria
| | - Amelia N Odo
- Department of Human Kinetics and Health Education, University of Nigeria, Nsukka, Enugu, Nigeria
| | - Chinedu M Chukwubike
- Molecular Virology Unit, Department of Microbiology University of Nigeria Teaching Hospital, Ituku-Ozalla, Enugu, Nigeria
| | - Sunday A Omilabu
- Centre for Human and Zoonotic Virology, College of Medicine, University of Lagos, Idi-Araba, Lagos, Nigeria
- Department of Medical Microbiology and Parasitology, College of Medicine, University of Lagos, Idi-Araba, Lagos, Nigeria
| | - Olumuyiwa B Salu
- Centre for Human and Zoonotic Virology, College of Medicine, University of Lagos, Idi-Araba, Lagos, Nigeria
- Department of Medical Microbiology and Parasitology, College of Medicine, University of Lagos, Idi-Araba, Lagos, Nigeria
| | - Bukola S Owolabi
- Department of Surgery, Lagos University Teaching Hospital, Idi-Araba, Mushin, Lagos, Nigeria
| | - Bodunrin I Osikomaiya
- Lagos State Blood Transfusion Service, Lagos, Nigeria
- Department of Haematology, Lagos State University Teaching Hospital, Ikeja, Lagos, Nigeria
| | - Samuel O Ebede
- Department of Medical Microbiology, University of Nigeria Teaching Hospital, Ituku-Ozalla, Enugu, Nigeria
| | | | - Abimbola O Olaitan
- Department of Medicine, University of Nigeria Teaching Hospital, Ituku-Ozalla, Enugu, Nigeria
- Olabisi Onabanjo University Teaching Hospital, Sagamu, Ogun, Nigeria
| | - Christopher U Chukwu
- Family Medicine Department, University of Nigeria Teaching Hospital, Ituku-Ozalla, Enugu, Nigeria
| | - Chibuzo O Ndiokwelu
- Department of Paediatrics, University of Nigeria Teaching Hospital, Ituku-Ozalla, Enugu, Nigeria
| | - Chioma Edu-Alamba
- Molecular Virology Unit, Department of Microbiology University of Nigeria Teaching Hospital, Ituku-Ozalla, Enugu, Nigeria
| | - Constance Azubuike
- Molecular Virology Unit, Department of Microbiology University of Nigeria Teaching Hospital, Ituku-Ozalla, Enugu, Nigeria
| | | | - Yusuf A Hassan
- Medical Emergency Unit, Lagos State University Teaching Hospital, Ikeja, Lagos, Nigeria
| | | | | | | | - Amina Saliu
- Lagos State Health Service Commission, Lagos, Nigeria
| | - Ololade O Fadipe
- Medical Emergency Unit, Lagos State University Teaching Hospital, Ikeja, Lagos, Nigeria
| | - Roosevelt A Anyanwu
- Centre for Human and Zoonotic Virology, College of Medicine, University of Lagos, Idi-Araba, Lagos, Nigeria
| | - Mercy R Orenolu
- Centre for Human and Zoonotic Virology, College of Medicine, University of Lagos, Idi-Araba, Lagos, Nigeria
| | - Maryam A Abdullah
- Centre for Human and Zoonotic Virology, College of Medicine, University of Lagos, Idi-Araba, Lagos, Nigeria
| | - Onyinye D Ishaya
- Usmanu Danfodiyo University Teaching Hospital, Garba Nadama, Sokoto, Nigeria
| | - Chinenye J Agulefo
- Centre for Human and Zoonotic Virology, College of Medicine, University of Lagos, Idi-Araba, Lagos, Nigeria
| | - Iorhen E Akase
- Lagos University Teaching Hospital, Idi-Araba, Mushin, Lagos, Nigeria
- College of Medicine, University of Lagos (CMUL), Idi-Araba, Lagos, Nigeria
| | - Megan E Gauck
- Department of Anthropology, Binghamton University, Binghamton, NY, USA
| | - Zifan Huang
- Department of Mathematics and Statistics, Binghamton University, Binghamton, NY, USA
| | - Mei-Hsiu Chen
- Department of Mathematics and Statistics, Binghamton University, Binghamton, NY, USA
| | - Titilayo A Okoror
- Department of Africana Studies, Binghamton University, Binghamton, NY, USA
| | - Masako Fujita
- Department of Anthropology, Michigan State University, East Lansing, MI, USA
| |
Collapse
|
28
|
Park MY, Agoro R, Jankauskas SS, Le Henaff C, Sitara D. Phosphorus-independent role of FGF23 in erythropoiesis and iron homeostasis. PLoS One 2024; 19:e0315228. [PMID: 39666728 PMCID: PMC11637385 DOI: 10.1371/journal.pone.0315228] [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: 07/23/2024] [Accepted: 11/21/2024] [Indexed: 12/14/2024] Open
Abstract
A number of studies have reported an association between phosphorus, red blood cell (RBC) production, and iron metabolism. However, it is difficult to distinguish whether the effect of phosphorus is direct or through the actions of FGF23, and it is not clear whether phosphorus is positively or negatively associated with RBC production. In the present study, we investigated the effects of a) increased phosphorus load and b) phosphorus deficiency on erythropoiesis and iron metabolism in association with FGF23. Mice were fed either a 1.2% or 1.65% phosphorus diet and compared to mice fed a control diet containing 0.6% of phosphorus. Moreover, we used two mouse models of hypophosphatemia-induced either by dietary intervention in the form of a low phosphorus (LP) diet (0.02% of Pi) or genetically in a mouse model of X-linked hypophosphatemia (XLH)-that had opposite FGF23 levels. Phosphorus supplementation appropriately increased FGF23 levels leading to excretion of excess phosphorus and normalization of serum phosphorus levels. We also found that a phosphorus-rich diet results in inflammation-induced hypoferremia associated with reduced iron export leading to tissue iron overload. Moreover, high phosphorus intake results in ineffective erythropoiesis caused by decreased production (decreased RBCs, hemoglobin, hematocrit, and erythroid progenitors in the bone marrow) and increased destruction of RBCs, leading to anemia despite increased EPO secretion. These complications occur through the actions of elevated FGF23 in the presence of normophosphatemia. Our data also show that LP diet induces a decrease in the serum concentrations of phosphorus and FGF23, resulting in increased RBC counts, hemoglobin concentration, and hematocrit compared to mice fed normal diet. Moreover, serum iron and transferrin saturation were increased and positively correlated with serum ferritin, liver ferritin protein and mRNA expression in mice fed LP diet. However, hyp mice, the murine model of XLH, exhibit hypophosphatemia and high serum FGF23 levels, along with low number of circulating RBCs, hemoglobin, and hematocrit compared to wild-type mice. In the bone marrow, hyp mice showed reduced number of erythroid progenitors and formed significantly less BFU-E colonies compared to control mice. Serum iron levels and transferrin saturation were also decreased in hyp mice in comparison to control mice. Taken together, our data show that FGF23 acts independent of phosphorus levels to regulate erythropoiesis and iron homeostasis.
Collapse
Affiliation(s)
- Min Young Park
- Department of Molecular Pathobiology, New York University College of Dentistry, New York, NY, United States of America
| | - Rafiou Agoro
- Department of Mammalian Genetics, The Jackson Laboratory, Bar Harbor, ME, United States of America
| | | | - Carole Le Henaff
- Department of Molecular Pathobiology, New York University College of Dentistry, New York, NY, United States of America
| | - Despina Sitara
- Department of Molecular Pathobiology, New York University College of Dentistry, New York, NY, United States of America
- Department of Medicine, Holman Division of Endocrinology, Diabetes and Metabolism, New York University Grossman School of Medicine, New York, NY, United States of America
| |
Collapse
|
29
|
Tembhurne S, Malagi SK, Abraham DV, Pandaw KS, Mahato A, Agrawal P. Comparative Evaluation of Salivary Ferritin Levels as a Predictor Inflammatory Biomarker Before and After Non Surgical Periodontal Therapy- An Interventional Study. JOURNAL OF PHARMACY AND BIOALLIED SCIENCES 2024; 16:S3266-S3268. [PMID: 39926847 PMCID: PMC11805173 DOI: 10.4103/jpbs.jpbs_759_24] [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: 07/01/2024] [Revised: 07/06/2024] [Accepted: 07/15/2024] [Indexed: 02/11/2025] Open
Abstract
Background Ferritin plays a crucial role in the host immune response, with elevated serum ferritin levels established in various chronic inflammation-related diseases, including periodontitis. This study aimed to evaluate the efficacy of nonsurgical periodontal treatment on salivary ferritin levels in periodontally healthy and chronic periodontitis patients. Materials and Methods Fifty patients were selected and divided into two groups: a control group of healthy individuals and an experimental group of chronic periodontitis patients. Salivary samples were collected using the drooling method to assess ferritin levels. The experimental group underwent nonsurgical periodontal therapy, with clinical parameters [plaque index (PI), gingival index (GI), oral hygiene index-simplified (OHIS), bleeding point index (BPI), probing pocket depth (PPD), and clinical attachment level (CAL)] recorded before and one month after therapy. Salivary samples were analyzed using CLIA, and statistical analysis was performed. Results The control group showed significantly lower ferritin levels compared to the periodontitis group. Post-therapy, the periodontitis group exhibited significant improvements in both clinical parameters and ferritin levels. Conclusion Salivary ferritin levels effectively predict periodontal disease and the impact of periodontal therapy, demonstrating its potential as a valuable biomarker for diagnosing and monitoring periodontal health.
Collapse
Affiliation(s)
- Saurabh Tembhurne
- Department of Periodontology, Maitri College of Dentistry and Research Centre, Anjora, Chhattisgarh, India
| | - Sachin K. Malagi
- Department of Periodontology, Maitri College of Dentistry and Research Centre, Anjora, Chhattisgarh, India
| | - Dennis V. Abraham
- Department of Periodontology, Maitri College of Dentistry and Research Centre, Anjora, Chhattisgarh, India
| | - Kshitibhushan S. Pandaw
- Department of Periodontology, Maitri College of Dentistry and Research Centre, Anjora, Chhattisgarh, India
| | - Arundhuti Mahato
- Department of Periodontology, Maitri College of Dentistry and Research Centre, Anjora, Chhattisgarh, India
| | - Priyal Agrawal
- Department of Periodontology, Maitri College of Dentistry and Research Centre, Anjora, Chhattisgarh, India
| |
Collapse
|
30
|
Vinhas S, de Castro B, Rangel M. Synthesis of 3-hydroxy-4-pyridinone hexadentate chelators, and biophysical evaluation of their affinity towards lipid bilayers. Bioorg Chem 2024; 153:107806. [PMID: 39255611 DOI: 10.1016/j.bioorg.2024.107806] [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: 06/27/2024] [Revised: 08/28/2024] [Accepted: 09/03/2024] [Indexed: 09/12/2024]
Abstract
Iron is an essential micronutrient for almost every living organism, namely pathogenic bacteria. In an infection scenario, host-pathogen competitive relationships for the element are present and Fe withholding is a well known response of the host. Also, bacterial resistance is a major concern that can compromise public health and the WHO underlines an urgent need to search for new pharmaceutical ingredients or strategies to fight opportunistic bacteria. Iron metabolism, and in particular, deprivation is a strategy that currently constitutes another option to fight bacterial infection. In this work we report the synthesis of a new hexadentate chelator with enhanced hydrophilicity (MRHT) and the improved synthesis of two other chelators. The affinity towards charged and non-charged phospholipid bilayers was evaluated for three hexadentate chelators: MRHT, CP256 and RH8b using NMR and EPR spectroscopies. The results revealed that these structures, bearing 3,4-HPO units have a high affinity towards the hydrophilic region of the phospholipid bilayer. From the three hexadentate chelators, MRHT stood out, especially for liposomes with a charged surface, suggesting that this molecule could more efficiently compete with natural siderophores, creating an iron gradient near bacteria organisms.
Collapse
Affiliation(s)
- Sílvia Vinhas
- REQUIMTE, LAQV, Departamento de Química e Bioquímica, Faculdade de Ciências, Universidade do Porto, 4069-007 Porto, Portugal
| | - Baltazar de Castro
- REQUIMTE, LAQV, Departamento de Química e Bioquímica, Faculdade de Ciências, Universidade do Porto, 4069-007 Porto, Portugal
| | - Maria Rangel
- REQUIMTE, LAQV, Instituto de Ciências Biomédicas de Abel Salazar, Universidade do Porto, 4050-313 Porto, Portugal.
| |
Collapse
|
31
|
Sandnes M, Reikvam H. Hepcidin as a therapeutic target in iron overload. Expert Opin Ther Targets 2024; 28:1039-1046. [PMID: 39679683 DOI: 10.1080/14728222.2024.2443081] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2024] [Revised: 11/18/2024] [Accepted: 12/12/2024] [Indexed: 12/17/2024]
Abstract
INTRODUCTION Dysregulation of the hepcidin-ferroportin axis is a hallmark in the pathogenesis of iron overload, ultimately leading to end-organ injury. Hereditary hemochromatosis and iron-loading anemias are characterized by a hepcidin deficiency, making hepcidin a novel therapeutic target for preventing and managing iron overload. AREAS COVERED Modulators of hepcidin expression and molecules mimicking hepcidin are emerging as highly promising therapeutic strategies. We present a summary of results from preclinical and clinical trials of such therapies in models of iron overload. EXPERT OPINION Current treatment alternatives in iron overload fail to address the underlying hepcidin deficiency - and may even exacerbate it. Until hepcidin-targeting therapies become available, several challenges remain, including the need to optimize dosing in order to manage the narrow treatment window and improving specificity in targeting iron metabolism pathways exclusively. Long-term studies are crucial to fully assess both the benefits and risks of these therapies and to explore their potential utility in combination with existing treatment guidelines. Furthermore, these therapies are expected to have applications, particularly in addressing other iron-maldistributed disorders, as seen in anemia of chronic disease and inflammation.
Collapse
Affiliation(s)
- Miriam Sandnes
- Department of Medicine, Haukeland University Hospital, Bergen, Norway
| | - Håkon Reikvam
- Department of Medicine, Haukeland University Hospital, Bergen, Norway
- K.G. Jebsen Center of Myeloid Malignancies, Department of Clinical Science, University of Bergen, Bergen, Norway
| |
Collapse
|
32
|
Alrouji M, Anwar S, Venkatesan K, Shahwan M, Hassan MI, Islam A, Shamsi A. Iron homeostasis and neurodegeneration in the ageing brain: Insight into ferroptosis pathways. Ageing Res Rev 2024; 102:102575. [PMID: 39515619 DOI: 10.1016/j.arr.2024.102575] [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: 06/26/2024] [Revised: 10/25/2024] [Accepted: 11/03/2024] [Indexed: 11/16/2024]
Abstract
Ageing is a major risk factor for various chronic diseases and offers a potential target for developing novel and broadly effective preventatives or therapeutics for age-related conditions, including those affecting the brain. Mechanisms contributing to ageing have been summarized as the hallmarks of ageing, with iron imbalance being one of the major factors. Ferroptosis, an iron-mediated lipid peroxidation-induced programmed cell death, has recently been implicated in neurodegenerative diseases such as Alzheimer's disease (AD), Parkinson's disease (PD) and Huntington's disease (HD). Addressing ferroptosis offers both opportunities and challenges for treating neurodegenerative diseases, though the specific mechanisms remain unclear. This research explores the key processes behind how ferroptosis contributes to brain ageing, with a focus on the complex signaling networks that are involved. The current article aims to uncover that how ferroptosis, a specific type of cell death, may drive age-related changes in the brain. Additionally, the article also unveils its role in neurodegenerative diseases, discussing how understanding these mechanisms could open up new therapeutic avenues.
Collapse
Affiliation(s)
- Mohammed Alrouji
- Department of Medical Laboratories, College of Applied Medical Sciences, Shaqra University, Shaqra 11961, Saudi Arabia.
| | - Saleha Anwar
- Center for Global Health Research, Saveetha medical college, Saveetha institute of Medical and Technical Sciences, Chennai, India.
| | - Kumar Venkatesan
- Department of Pharmaceutical Chemistry, College of Pharmacy, King Khalid University, Abha 62529, Saudi Arabia.
| | - Moyad Shahwan
- Centre of Medical and Bio-allied Health Sciences Research, Ajman University, United Arab Emirates.
| | - Md Imtaiyaz Hassan
- Center for Interdsicplinary Research in Basic Sciences, Jamia Millia Islamia, New Delhi, India.
| | - Asimul Islam
- Center for Interdsicplinary Research in Basic Sciences, Jamia Millia Islamia, New Delhi, India.
| | - Anas Shamsi
- Centre of Medical and Bio-allied Health Sciences Research, Ajman University, United Arab Emirates.
| |
Collapse
|
33
|
Yadav A, Narain U, Gupta A, Maurya S. Iron Deficiency and Renal Phosphate Handling: The Role of Maximal Tubular Reabsorption of Phosphate Normalized to Glomerular Filtration Rate (TmP/GFR) in Proximal Tubular Dysfunction. Cureus 2024; 16:e76329. [PMID: 39850154 PMCID: PMC11756918 DOI: 10.7759/cureus.76329] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/23/2024] [Indexed: 01/25/2025] Open
Abstract
BACKGROUND AND AIM Phosphate dysregulation is often associated with chronic kidney disease (CKD), and recent studies suggest that it may also be present in non-CKD patients with systemic conditions including iron deficiency anemia. This study aimed to evaluate the relationship between iron deficiency parameters (total iron-binding capacity {TIBC}, hemoglobin, and serum ferritin) and markers of proximal tubular dysfunction (the maximal tubular reabsorption of phosphate normalized to glomerular filtration rate {TmP/GFR} and tubular reabsorption of phosphate {TRP}) in non-CKD patients with iron deficiency anemia. METHODS This was a hospital-based analytical cross-sectional study conducted in the outpatient department and/or inpatient wards of the Department of Internal Medicine, Swaroop Rani Nehru (SRN) Hospital associated with Moti Lal Nehru (MLN) Medical College, Prayagraj, Uttar Pradesh, India, between July 2023 and August 2024. RESULTS This study analyzed 40 anemic patients without CKD, with a mean age of 33.9 years. Most participants (n=24, 60%) were aged 18-35 years, and the majority (n=27, 67.5%) were female. Peripheral smear analysis revealed that 72.5% (n=29) had microcytic hypochromic anemia. Hemoglobin levels averaged 7.7 g/dL, serum iron was 91.0 µg/dL, total iron-binding capacity (TIBC) was 316.3 µg/dL, and serum ferritin was 199.7 ng/mL. Phosphate handling was assessed with TmP/GFR and tubular reabsorption of phosphate (TRP) showing mean values of 4.1 mg/dL and 99.2%, respectively. This study found that TmP/GFR had a significant positive correlation with TIBC (r=0.402, p=0.010), but non-significant negative correlations with hemoglobin and serum iron. TRP was negatively correlated with hemoglobin and serum ferritin, but not significantly. Among patients with microcytic hypochromic anemia, 55.2% (n=16) had increased TmP/GFR, and 61.1% (n=20) of patients with iron deficiency anemia exhibited increased TmP/GFR. Regression analysis revealed that TIBC significantly predicted TmP/GFR (p=0.022), indicating that higher TIBC values are associated with increased TmP/GFR, suggesting a potential link between iron metabolism and renal phosphate handling. CONCLUSION Higher TIBC levels were associated with increased TmP/GFR, suggesting that iron deficiency anemia may influence proximal tubular function. The findings emphasize the importance of considering renal phosphate handling in patients with iron deficiency anemia.
Collapse
Affiliation(s)
- Aman Yadav
- General Medicine, Moti Lal Nehru Medical College, Prayagraj, IND
| | - Upma Narain
- Microbiology, Tejas Micro Diagnostic Centre, Prayagraj, IND
| | - Arvind Gupta
- Nephrology, Moti Lal Nehru Medical College, Prayagraj, IND
| | - Santosh Maurya
- Nephrology, Moti Lal Nehru Medical College, Prayagraj, IND
| |
Collapse
|
34
|
Paudel S, Severin GB, Pirani A, Snitkin ES, Mobley HLT. Multiplexed PCR to measure in situ growth rates of uropathogenic E. coli during experimental urinary tract infection. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.11.21.624689. [PMID: 39605434 PMCID: PMC11601645 DOI: 10.1101/2024.11.21.624689] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/29/2024]
Abstract
Measuring bacterial growth rates in vitro is routine, however, determining growth rates during infection in host has been more challenging. Peak-to-trough ratio (PTR) is a technique for studying microbial growth dynamics, calculated using the ratio of replication origin (ori) copies to that of the terminus (ter), as originally defined by whole genome sequencing (WGS). WGS presents significant challenges in terms of expense and data analysis complexity due to the presence of host DNA in the samples. Here, we used multiplexed PCR with fluorescent probes to estimate bacterial growth rates based on the abundance of ori- and ter-adjacent loci, without the need for WGS. We establish the utility of this approach by comparing growth rates of the uropathogenic Escherichia coli (UPEC) strain HM86 by WGS (PTR) and qPCR to measure the equivalent ori:ter (O:TPCR ). We found that PTR and O:TPCR were highly correlated and that O:TPCR reliably predicted growth rates calculated by conventional methods. O:TPCR was then used to calculate the in situ E. coli growth rates in urine, bladder, and kidneys collected over the course of a week from a murine model of urinary tract infection (UTI). These analyses revealed that growth rate of UPEC strains gradually increased during the early stages of infection (0-6h), followed by a slow decrease in growth rates during later time points (1-7 days). This rapid and convenient method provides valuable insights into bacterial growth dynamics during infection and can be applied to other bacterial species in both animal models and clinical infections.
Collapse
Affiliation(s)
- Santosh Paudel
- Department of Microbiology and Immunology, and University of Michigan Medical School, Ann Arbor, USA
| | - Geoffrey B Severin
- Department of Microbiology and Immunology, and University of Michigan Medical School, Ann Arbor, USA
| | - Ali Pirani
- Department of Microbiology and Immunology, and University of Michigan Medical School, Ann Arbor, USA
| | - Evan S Snitkin
- Department of Microbiology and Immunology, and University of Michigan Medical School, Ann Arbor, USA
- Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, USA
| | - Harry L T Mobley
- Department of Microbiology and Immunology, and University of Michigan Medical School, Ann Arbor, USA
| |
Collapse
|
35
|
Wu L, Yuan Z, Wang M, Fu X, Liu X, Wei B, Liu Y. Hepcidin Exacerbates Iron Metabolism Imbalance in Septic Mice. Infect Drug Resist 2024; 17:5027-5036. [PMID: 39554472 PMCID: PMC11566574 DOI: 10.2147/idr.s484103] [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] [Subscribe] [Scholar Register] [Received: 06/24/2024] [Accepted: 11/07/2024] [Indexed: 11/19/2024] Open
Abstract
Purpose Sepsis is a life-threatening condition associated with acute organ dysfunction. Iron is an essential trace element for multicellular organisms and almost all microorganisms, and its role in sepsis has been increasingly recognized. The aim of this study was to investigate the changes in iron metabolism in caecal ligation and puncture solution (CLP) -induced septic mice and the effects of hepcidin pretreatment on serum inflammatory marker levels and liver iron metabolism in CLP-induced septic mice. Methods C57BL/6 mice were given normal saline, CLP (peritonitis model) or 100 μg of hepcidin via intraperitoneal injection. The experimental animals were divided into 4 groups: the control group, model group (CLP), hepcidin pretreatment Groups CLP+hepcidin-2h and CLP+hepcidin-24 h. Blood samples were collected at 6, 12 and 24 hours after CLP surgery, and the mice were euthanized and livers were obtained. Results ELISA revealed that hepcidin pretreatment, especially 2 hours in advance (p<0.01), increased the serum hepcidin, TNF-a and IL-6 in CLP-induced septic mice; the serum iron content of CLP-related septic mice decreased (P<0.01), while the liver iron content increased (P<0.01); Hepcidin pretreatment reduced the serum iron (P<0.05) at 6 h and 12 h and liver iron concentrations (P<0.01) at 6 h, 12 h and 24 h in CLP-related septic mice. Western blotting revealed that the hepatic iron absorption-related proteins transferrin receptor-2 (TFR2), ZRT/IRT-like protein 14 (ZIP14) and divalent meta lion transporter-1 (DMT1) were elevated (P<0.01); The iron-exporting protein ferroportin (SLC40A1) was decreased (P<0.01) throughout CLP and CLP+hepcidin sepsis. Compared with CLP group, the protein expressions in the CLP+ hepcidin-2 h group were more obvious than that in the CLP+ hepcidin-24 h group. Conclusion Hepcidin has proinflammatory effect. Hepcidin exacerbates iron metabolism imbalances in sepsis by influencing the expression of iron absorption-related proteins and iron export-related proteins.
Collapse
Affiliation(s)
- Liyan Wu
- Department of Infectious Diseases and Clinical Microbiology, Beijing Chaoyang Hospital, Capital Medical University, Beijing, 100043, People’s Republic of China
| | - Zhenyan Yuan
- Department of Infectious Diseases and Clinical Microbiology, Beijing Chaoyang Hospital, Capital Medical University, Beijing, 100043, People’s Republic of China
| | - Min Wang
- Department of Infectious Diseases and Clinical Microbiology, Beijing Chaoyang Hospital, Capital Medical University, Beijing, 100043, People’s Republic of China
| | - Xiaomeng Fu
- Department of Infectious Diseases and Clinical Microbiology, Beijing Chaoyang Hospital, Capital Medical University, Beijing, 100043, People’s Republic of China
| | - Xiaohui Liu
- Department of Infectious Diseases and Clinical Microbiology, Beijing Chaoyang Hospital, Capital Medical University, Beijing, 100043, People’s Republic of China
| | - Bing Wei
- Emergency Medicine Clinical Research Center, Beijing Chaoyang Hospital, Capital Medical University, & Beijing Key Laboratory of Cardiopulmonary Cerebral Resuscitation, Clinical Center for Medicine in Acute Infection, Capital Medical University, Beijing, 100043, People’s Republic of China
| | - Yugeng Liu
- Department of Infectious Diseases and Clinical Microbiology, Beijing Chaoyang Hospital, Capital Medical University, Beijing, 100043, People’s Republic of China
| |
Collapse
|
36
|
Darand M, Golpour-Hamedani S, Karimi E, Hassanizadeh S, Mirzaei M, Arabi V, Nadjarzadeh A, Hosseinzadeh M. The association between adherence to unhealthy plant-based diet and risk of COVID-19: a cross-sectional study. BMC Infect Dis 2024; 24:1245. [PMID: 39501206 PMCID: PMC11536890 DOI: 10.1186/s12879-024-10115-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2023] [Accepted: 10/24/2024] [Indexed: 11/09/2024] Open
Abstract
BACKGROUND The fast spread of the coronavirus disease 2019 (COVID-19) epidemic and its high mortality were quickly considered by the health community. Dietary patterns play an important role in strengthening or weakening the immune system and thus incidence of diseases. AIM The present study can provide a comprehensive picture of the association between adherence to unhealthy plant-based diet (uPDI) and COVID-19 incidence. METHODS This study was undertaken on 8157 adults' participants of the Yazd Health Study (YaHS) and Taghzieh Mardom-e-Yazd (TAMIZ) study aged 20 to 70 years. Data on dietary intakes were obtained using a validated food frequency questionnaire (FFQ). Multivariable logistic regression analysis was used to assess the association between uPDI and COVID-19. RESULTS We found a significant association between uPDI and the risk of COVID-19 (OR: 1.36; 95% CI: 1.05-1.75) in the crude model. After adjusting potential confounders, a significant increasing trend in the odds of COVID-19 across increasing quintiles of uPDI (OR: 1.58;95% CI: 1.05-2.37; P-value: 0.027) was observed. Stratified analysis based on sex indicated that uPDI significantly increased the risk of COVID-19 only in males (OR: 1.73;95% CI: 1.12-2.67; P-value: 0.012) and had no effect on females. CONCLUSIONS Participants in the highest quintiles of the uPDI had 58% higher odds of COVID-19 compared to subjects in the lowest quintile of uPDI. Although our study has promising results, stronger clinical studies are needed.
Collapse
Affiliation(s)
- Mina Darand
- Prevention of Cardiovascular Disease Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Sahar Golpour-Hamedani
- Department of Community Nutrition, School of Nutrition and Food Science, Food Security Research Center, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Elham Karimi
- Research Development Center, Arash Women's Hospital, Tehran University of Medical Sciences, Tehran, Iran
- Department of Clinical Nutrition, School of Nutrition and Food Science, Food Security Research Center, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Shirin Hassanizadeh
- Department of Clinical Nutrition, School of Nutrition and Food Science, Food Security Research Center, Isfahan University of Medical Sciences, Isfahan, Iran
- Student Research Committee, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Masoud Mirzaei
- Yazd Cardiovascular Research Centre, Non-Communicable Diseases Research Institute, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Vahid Arabi
- Research Center for Food Hygiene and Safety, School of Public Health, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
- Department of Nutrition, School of Public Health, Shahid Sadughi University of Medical Sciences, Yazd, Iran
| | - Azadeh Nadjarzadeh
- Research Center for Food Hygiene and Safety, School of Public Health, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
- Department of Nutrition, School of Public Health, Shahid Sadughi University of Medical Sciences, Yazd, Iran
| | - Mahdieh Hosseinzadeh
- Research Center for Food Hygiene and Safety, School of Public Health, Shahid Sadoughi University of Medical Sciences, Yazd, Iran.
- Department of Nutrition, School of Public Health, Shahid Sadughi University of Medical Sciences, Yazd, Iran.
| |
Collapse
|
37
|
Yin D, Mao R, Wang D, Yu P, Zhou C, Liu J, Li S, Nie Y, Liao H, Peng C. Association of Plasma Metal Levels with Outcomes of Assisted Reproduction in Polycystic Ovary Syndrome. Biol Trace Elem Res 2024; 202:4961-4977. [PMID: 38441797 DOI: 10.1007/s12011-024-04085-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/23/2023] [Accepted: 01/28/2024] [Indexed: 10/01/2024]
Abstract
The objective of this study is to explore the correlation of metal levels with assisted reproductive technology (ART) outcomes in polycystic ovary syndrome (PCOS) patients. The individuals were recruited who met the research criteria, only tubal factor or male infertility served as the control group (n = 40) and patient group was PCOS patients (n = 35). Individuals (n = 75) were divided into PCOS group (n = 35) and control group (n = 40). The normal body mass index (BMI) group (control) includes women with BMI < 25 kg/m2 in PCOS group (n = 24) and control group (n = 33), and BMI ≥ 25 kg/m2 in PCOS group (n = 11) and control group (n = 7). We performed an analysis of insulin resistance (IR) (n = 15) group and without insulin resistance (NIR) group (n = 20) in PCOS patient and control patients. Comparing difference demographic data, ART outcomes and the metal levels in every group respectively, the correlation of metal levels and ART outcomes in control participants and PCOS patients were analyzed by the Spearman correlation analysis, and multiple linear regression model was used to examine the association between the concentration of 19 metals and ART outcomes in PCOS group and control group. Plasma manganese (Mn), titanium (Ti), sodium (Na), magnesium (Mg), copper (Cu), calcium (Ca)/Mg ratio, and Cu/zinc (Zn) ratio levels in PCOS patients were higher than that in control, while Zn and Ca levels were lower in PCOS patients than that in control. The Mg levels had a positive connection with the number of eggs recovered, and the iron (Fe) levels were positively associated with the number of transplanted embryos in PCOS-IR. In PCOS-NIR, Mn levels positively correlated with the number of follicles and the number of good embryos. Silver (Ag) levels were negatively correlated with the number of follicles, and aluminum (Al) levels were negatively related with the normal fertilization and the number of good embryos. The Spearman analysis in PCOS-BMI ≥ 25 group exhibited that nickel (Ni) levels were negatively associated with the number of follicles. The plasma metal levels seem to affect the clinical manifestations and in vitro fertilization outcomes in assisted reproduction.
Collapse
Affiliation(s)
- Dan Yin
- Department of Cell Biology and Genetics, Institute of Cytology and Genetics, School of Basic Medical Sciences, Hengyang Medical School, Key Laboratory of Ecological Environment and Critical Human Diseases Prevention of Hunan Province, Department of Education, Key Laboratory of Hengyang City On Biological Toxicology and Ecological Restoration, Key Laboratory of Typical Environmental Pollution and Health Hazards, University of South China, Hengyang, 421001, Hunan, China
| | - Rui Mao
- Department of Cell Biology and Genetics, Institute of Cytology and Genetics, School of Basic Medical Sciences, Hengyang Medical School, Key Laboratory of Ecological Environment and Critical Human Diseases Prevention of Hunan Province, Department of Education, Key Laboratory of Hengyang City On Biological Toxicology and Ecological Restoration, Key Laboratory of Typical Environmental Pollution and Health Hazards, University of South China, Hengyang, 421001, Hunan, China
| | - Die Wang
- Department of Cell Biology and Genetics, Institute of Cytology and Genetics, School of Basic Medical Sciences, Hengyang Medical School, Key Laboratory of Ecological Environment and Critical Human Diseases Prevention of Hunan Province, Department of Education, Key Laboratory of Hengyang City On Biological Toxicology and Ecological Restoration, Key Laboratory of Typical Environmental Pollution and Health Hazards, University of South China, Hengyang, 421001, Hunan, China
| | - Ping Yu
- Department of Cell Biology and Genetics, Institute of Cytology and Genetics, School of Basic Medical Sciences, Hengyang Medical School, Key Laboratory of Ecological Environment and Critical Human Diseases Prevention of Hunan Province, Department of Education, Key Laboratory of Hengyang City On Biological Toxicology and Ecological Restoration, Key Laboratory of Typical Environmental Pollution and Health Hazards, University of South China, Hengyang, 421001, Hunan, China
| | - Cuilan Zhou
- Department of Cell Biology and Genetics, Institute of Cytology and Genetics, School of Basic Medical Sciences, Hengyang Medical School, Key Laboratory of Ecological Environment and Critical Human Diseases Prevention of Hunan Province, Department of Education, Key Laboratory of Hengyang City On Biological Toxicology and Ecological Restoration, Key Laboratory of Typical Environmental Pollution and Health Hazards, University of South China, Hengyang, 421001, Hunan, China
| | - Jun Liu
- Department of Cell Biology and Genetics, Institute of Cytology and Genetics, School of Basic Medical Sciences, Hengyang Medical School, Key Laboratory of Ecological Environment and Critical Human Diseases Prevention of Hunan Province, Department of Education, Key Laboratory of Hengyang City On Biological Toxicology and Ecological Restoration, Key Laboratory of Typical Environmental Pollution and Health Hazards, University of South China, Hengyang, 421001, Hunan, China
| | - Suyun Li
- Department of Cell Biology and Genetics, Institute of Cytology and Genetics, School of Basic Medical Sciences, Hengyang Medical School, Key Laboratory of Ecological Environment and Critical Human Diseases Prevention of Hunan Province, Department of Education, Key Laboratory of Hengyang City On Biological Toxicology and Ecological Restoration, Key Laboratory of Typical Environmental Pollution and Health Hazards, University of South China, Hengyang, 421001, Hunan, China
| | - Yulin Nie
- Department of Obstetrics and Gynecology, the Second Affiliated Hospital, University of South China, 30# Jiefang Road, Hengyang, 421001, Hunan, China
| | - Hongqing Liao
- Department of Obstetrics and Gynecology, the Second Affiliated Hospital, University of South China, 30# Jiefang Road, Hengyang, 421001, Hunan, China.
| | - Cuiying Peng
- Department of Cell Biology and Genetics, Institute of Cytology and Genetics, School of Basic Medical Sciences, Hengyang Medical School, Key Laboratory of Ecological Environment and Critical Human Diseases Prevention of Hunan Province, Department of Education, Key Laboratory of Hengyang City On Biological Toxicology and Ecological Restoration, Key Laboratory of Typical Environmental Pollution and Health Hazards, University of South China, Hengyang, 421001, Hunan, China.
| |
Collapse
|
38
|
Iriarte-Gahete M, Tarancon-Diez L, Garrido-Rodríguez V, Leal M, Pacheco YM. Absolute and functional iron deficiency: Biomarkers, impact on immune system, and therapy. Blood Rev 2024; 68:101227. [PMID: 39142965 DOI: 10.1016/j.blre.2024.101227] [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: 06/14/2024] [Revised: 08/02/2024] [Accepted: 08/07/2024] [Indexed: 08/16/2024]
Abstract
Iron is essential for numerous physiological processes and its deficiency often leads to anemia. Iron deficiency (ID) is a global problem, primarily affecting reproductive-age women and children, especially in developing countries. Diagnosis uses classical biomarkers like ferritin or transferrin saturation. Recent advancements include using soluble transferrin receptor (sTfR) or hepcidin for improved detection and classification of absolute and functional iron deficiencies, though mostly used in research. ID without anemia may present symptoms like asthenia and fatigue, even without relevant clinical consequences. ID impacts not only red-blood cells but also immune system cells, highlighting its importance in global health and immune-related comorbidities. Managing ID, requires addressing its cause and selecting appropriate iron supplementation. Various improved oral and intravenous products are available, but further research is needed to refine treatment strategies. This review updates on absolute and functional iron deficiencies, their relationships with the immune system and advancements in diagnosis and therapies.
Collapse
Affiliation(s)
- Marianela Iriarte-Gahete
- Immunology Service, Unit of Clinical Laboratories, Institute of Biomedicine of Seville, IBiS / Virgen del Rocío University Hospital / CSIC / University of Seville, Seville, Spain
| | - Laura Tarancon-Diez
- Group of Infections in the Pediatric Population, Health Research Institute Gregorio Marañón (IiSGM), Madrid, Spain; Centro de Investigación Biomédica en Red de Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
| | - Vanesa Garrido-Rodríguez
- Immunology Service, Unit of Clinical Laboratories, Institute of Biomedicine of Seville, IBiS / Virgen del Rocío University Hospital / CSIC / University of Seville, Seville, Spain
| | - Manuel Leal
- Internal Medicine Service, Viamed Santa Ángela de la Cruz Hospital, Seville, Spain
| | - Yolanda María Pacheco
- Immunology Service, Unit of Clinical Laboratories, Institute of Biomedicine of Seville, IBiS / Virgen del Rocío University Hospital / CSIC / University of Seville, Seville, Spain; Universidad Loyola Andalucía, Facultad de Ciencias de la Salud, Campus Sevilla, 41704, Dos Hermanas, Sevilla, Spain.
| |
Collapse
|
39
|
Liu S, Yin J, Wan D, Yin Y. The Role of Iron in Intestinal Mucus: Perspectives from Both the Host and Gut Microbiota. Adv Nutr 2024; 15:100307. [PMID: 39341502 PMCID: PMC11533511 DOI: 10.1016/j.advnut.2024.100307] [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: 06/02/2024] [Revised: 09/10/2024] [Accepted: 09/22/2024] [Indexed: 10/01/2024] Open
Abstract
Although research on the role of iron in host immunity has a history spanning decades, it is only relatively recently that attention has been directed toward the biological effects of iron on the intestinal mucus layer, prompted by an evolving understanding of the role of this material in immune defense. The mucus layer, secreted by intestinal goblet cells, covers the intestinal epithelium, and given its unique location, interactions between the host and gut microbiota, as well as among constituent microbiota, occur frequently within the mucus layer. Iron, as an essential nutrient for the vast majority of life forms, regulates immune responses from both the host and microbial perspectives. In this review, we summarize the iron metabolism of both the host and gut microbiota and describe how iron contributes to intestinal mucosal homeostasis via the intestinal mucus layer with respect to both host and constituent gut microbiota. The findings described herein offer a new perspective on iron-mediated intestinal mucosal barrier function.
Collapse
Affiliation(s)
- Shuan Liu
- Laboratory of Animal Nutritional Physiology and Metabolic Process, Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, Hunan, China; University of Chinese Academy of Sciences, Beijing, China
| | - Jie Yin
- College of Animal Science and Technology, Hunan Agriculture University, Changsha, China
| | - Dan Wan
- Laboratory of Animal Nutritional Physiology and Metabolic Process, Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, Hunan, China.
| | - Yulong Yin
- Laboratory of Animal Nutritional Physiology and Metabolic Process, Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, Hunan, China; University of Chinese Academy of Sciences, Beijing, China.
| |
Collapse
|
40
|
Liu J, Feng G. The causal relationship between trace element status and upper gastrointestinal ulcers: a Mendelian randomization study. Front Nutr 2024; 11:1443090. [PMID: 39539362 PMCID: PMC11557352 DOI: 10.3389/fnut.2024.1443090] [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: 06/03/2024] [Accepted: 10/08/2024] [Indexed: 11/16/2024] Open
Abstract
Background This study aimed to investigate the bidirectional causal relationships between trace elements (such as zinc, magnesium, phosphate, and folate) and upper gastrointestinal ulcers (including gastric and duodenal ulcers). We utilized a two-sample Mendelian randomization (MR) analysis to achieve this. Methods We conducted a two-sample MR analysis using summary-level data from genome-wide association studies (GWAS) obtained from public genomics repositories. We utilized a range of MR methods, including inverse-variance weighted (IVW), MR-Egger, and weighted median methods, and conducted a meta-analysis to synthesize results across different datasets. To ensure the robustness of our findings, we performed extensive sensitivity analyses, including pleiotropy assessment, heterogeneity tests, and leave-one-out analysis. Results Our findings are significant, indicating a positive causal relationship between increased zinc levels and the risk of gastric ulcers. Moreover, magnesium and folate appear to offer potential protective effects against gastroduodenal ulcers (p < 0.05). The meta-analysis further supports the causal relationship between zinc and gastric ulcers (p < 0.05), confirming zinc's significant causal impact on this condition. Conclusion The study confirms a positive causal relationship between zinc and gastric ulcers and highlights the complexity of how trace elements regulate the progression of upper gastrointestinal ulcers. These results provide a scientific basis for dietary recommendations regarding trace element intake in clinical and public health practices. They also offer new insights into effective prevention and treatment strategies for gastric and duodenal ulcers.
Collapse
Affiliation(s)
- Jianwei Liu
- Department of Gastroenterology, Qilu Hospital (Qingdao) of Shandong University, Qingdao, China
| | - Gege Feng
- Department of Hematology, Qilu Hospital of Shandong University, Jinan, China
| |
Collapse
|
41
|
Ru Q, Li Y, Chen L, Wu Y, Min J, Wang F. Iron homeostasis and ferroptosis in human diseases: mechanisms and therapeutic prospects. Signal Transduct Target Ther 2024; 9:271. [PMID: 39396974 PMCID: PMC11486532 DOI: 10.1038/s41392-024-01969-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Revised: 08/08/2024] [Accepted: 09/02/2024] [Indexed: 10/15/2024] Open
Abstract
Iron, an essential mineral in the body, is involved in numerous physiological processes, making the maintenance of iron homeostasis crucial for overall health. Both iron overload and deficiency can cause various disorders and human diseases. Ferroptosis, a form of cell death dependent on iron, is characterized by the extensive peroxidation of lipids. Unlike other kinds of classical unprogrammed cell death, ferroptosis is primarily linked to disruptions in iron metabolism, lipid peroxidation, and antioxidant system imbalance. Ferroptosis is regulated through transcription, translation, and post-translational modifications, which affect cellular sensitivity to ferroptosis. Over the past decade or so, numerous diseases have been linked to ferroptosis as part of their etiology, including cancers, metabolic disorders, autoimmune diseases, central nervous system diseases, cardiovascular diseases, and musculoskeletal diseases. Ferroptosis-related proteins have become attractive targets for many major human diseases that are currently incurable, and some ferroptosis regulators have shown therapeutic effects in clinical trials although further validation of their clinical potential is needed. Therefore, in-depth analysis of ferroptosis and its potential molecular mechanisms in human diseases may offer additional strategies for clinical prevention and treatment. In this review, we discuss the physiological significance of iron homeostasis in the body, the potential contribution of ferroptosis to the etiology and development of human diseases, along with the evidence supporting targeting ferroptosis as a therapeutic approach. Importantly, we evaluate recent potential therapeutic targets and promising interventions, providing guidance for future targeted treatment therapies against human diseases.
Collapse
Affiliation(s)
- Qin Ru
- Institute of Intelligent Sport and Proactive Health, Department of Health and Physical Education, Jianghan University, Wuhan, China
| | - Yusheng Li
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Lin Chen
- Institute of Intelligent Sport and Proactive Health, Department of Health and Physical Education, Jianghan University, Wuhan, China
| | - Yuxiang Wu
- Institute of Intelligent Sport and Proactive Health, Department of Health and Physical Education, Jianghan University, Wuhan, China.
| | - Junxia Min
- The First Affiliated Hospital, Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, China.
| | - Fudi Wang
- The Second Affiliated Hospital, School of Public Health, State Key Laboratory of Experimental Hematology, Zhejiang University School of Medicine, Hangzhou, China.
| |
Collapse
|
42
|
Nonejuie P, Wilantho A, McDonald D, Htoo HH, Chalerm J, Tripathi A, Ngamphiw C, Tongsima S, Knight R, Paiboonsukwong K, Fucharoen S. Differential gut microbiota composition in β-Thalassemia patients and its correlation with iron overload. Sci Rep 2024; 14:23858. [PMID: 39394230 PMCID: PMC11470119 DOI: 10.1038/s41598-024-75456-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Accepted: 10/04/2024] [Indexed: 10/13/2024] Open
Abstract
Recent research highlights the significant impact of the gut microbiota on health and disease. Thalassemia, a hereditary blood disorder, requires regular blood transfusions, leading to an accumulation of iron in the body. Such changes could potentially alter the intestinal microbiota, thereby increasing the susceptibility of thalassemic patients to infection. In this study, we analyzed the fecal microbiota of 70 non-transfusion-dependent (NTDT) β-thalassemia/HbE patients and 30 healthy controls. Our findings indicate that iron chelation intervention had no detectable effect on the microbiome profile of thalassemic patients. However, the cross-sectional analysis revealed that the bacterial diversity and community structure in patients were significantly less diverse and distinct compared to those of healthy subjects. Using reference frames, we were also able to demonstrate that bacterial taxa that are known to produce short chain fatty acids, from the genera Alistipes, Coprococcus, and Oscillospira, and those from the family Ruminococcaceae, were less prevalent in the patients. In contrast, bacterial taxa associated with an unhealthy gut, including the genus Clostridium and those from the families Fusobacteriaceae, Enterobacteriaceae, and Peptostrptococcaceae, were more prevalent in patients and found to be correlated with higher levels of ferritin. Collectively, these changes in the microbiota could be regarded as markers of raised ferritin levels, and therefore, awareness should be exercised as they could interfere, albeit indirectly, with the treatment of the co-morbidities of thalassemia.
Collapse
Affiliation(s)
- Poochit Nonejuie
- Institute of Molecular Biosciences, Mahidol University, Nakhon Pathom, Thailand
| | - Alisa Wilantho
- National Center for Genetic Engineering and Biotechnology (BIOTEC), National Biobank of Thailand, Pathum Thani, Thailand
| | - Daniel McDonald
- Department of Pediatrics, University of California San Diego, La Jolla, CA, USA
| | - Htut Htut Htoo
- Institute of Molecular Biosciences, Mahidol University, Nakhon Pathom, Thailand
| | - Jenjira Chalerm
- Thalassemia Research Center, Institute of Molecular Biosciences, Mahidol University, Nakhon Pathom, Thailand
| | - Anupriya Tripathi
- Department of Pediatrics, University of California San Diego, La Jolla, CA, USA
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, La Jolla, CA, USA
| | - Chumpol Ngamphiw
- National Center for Genetic Engineering and Biotechnology (BIOTEC), National Biobank of Thailand, Pathum Thani, Thailand
| | - Sissades Tongsima
- National Center for Genetic Engineering and Biotechnology (BIOTEC), National Biobank of Thailand, Pathum Thani, Thailand
| | - Rob Knight
- Department of Pediatrics, University of California San Diego, La Jolla, CA, USA
- Shu Chien-Gene Lay Department of Engineering, University of California San Diego, La Jolla, CA, USA
- Department of Computer Science and Engineering, University of California San Diego, La Jolla, CA, USA
- Halıcıoğlu Data Science Institute, University of California San Diego, La Jolla, CA, USA
| | - Kittiphong Paiboonsukwong
- Thalassemia Research Center, Institute of Molecular Biosciences, Mahidol University, Nakhon Pathom, Thailand
| | - Suthat Fucharoen
- Thalassemia Research Center, Institute of Molecular Biosciences, Mahidol University, Nakhon Pathom, Thailand.
| |
Collapse
|
43
|
Davaanyam D, Seol SI, Oh SA, Lee H, Lee JK. Hepatocyte activation and liver injury following cerebral ischemia promote HMGB1-mediated hepcidin upregulation in hepatocytes and regulation of systemic iron levels. Exp Mol Med 2024; 56:2171-2183. [PMID: 39349828 PMCID: PMC11541749 DOI: 10.1038/s12276-024-01314-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: 12/26/2023] [Accepted: 06/24/2024] [Indexed: 10/03/2024] Open
Abstract
We previously reported that high mobility group box 1 (HMGB1), a danger-associated molecular pattern (DAMP), increases intracellular iron levels in the postischemic brain by upregulating hepcidin, a key regulator of iron homeostasis, triggering ferroptosis. Since hepatocytes are the primary cells that produce hepcidin and control systemic iron levels, we investigated whether cerebral ischemia induces hepcidin upregulation in hepatocytes. Following middle cerebral artery occlusion (MCAO) in a rodent model, significant liver injury was observed. This injury was evidenced by significantly elevated Eckhoff's scores and increased serum levels of alanine aminotransferase (ALT) and aspartate aminotransferase (AST). Additionally, total iron levels were significantly elevated in the liver, with intracellular iron accumulation detected in hepatocytes. Hepcidin expression in the liver, which is primarily localized in hepatocytes, increased significantly starting at 3 h after MCAO and continued to increase rapidly, reaching a peak at 24 h. Interestingly, HMGB1 levels in the liver were also significantly elevated after MCAO, with the disulfide form of HMGB1 being the major subtype. In vitro experiments using AML12 hepatocytes showed that recombinant disulfide HMGB1 significantly upregulated hepcidin expression in a Toll-like receptor 4 (TLR4)- and RAGE-dependent manner. Furthermore, treatment with a ROS scavenger and a peptide HMGB1 antagonist revealed that both ROS generation and HMGB1 induction contributed to hepatocyte activation and liver damage following MCAO-reperfusion. In conclusion, this study revealed that cerebral ischemia triggers hepatocyte activation and liver injury. HMGB1 potently induces hepcidin not only in the brain but also in the liver, thereby influencing systemic iron homeostasis following ischemic stroke.
Collapse
Affiliation(s)
- Dashdulam Davaanyam
- Department of Anatomy, Inha University School of Medicine, Incheon, 22212, Korea
| | - Song-I Seol
- Department of Anatomy, Inha University School of Medicine, Incheon, 22212, Korea
| | - Sang-A Oh
- Department of Anatomy, Inha University School of Medicine, Incheon, 22212, Korea
| | - Hahnbi Lee
- Department of Anatomy, Inha University School of Medicine, Incheon, 22212, Korea
| | - Ja-Kyeong Lee
- Department of Anatomy, Inha University School of Medicine, Incheon, 22212, Korea.
| |
Collapse
|
44
|
Reid BM. Early life stress and iron metabolism in developmental psychoneuroimmunology. Brain Behav Immun Health 2024; 40:100824. [PMID: 39161875 PMCID: PMC11331713 DOI: 10.1016/j.bbih.2024.100824] [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: 11/29/2023] [Revised: 06/03/2024] [Accepted: 07/15/2024] [Indexed: 08/21/2024] Open
Abstract
An estimated 250 million children face adverse health outcomes from early life exposure to severe or chronic social, economic, and nutritional adversity, highlighting/emphasizing the pressing concern about the link between ELS and long-term implications on mental and physical health. There is significant overlap between populations experiencing high levels of chronic stress and those experiencing iron deficiency, spotlighting the potential role of iron as a key mediator in this association. Iron, an essential micronutrient for brain development and immune function, is often depleted in stress conditions. Iron deficiency among the most common nutrient deficiencies in the world. Fetal and infant iron status may thus serve as a crucial intermediary between early chronic psychological stress and subsequent immune system changes to impact neurodevelopment. The review presents a hypothesized pathway between early life stress (ELS), iron deficiency, and neurodevelopment through the hypothalamic-pituitary-adrenocortical (HPA) axis and the IL-6-hepcidin axis. This hypothesis is derived from (1) evidence that stress impacts iron status (2) long-term neurodevelopmental outcomes that are shared by ELS and iron deficiency exposure, and (3) possible mechanisms for how iron may mediate the relation between ELS and iron deficiency through alterations in the developing immune system. The article concludes by proposing future research directions, emphasizing the need for rigorous studies to elucidate how stress and iron metabolism interact to modify the developing immune system. Understanding these mechanisms could open new avenues for improving human health and neurodevelopment for women and children globally, making it a timely and vital area of study in psychoneuroimmunology research.
Collapse
Affiliation(s)
- Brie M. Reid
- Department of Psychiatry and Human Behavior, Warren Alpert Medical School, Brown University, USA
- Center for Behavioral and Preventive Medicine, The Miriam Hospital, USA
- Department of Psychology, Department of Health Sciences, Northeastern University, USA
| |
Collapse
|
45
|
Babar S, Saboor M. Erythroferrone in focus: emerging perspectives in iron metabolism and hematopathologies. BLOOD SCIENCE 2024; 6:e00198. [PMID: 39027903 PMCID: PMC11254117 DOI: 10.1097/bs9.0000000000000198] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2024] [Accepted: 06/25/2024] [Indexed: 07/20/2024] Open
Abstract
Beyond its core role in iron metabolism, erythroferrone (ERFE) has emerged as a key player with far-reaching implications in various hematologic disorders. Its regulatory effect on hepcidin underlines its significance in conditions characterized by disrupted iron homeostasis. In β-thalassemia and myelodysplastic syndromes, its dysregulation intricately contributes to the clinical challenges of anemia and iron overload which highlights its potential as a therapeutic target. In anemia of chronic disease and iron deficiency anemia, ERFE presents a unique profile. In chronic kidney disease (CKD), the intricate interplay between ERFE, erythropoietin, and hepcidin undergoes dysregulation, contributing to the complex iron imbalance characteristic of this condition. Recent research suggests that ERFE plays a multifaceted role in restoring iron balance in CKD, beyond simply suppressing hepcidin production. The potential to modulate ERFE activity offers a novel approach to treating a spectrum of disorders associated with iron dysregulation. As our understanding of ERFE continues to evolve, it is poised to become a key focus in the development of targeted treatments, making it an exciting and dynamic area of ongoing research. Modulating ERFE activity presents a groundbreaking approach to treat iron dysregulation in conditions like iron deficiency anemia, thalassemia, and hemochromatosis. As new research unveils its intricate roles, ERFE has rapidly emerged as a key target for developing targeted therapies like ERFE agonists and antagonists. With promising studies underway, this dynamic field holds immense potential to improve patient outcomes, reduce complications, and offer personalized treatment options in hematology research. This comprehensive overview of ERFE's role across various conditions underscores its pivotal function in iron metabolism and associated pathologies.
Collapse
Affiliation(s)
- Sadia Babar
- Baqai Institute of Hematology, Baqai Medical University, Karachi, Pakistan
- Baqai Institute of Medical Technology, Baqai Medical University, Karachi, Pakistan
| | - Muhammad Saboor
- Department of Medical Laboratory Sciences, College of Health Sciences, University of Sharjah, Sharjah, United Arab Emirates
- Research Institute for Medical and Health Sciences, University of Sharjah, Sharjah, United Arab Emirates
| |
Collapse
|
46
|
Imelio JA, Trajtenberg F, Mondino S, Zarantonelli L, Vitrenko I, Lemée L, Cokelaer T, Picardeau M, Buschiazzo A. Signal-sensing triggers the shutdown of HemKR, regulating heme and iron metabolism in the spirochete Leptospira biflexa. PLoS One 2024; 19:e0311040. [PMID: 39325783 PMCID: PMC11426443 DOI: 10.1371/journal.pone.0311040] [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: 07/12/2024] [Accepted: 09/11/2024] [Indexed: 09/28/2024] Open
Abstract
Heme and iron metabolic pathways are highly intertwined, both compounds being essential for key biological processes, yet becoming toxic if overabundant. Their concentrations are exquisitely regulated, including via dedicated two-component systems (TCSs) that sense signals and regulate adaptive responses. HemKR is a TCS present in both saprophytic and pathogenic Leptospira species, involved in the control of heme metabolism. However, the molecular means by which HemKR is switched on/off in a signal-dependent way, are still unknown. Moreover, a comprehensive list of HemKR-regulated genes, potentially overlapped with iron-responsive targets, is also missing. Using the saprophytic species Leptospira biflexa as a model, we now show that 5-aminolevulinic acid (ALA) triggers the shutdown of the HemKR pathway in live cells, and does so by stimulating the phosphatase activity of HemK towards phosphorylated HemR. Phospho~HemR dephosphorylation leads to differential expression of multiple genes, including of heme metabolism and transport systems. Besides the heme-biosynthetic genes hemA and the catabolic hmuO, which we had previously reported as phospho~HemR targets, we now extend the regulon identifying additional genes. Finally, we discover that HemR inactivation brings about an iron-deficit tolerant phenotype, synergistically with iron-responsive signaling systems. Future studies with pathogenic Leptospira will be able to confirm whether such tolerance to iron deprivation is conserved among Leptospira spp., in which case HemKR could play a vital role during infection where available iron is scarce. In sum, HemKR responds to abundance of porphyrin metabolites by shutting down and controlling heme homeostasis, while also contributing to integrate the regulation of heme and iron metabolism in the L. biflexa spirochete model.
Collapse
Affiliation(s)
- Juan Andrés Imelio
- Laboratory of Molecular & Structural Microbiology, Institut Pasteur de Montevideo, Montevideo, Uruguay
| | - Felipe Trajtenberg
- Laboratory of Molecular & Structural Microbiology, Institut Pasteur de Montevideo, Montevideo, Uruguay
| | - Sonia Mondino
- Laboratory of Molecular & Structural Microbiology, Institut Pasteur de Montevideo, Montevideo, Uruguay
| | | | - Iakov Vitrenko
- Plateforme Technologique Biomics, C2RT, Institut Pasteur, Université Paris Cité, Paris, France
| | - Laure Lemée
- Plateforme Technologique Biomics, C2RT, Institut Pasteur, Université Paris Cité, Paris, France
| | - Thomas Cokelaer
- Plateforme Technologique Biomics, C2RT, Institut Pasteur, Université Paris Cité, Paris, France
- Bioinformatics and Biostatistics Hub, Institut Pasteur, Paris, France
| | - Mathieu Picardeau
- Biology of Spirochetes Unit, Institut Pasteur, Université Paris Cité, Paris, France
| | - Alejandro Buschiazzo
- Laboratory of Molecular & Structural Microbiology, Institut Pasteur de Montevideo, Montevideo, Uruguay
- Dept of Microbiology, Institut Pasteur, Université Paris Cité, Paris, France
| |
Collapse
|
47
|
Strobel S, Sian-Hulsmann J, Tappe D, Jellinger K, Riederer P, Monoranu CM. Postencephalitic Parkinsonism: Unique Pathological and Clinical Features-Preliminary Data. Cells 2024; 13:1511. [PMID: 39329695 PMCID: PMC11430219 DOI: 10.3390/cells13181511] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Revised: 08/29/2024] [Accepted: 09/04/2024] [Indexed: 09/28/2024] Open
Abstract
Postencephalitic parkinsonism (PEP) is suggested to show a virus-induced pathology, which is different from classical idiopathic Parkinson's disease (PD) as there is no α-synuclein/Lewy body pathology. However, PEP shows a typical clinical representation of motor disturbances. In addition, compared to PD, there is no iron-induced pathology. The aim of this preliminary study was to compare PEP with PD regarding iron-induced pathology, using histochemistry methods on paraffin-embedded post-mortem brain tissue. In the PEP group, iron was not seen, except for one case with sparse perivascular depositions. Rather, PEP offers a pathology related to tau-protein/neurofibrillary tangles, with mild to moderate memory deficits only. It is assumed that this virus-induced pathology is due to immunological dysfunctions causing (neuro)inflammation-induced neuronal network disturbances as events that trigger clinical parkinsonism. The absence of iron deposits implies that PEP cannot be treated with iron chelators. The therapy with L-Dopa is also not an option, as L-Dopa only leads to an initial slight improvement in symptoms in isolated cases.
Collapse
Affiliation(s)
- Sabrina Strobel
- Institute of Pathology, Department of Neuropathology, University of Wuerzburg, 97080 Wuerzburg, Germany;
| | - Jeswinder Sian-Hulsmann
- Department of Medical Physiology, University of Nairobi, P.O. Box 30197, Nairobi 00100, Kenya;
| | - Dennis Tappe
- Bernhard Nocht Institute for Tropical Medicine, 20359 Hamburg, Germany;
| | - Kurt Jellinger
- Institute of Clinical Neurobiology, A-1150 Vienna, Austria;
| | - Peter Riederer
- Clinic and Policlinic for Psychiatry, Psychosomatics and Psychotherapy, University Hospital Wuerzburg, 97080 Wuerzburg, Germany;
- Department of Psychiatry, University of Southern Denmark, 5000 Odense, Denmark
| | - Camelia-Maria Monoranu
- Institute of Pathology, Department of Neuropathology, University of Wuerzburg, 97080 Wuerzburg, Germany;
| |
Collapse
|
48
|
Torres-Escobar A, Wilkins A, Juárez-Rodríguez MD, Circu M, Latimer B, Dragoi AM, Ivanov SS. Iron-depleting nutritional immunity controls extracellular bacterial replication in Legionella pneumophila infections. Nat Commun 2024; 15:7848. [PMID: 39245746 PMCID: PMC11381550 DOI: 10.1038/s41467-024-52184-x] [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: 02/02/2024] [Accepted: 08/29/2024] [Indexed: 09/10/2024] Open
Abstract
The accidental human pathogen Legionella pneumophila (Lp) is the etiological agent for a severe atypical pneumonia known as Legionnaires' disease. In human infections and animal models of disease alveolar macrophages are the primary cellular niche that supports bacterial replication within a unique intracellular membrane-bound organelle. The Dot/Icm apparatus-a type IV secretion system that translocates ~300 bacterial proteins within the cytosol of the infected cell-is a central virulence factor required for intracellular growth. Mutant strains lacking functional Dot/Icm apparatus are transported to and degraded within the lysosomes of infected macrophages. The early foundational work from Dr. Horwitz's group unequivocally established that Legionella does not replicate extracellularly during infection-a phenomenon well supported by experimental evidence for four decades. Our data challenges this paradigm by demonstrating that macrophages and monocytes provide the necessary nutrients and support robust Legionella extracellular replication. We show that the previously reported lack of Lp extracellular replication is not a bacteria intrinsic feature but rather a result of robust restriction by serum-derived nutritional immunity factors. Specifically, the host iron-sequestering protein Transferrin is identified here as a critical suppressor of Lp extracellular replication in an iron-dependent manner. In iron-overload conditions or in the absence of Transferrin, Lp bypasses growth restriction by IFNγ-primed macrophages though extracellular replication. It is well established that certain risk factors associated with development of Legionnaires' disease, such as smoking, produce a chronic pulmonary environment of iron-overload. Our work indicates that iron-overload could be an important determinant of severe infection by allowing Lp to overcome nutritional immunity and replicate extracellularly, which in turn would circumvent intracellular cell intrinsic host defenses. Thus, we provide evidence for nutritional immunity as a key underappreciated host defense mechanism in Legionella pathogenesis.
Collapse
Affiliation(s)
- Ascención Torres-Escobar
- Department of Microbiology and Immunology, Louisiana State University Health Sciences Center-Shreveport, Shreveport, LA, 71130, USA
| | - Ashley Wilkins
- Department of Microbiology and Immunology, Louisiana State University Health Sciences Center-Shreveport, Shreveport, LA, 71130, USA
- Bacterial Physiology and Metabolism Unit, Laboratory of Bacteriology, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, 59840, USA
| | - María D Juárez-Rodríguez
- Department of Molecular and Cellular Physiology, Louisiana State University Health Sciences Center-Shreveport, Shreveport, LA, 71130, USA
| | - Magdalena Circu
- Department of Microbiology and Immunology, Louisiana State University Health Sciences Center-Shreveport, Shreveport, LA, 71130, USA
| | - Brian Latimer
- Innovative North Louisiana Experimental Therapeutics program (INLET), Feist-Weiller Cancer Center, Louisiana State University Health Sciences Center-Shreveport, Shreveport, LA, 71130, USA
| | - Ana-Maria Dragoi
- Department of Molecular and Cellular Physiology, Louisiana State University Health Sciences Center-Shreveport, Shreveport, LA, 71130, USA
- Innovative North Louisiana Experimental Therapeutics program (INLET), Feist-Weiller Cancer Center, Louisiana State University Health Sciences Center-Shreveport, Shreveport, LA, 71130, USA
| | - Stanimir S Ivanov
- Department of Microbiology and Immunology, Louisiana State University Health Sciences Center-Shreveport, Shreveport, LA, 71130, USA.
| |
Collapse
|
49
|
Delanoy G, Lupardus C, Vali SW, Wofford JD, Thorat S, Lindahl PA. Mössbauer and EPR detection of iron trafficking kinetics and possibly labile iron pools in whole Saccharomyces cerevisiae cells. J Biol Chem 2024; 300:107711. [PMID: 39178945 PMCID: PMC11422575 DOI: 10.1016/j.jbc.2024.107711] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2024] [Revised: 08/06/2024] [Accepted: 08/19/2024] [Indexed: 08/26/2024] Open
Abstract
The kinetics of iron trafficking in whole respiring Saccharomyces cerevisiae cells were investigated using Mössbauer and EPR spectroscopies. The Mössbauer-active isotope 57Fe was added to cells growing under iron-limited conditions; cells were analyzed at different times post iron addition. Spectroscopic changes suggested that the added 57Fe initially entered the labile iron pool, and then distributed to vacuoles and mitochondria. The first spectroscopic feature observed, ∼ 3 min after adding 57Fe plus a 5 to 15 min processing dead time, was a quadrupole doublet typical of nonheme high-spin FeII. This feature likely arose from labile FeII pools in the cell. At later times (15-150 min), magnetic features due to S = 5/2 FeIII developed; these likely arose from FeIII in vacuoles. Corresponding EPR spectra were dominated by a g = 4.3 signal from the S = 5/2 FeIII ions that increased in intensity over time. Developing at a similar rate was a quadrupole doublet typical of S = 0 [Fe4S4]2+ clusters and low-spin FeII hemes; such centers are mainly in mitochondria, cytosol, and nuclei. Development of these features was simulated using a published mathematical model, and simulations compared qualitatively well with observations. In the five sets of experiments presented, all spectroscopic features developed within the doubling time of the cells, implying that the detected iron trafficking species are physiologically relevant. These spectroscopy-based experiments allow the endogenous labile iron pool within growing cells to be detected without damaging or altering the pool, as definitely occurs using chelator-probe detection and possibly occurs using chromatographic separations.
Collapse
Affiliation(s)
- Grant Delanoy
- Department of Chemistry, Texas A&M University, College Station, Texas, USA
| | - Cody Lupardus
- Department of Biochemistry and Biophysics, Texas A&M University, College Station, Texas, USA
| | - Shaik Waseem Vali
- Department of Biochemistry and Biophysics, Texas A&M University, College Station, Texas, USA
| | - Joshua D Wofford
- Department of Chemistry, Texas A&M University, College Station, Texas, USA
| | - Shantanu Thorat
- Department of Computer Science and Engineering, Texas A&M University, College Station, Texas, USA
| | - Paul A Lindahl
- Department of Chemistry, Texas A&M University, College Station, Texas, USA; Department of Biochemistry and Biophysics, Texas A&M University, College Station, Texas, USA.
| |
Collapse
|
50
|
Murante D, Hogan DA. Drivers of diversification in fungal pathogen populations. PLoS Pathog 2024; 20:e1012430. [PMID: 39264909 PMCID: PMC11392411 DOI: 10.1371/journal.ppat.1012430] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/14/2024] Open
Abstract
To manage and treat chronic fungal diseases effectively, we require an improved understanding of their complexity. There is an increasing appreciation that chronic infection populations are often heterogeneous due to diversification and drift, even within a single microbial species. Genetically diverse populations can contribute to persistence and resistance to treatment by maintaining cells with different phenotypes capable of thriving in these dynamic environments. In chronic infections, fungal pathogens undergo prolonged challenges that can drive trait selection to convergent adapted states through restricted access to critical nutrients, assault by immune effectors, competition with other species, and antifungal drugs. This review first highlights the various genetic and epigenetic mechanisms that promote diversity in pathogenic fungal populations and provide an additional barrier to assessing the actual heterogeneity of fungal infections. We then review existing studies of evolution and genetic heterogeneity in fungal populations from lung infections associated with the genetic disease cystic fibrosis. We conclude with a discussion of open research questions that, once answered, may aid in diagnosing and treating chronic fungal infections.
Collapse
Affiliation(s)
- Daniel Murante
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Hanover, New Hampshire, United States of America
| | - Deborah Ann Hogan
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Hanover, New Hampshire, United States of America
| |
Collapse
|