1
|
Wang TW, Zhou LL, Yuan J, Zhou WX, Wang HR, Yu TT, Zhai JC, Tang CB, Jiang W, Yu JQ, Zheng RQ, Yu HL, Shao J. Study of the relationship between iron metabolism disorders and sepsis-associated liver injury: A prospective observational study. World J Gastroenterol 2025; 31:104584. [PMID: 40248384 PMCID: PMC12001195 DOI: 10.3748/wjg.v31.i14.104584] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/25/2024] [Revised: 02/17/2025] [Accepted: 03/25/2025] [Indexed: 04/11/2025] Open
Abstract
BACKGROUND Sepsis-associated liver injury (SALI) refers to secondary liver function impairment caused by sepsis, patients with SALI often have worse clinical outcomes. The early identification and assessment of the occurrence and progression of SALI are pressing issues that urgently need to be resolved. AIM To investigate the relationship between iron metabolism and SALI. METHODS In this prospective study, 139 patients were recruited, with 53 assigned to the SALI group. The relationships between SALI and various iron metabolism-related biomarkers were examined. These biomarkers included serum iron (SI), total iron-binding capacity (TIBC), serum ferritin, transferrin, and transferrin saturation. To identify independent risk factors for SALI, both univariate and multivariate logistic regression analyses were performed. Additionally, receiver operating characteristic curve analysis was utilized to assess the predictive value of these biomarkers for the occurrence of SALI. RESULTS There were no statistically significant differences in age, sex, body mass index, Sequential Organ Failure Assessment scores (excluding liver function), or APACHE II scores between the two groups of patients. Compared with the sepsis group, the SALI group presented significantly higher SI (P < 0.001), TIBC (P < 0.001), serum ferritin (P = 0.001), transferrin (P = 0.005), and transferrin saturation levels (P < 0.001). Multivariate logistic regression analysis revealed that SI (odds ratio = 1.24, 95% confidence interval: 1.11-1.40, P < 0.001) and TIBC levels (odds ratio = 1.13, 95% confidence interval: 1.05-1.21, P < 0.001) were independent predictors of SALI. Receiver operating characteristic curve analysis revealed that SI and TIBC had areas under the curve of 0.816 and 0.757, respectively, indicating moderate predictive accuracy for SALI. CONCLUSION Iron metabolism disorders are closely associated with the development of SALI, and SI and TIBC may serve as potential predictive biomarkers. The combined use of SI and TIBC has superior diagnostic efficacy for SALI. These findings provide valuable insights for the early identification and management of SALI among patients with sepsis.
Collapse
Affiliation(s)
- Tian-Wei Wang
- Department of Intensive Care Unit, Northern Jiangsu People’s Hospital Affiliated to Yangzhou University, Yangzhou 225001, Jiangsu Province, China
| | - Lu-Lu Zhou
- Department of Intensive Care Unit, Northern Jiangsu People’s Hospital Affiliated to Yangzhou University, Yangzhou 225001, Jiangsu Province, China
| | - Jing Yuan
- Department of Functional Examination, Northern Jiangsu People’s Hospital Affiliated to Yangzhou University, Yangzhou 225001, Jiangsu Province, China
| | - Wen-Xin Zhou
- Department of Oncology, First Affiliated Hospital of Nanjing Medical University, Nanjing 210000, Jiangsu Province, China
| | - Hao-Ran Wang
- Department of Intensive Care Unit, Northern Jiangsu People’s Hospital Affiliated to Yangzhou University, Yangzhou 225001, Jiangsu Province, China
| | - Ting-Ting Yu
- Department of Intensive Care Unit, Northern Jiangsu People’s Hospital Affiliated to Yangzhou University, Yangzhou 225001, Jiangsu Province, China
| | - Ji-Chao Zhai
- Department of Intensive Care Unit, Northern Jiangsu People’s Hospital Affiliated to Yangzhou University, Yangzhou 225001, Jiangsu Province, China
| | - Cheng-Bin Tang
- Department of Center for Cardiac Macrovascular Disease, Northern Jiangsu People’s Hospital Affiliated to Yangzhou University, Yangzhou 225001, Jiangsu Province, China
| | - Wei Jiang
- Department of Intensive Care Unit, Northern Jiangsu People’s Hospital Affiliated to Yangzhou University, Yangzhou 225001, Jiangsu Province, China
| | - Jiang-Quan Yu
- Department of Intensive Care Unit, Northern Jiangsu People’s Hospital Affiliated to Yangzhou University, Yangzhou 225001, Jiangsu Province, China
| | - Rui-Qiang Zheng
- Department of Intensive Care Unit, Northern Jiangsu People’s Hospital Affiliated to Yangzhou University, Yangzhou 225001, Jiangsu Province, China
| | - Hai-Long Yu
- Department of Neuro Intensive Care Unit, Northern Jiangsu People’s Hospital Affiliated to Yangzhou University, Yangzhou 225001, Jiangsu Province, China
- Department of Neurology, Northern Jiangsu People’s Hospital Affiliated to Yangzhou University, Yangzhou 225001, Jiangsu Province, China
| | - Jun Shao
- Department of Center for Cardiac Macrovascular Disease, Northern Jiangsu People’s Hospital Affiliated to Yangzhou University, Yangzhou 225001, Jiangsu Province, China
| |
Collapse
|
2
|
Sun Y, Ren Y, Song LY, Wang YY, Li TG, Wu YL, Li L, Yang ZS. Targeting iron-metabolism:a potential therapeutic strategy for pulmonary fibrosis. Biomed Pharmacother 2024; 172:116270. [PMID: 38364737 DOI: 10.1016/j.biopha.2024.116270] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2023] [Revised: 01/23/2024] [Accepted: 02/07/2024] [Indexed: 02/18/2024] Open
Abstract
Iron homeostasisis is integral to normal physiological and biochemical processes of lungs. The maintenance of iron homeostasis involves the process of intake, storage and output, dependening on iron-regulated protein/iron response element system to operate tightly metabolism-related genes, including TFR1, DMT1, Fth, and FPN. Dysregulation of iron can lead to iron overload, which increases the virulence of microbial colonisers and the occurrence of oxidative stress, causing alveolar epithelial cells to undergo necrosis and apoptosis, and form extracellular matrix. Accumulated iron drive iron-dependent ferroptosis to exacerbated pulmonary fibrosis. Notably, the iron chelator deferoxamine and the lipophilic antioxidant ferritin-1 have been shown to attenuate ferroptosis and inhibit lipid peroxidation in pulmonary fibrosis. The paper summarises the regulatory mechanisms of dysregulated iron metabolism and ferroptosis in the development of pulmonary fibrosis. Targeting iron metabolism may be a potential therapeutic strategy for the prevention and treatment of pulmonary fibrosis.
Collapse
Affiliation(s)
- Yi Sun
- Yunnan Provincial Key Laboratory of Integrated Traditional Chinese and Western Medicine for Chronic Disease in Prevention and Treatment, School of Basic Medical Sciences, Yunnan University of Chinese Medicine, 1076 Yuhua Road, Kunming, 650500, Yunnan, China
| | - Yu Ren
- Yunnan Provincial Key Laboratory of Integrated Traditional Chinese and Western Medicine for Chronic Disease in Prevention and Treatment, School of Basic Medical Sciences, Yunnan University of Chinese Medicine, 1076 Yuhua Road, Kunming, 650500, Yunnan, China
| | - Li-Yun Song
- Yunnan Provincial Key Laboratory of Integrated Traditional Chinese and Western Medicine for Chronic Disease in Prevention and Treatment, School of Basic Medical Sciences, Yunnan University of Chinese Medicine, 1076 Yuhua Road, Kunming, 650500, Yunnan, China
| | - Yin-Ying Wang
- The First School of Clinical Medicine, Yunnan University of Chinese Medicine, 1076 Yuhua Road Kunming, Yunnan 650500, China
| | - Tian-Gang Li
- Yunnan Provincial Key Laboratory of Integrated Traditional Chinese and Western Medicine for Chronic Disease in Prevention and Treatment, School of Basic Medical Sciences, Yunnan University of Chinese Medicine, 1076 Yuhua Road, Kunming, 650500, Yunnan, China
| | - Ying-Li Wu
- Yunnan Provincial Key Laboratory of Integrated Traditional Chinese and Western Medicine for Chronic Disease in Prevention and Treatment, School of Basic Medical Sciences, Yunnan University of Chinese Medicine, 1076 Yuhua Road, Kunming, 650500, Yunnan, China
| | - Li Li
- Yunnan Provincial Key Laboratory of Integrated Traditional Chinese and Western Medicine for Chronic Disease in Prevention and Treatment, School of Basic Medical Sciences, Yunnan University of Chinese Medicine, 1076 Yuhua Road, Kunming, 650500, Yunnan, China.
| | - Zhong-Shan Yang
- Yunnan Provincial Key Laboratory of Integrated Traditional Chinese and Western Medicine for Chronic Disease in Prevention and Treatment, School of Basic Medical Sciences, Yunnan University of Chinese Medicine, 1076 Yuhua Road, Kunming, 650500, Yunnan, China.
| |
Collapse
|
3
|
Noordine ML, Seyoum Y, Bruneau A, Baye K, Lefebvre T, Cherbuy C, Canonne-Hergaux F, Nicolas G, Humblot C, Thomas M. The microbiota and the host organism switch between cooperation and competition based on dietary iron levels. Gut Microbes 2024; 16:2361660. [PMID: 38935764 PMCID: PMC11212566 DOI: 10.1080/19490976.2024.2361660] [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: 02/13/2024] [Accepted: 05/24/2024] [Indexed: 06/29/2024] Open
Abstract
The microbiota significantly impacts digestive epithelium functionality, especially in nutrient processing. Given the importance of iron for both the host and the microbiota, we hypothesized that host-microbiota interactions fluctuate with dietary iron levels. We compared germ-free (GF) and conventional mice (SPF) fed iron-containing (65 mg/Kg) or iron-depleted (<6 mg/Kg) diets. The efficacy of iron privation was validated by iron blood parameters. Ferritin and Dmt1, which represent cellular iron storage and transport respectively, were studied in tissues where they are abundant: the duodenum, liver and lung. When the mice were fed an iron-rich diet, the microbiota increased blood hemoglobin and hepcidin and the intestinal ferritin levels, suggesting that the microbiota helps iron storage. When iron was limiting, the microbiota inhibited the expression of the intestinal Dmt1 transporter, likely via the pathway triggered by Hif-2α. The microbiota assists the host in storing intestinal iron when it is abundant and competes with the host by inhibiting Dmt1 in conditions of iron scarcity. Comparison between duodenum, liver and lung indicates organ-specific responses to microbiota and iron availability. Iron depletion induced temporal changes in microbiota composition and activity, reduced α-diversity of microbiota, and led to Lactobacillaceae becoming particularly more abundant after 60 days of privation. By inoculating GF mice with a simplified bacterial mixture, we show that the iron-depleted host favors the gut fitness of Bifidobacterium longum.
Collapse
Affiliation(s)
- Marie-Louise Noordine
- Micalis Institute, Institut national de recherche pour l’agriculture, l’alimentation et l’environnement (INRAE), AgroParisTech, Université Paris-Saclay, UMR1319, Jouy-en-Josas, France
- Center for Microbiome Medicine (PaCeMM) FHU, AP-HP, Paris, Ile-de-France, France
| | - Yohannes Seyoum
- Micalis Institute, Institut national de recherche pour l’agriculture, l’alimentation et l’environnement (INRAE), AgroParisTech, Université Paris-Saclay, UMR1319, Jouy-en-Josas, France
- Center for Food Science and Nutrition, College of Natural and Computational Sciences, Addis Ababa University, Addis Ababa, Ethiopia
- QualiSud, Université de Montpellier, Avignon Université, CIRAD, Institut Agro, IRD, Université de la Réunion, Montpellier Cedex, France
| | - Aurélia Bruneau
- Micalis Institute, Institut national de recherche pour l’agriculture, l’alimentation et l’environnement (INRAE), AgroParisTech, Université Paris-Saclay, UMR1319, Jouy-en-Josas, France
- Center for Microbiome Medicine (PaCeMM) FHU, AP-HP, Paris, Ile-de-France, France
| | - Kaleab Baye
- Center for Food Science and Nutrition, College of Natural and Computational Sciences, Addis Ababa University, Addis Ababa, Ethiopia
| | - Thibaud Lefebvre
- Assistance Publique-Hôpitaux de Paris, Centre Français des Porphyries, Hôpital Louis Mourier, Colombes, France
- Institut National de la Santé et de la Recherche Médicale, U1149, Centre de Recherches sur l’Inflammation, Paris, France
| | - Claire Cherbuy
- Micalis Institute, Institut national de recherche pour l’agriculture, l’alimentation et l’environnement (INRAE), AgroParisTech, Université Paris-Saclay, UMR1319, Jouy-en-Josas, France
- Center for Microbiome Medicine (PaCeMM) FHU, AP-HP, Paris, Ile-de-France, France
| | - François Canonne-Hergaux
- IRSD, Université de Toulouse, INSERM, INRAE, ENVT, Univ Toulouse III - Paul Sabatier (UPS), Toulouse, France
- U1188 DéTROI, Université de La Réunion, Paris, France
| | - Gaël Nicolas
- Institut National de la Santé et de la Recherche Médicale, U1149, Centre de Recherches sur l’Inflammation, Paris, France
- Université Paris Diderot, site Bichat, Sorbonne Paris Cité, Paris, Ile-de-France, France
| | - Christèle Humblot
- QualiSud, Université de Montpellier, Avignon Université, CIRAD, Institut Agro, IRD, Université de la Réunion, Montpellier Cedex, France
| | - Muriel Thomas
- Micalis Institute, Institut national de recherche pour l’agriculture, l’alimentation et l’environnement (INRAE), AgroParisTech, Université Paris-Saclay, UMR1319, Jouy-en-Josas, France
- Center for Microbiome Medicine (PaCeMM) FHU, AP-HP, Paris, Ile-de-France, France
| |
Collapse
|
4
|
Hopkins CD, Wessel C, Chen O, El-Kersh K, Cave MC, Cai L, Huang J. Potential Roles of Metals in the Pathogenesis of Pulmonary and Systemic Hypertension. Int J Biol Sci 2023; 19:5036-5054. [PMID: 37928257 PMCID: PMC10620830 DOI: 10.7150/ijbs.85590] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Accepted: 09/08/2023] [Indexed: 11/07/2023] Open
Abstract
Pulmonary and systemic hypertension (PH, SH) are characterized by vasoconstriction and vascular remodeling resulting in increased vascular resistance and pulmonary/aortic artery pressures. The chronic stress leads to inflammation, oxidative stress, and infiltration by immune cells. Roles of metals in these diseases, particularly PH are largely unknown. This review first discusses the pathophysiology of PH including vascular oxidative stress, inflammation, and remodeling in PH; mitochondrial dysfunction and metabolic changes in PH; ion channel and its alterations in the pathogenesis of PH as well as PH-associated right ventricular (RV) remodeling and dysfunctions. This review then summarizes metal general features and essentiality for the cardiovascular system and effects of metals on systemic blood pressure. Lastly, this review explores non-essential and essential metals and potential roles of their dyshomeostasis in PH and RV dysfunction. Although it remains early to conclude the role of metals in the pathogenesis of PH, emerging direct and indirect evidence implicates the possible contributions of metal-mediated toxicities in the development of PH. Future research should focus on comprehensive clinical metallomics study in PH patients; mechanistic evaluations to elucidate roles of various metals in PH animal models; and novel therapy clinical trials targeting metals. These important discoveries will significantly advance our understandings of this rare yet fatal disease, PH.
Collapse
Affiliation(s)
- C. Danielle Hopkins
- Department of Anesthesiology and Perioperative Medicine, University of Louisville School of Medicine, Louisville, KY, USA
| | - Caitlin Wessel
- Department of Anesthesiology and Perioperative Medicine, University of Louisville School of Medicine, Louisville, KY, USA
| | - Oscar Chen
- Department of Anesthesiology and Perioperative Medicine, University of Louisville School of Medicine, Louisville, KY, USA
| | - Karim El-Kersh
- Department of Internal Medicine, Division of Pulmonary Critical Care and Sleep Medicine, University of Nebraska Medical Center, Omaha, NE, USA
| | - Matthew C. Cave
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Medicine, University of Louisville School of Medicine, Louisville, KY, USA
- The Center for Integrative Environmental Health Sciences, University of Louisville, Louisville, KY, 40202, USA
- Department of Pharmacology and Toxicology, University of Louisville School of Medicine, Louisville, KY, USA
- Department of Biochemistry and Molecular Genetics, University of Louisville School of Medicine, Louisville, KY, USA
- The Transplant Program at UofL Health - Jewish Hospital Trager Transplant Center, Louisville, KY, USA
| | - Lu Cai
- The Center for Integrative Environmental Health Sciences, University of Louisville, Louisville, KY, 40202, USA
- Department of Pharmacology and Toxicology, University of Louisville School of Medicine, Louisville, KY, USA
- Pediatric Research Institute, Department of Pediatrics, University of Louisville School of Medicine, Louisville, KY, USA
- Department of Radiation Oncology, University of Louisville School of Medicine, Louisville, KY, USA
| | - Jiapeng Huang
- Department of Anesthesiology and Perioperative Medicine, University of Louisville School of Medicine, Louisville, KY, USA
- The Center for Integrative Environmental Health Sciences, University of Louisville, Louisville, KY, 40202, USA
- Department of Pharmacology and Toxicology, University of Louisville School of Medicine, Louisville, KY, USA
- The Transplant Program at UofL Health - Jewish Hospital Trager Transplant Center, Louisville, KY, USA
- Cardiovascular Innovation Institute, Department of Cardiovascular and Thoracic Surgery, University of Louisville School of Medicine, Louisville, KY, USA
| |
Collapse
|
5
|
Abstract
Iron is essential to the virulence of Aspergillus species, and restricting iron availability is a critical mechanism of antimicrobial host defense. Macrophages recruited to the site of infection are at the crux of this process, employing multiple intersecting mechanisms to orchestrate iron sequestration from pathogens. To gain an integrated understanding of how this is achieved in aspergillosis, we generated a transcriptomic time series of the response of human monocyte-derived macrophages to Aspergillus and used this and the available literature to construct a mechanistic computational model of iron handling of macrophages during this infection. We found an overwhelming macrophage response beginning 2 to 4 h after exposure to the fungus, which included upregulated transcription of iron import proteins transferrin receptor-1, divalent metal transporter-1, and ZIP family transporters, and downregulated transcription of the iron exporter ferroportin. The computational model, based on a discrete dynamical systems framework, consisted of 21 3-state nodes, and was validated with additional experimental data that were not used in model generation. The model accurately captures the steady state and the trajectories of most of the quantitatively measured nodes. In the experimental data, we surprisingly found that transferrin receptor-1 upregulation preceded the induction of inflammatory cytokines, a feature that deviated from model predictions. Model simulations suggested that direct induction of transferrin receptor-1 (TfR1) after fungal recognition, independent of the iron regulatory protein-labile iron pool (IRP-LIP) system, explains this finding. We anticipate that this model will contribute to a quantitative understanding of iron regulation as a fundamental host defense mechanism during aspergillosis. IMPORTANCE Invasive pulmonary aspergillosis is a major cause of death among immunosuppressed individuals despite the best available therapy. Depriving the pathogen of iron is an essential component of host defense in this infection, but the mechanisms by which the host achieves this are complex. To understand how recruited macrophages mediate iron deprivation during the infection, we developed and validated a mechanistic computational model that integrates the available information in the field. The insights provided by this approach can help in designing iron modulation therapies as anti-fungal treatments.
Collapse
|
6
|
Role of Iron in Aging Related Diseases. Antioxidants (Basel) 2022; 11:antiox11050865. [PMID: 35624729 PMCID: PMC9137504 DOI: 10.3390/antiox11050865] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Revised: 04/17/2022] [Accepted: 04/25/2022] [Indexed: 02/05/2023] Open
Abstract
Iron progressively accumulates with age and can be further exacerbated by dietary iron intake, genetic factors, and repeated blood transfusions. While iron plays a vital role in various physiological processes within the human body, its accumulation contributes to cellular aging in several species. In its free form, iron can initiate the formation of free radicals at a cellular level and contribute to systemic disorders. This is most evident in high iron conditions such as hereditary hemochromatosis, when accumulation of iron contributes to the development of arthritis, cirrhosis, or cardiomyopathy. A growing body of research has further identified iron’s contributory effects in neurodegenerative diseases, ocular disorders, cancer, diabetes, endocrine dysfunction, and cardiovascular diseases. Reducing iron levels by repeated phlebotomy, iron chelation, and dietary restriction are the common therapeutic considerations to prevent iron toxicity. Chelators such as deferoxamine, deferiprone, and deferasirox have become the standard of care in managing iron overload conditions with other potential applications in cancer and cardiotoxicity. In certain animal models, drugs with iron chelating ability have been found to promote health and even extend lifespan. As we further explore the role of iron in the aging process, iron chelators will likely play an increasingly important role in our health.
Collapse
|
7
|
Ho T, Nichols M, Nair G, Radford K, Kjarsgaard M, Huang C, Bhalla A, Lavigne N, Mukherjee M, Surette M, Macri J, Nair P. Iron in airway macrophages and infective exacerbations of chronic obstructive pulmonary disease. Respir Res 2022; 23:8. [PMID: 35022042 PMCID: PMC8756761 DOI: 10.1186/s12931-022-01929-7] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Accepted: 01/04/2022] [Indexed: 12/31/2022] Open
Abstract
Background Excess pulmonary iron has been implicated in the pathogenesis of lung disease, including asthma and COPD. An association between higher iron content in sputum macrophages and infective exacerbations of COPD has previously been demonstrated. Objectives To assess the mechanisms of pulmonary macrophage iron sequestration, test the effect of macrophage iron-loading on cellular immune function, and prospectively determine if sputum hemosiderin index can predict infectious exacerbations of COPD. Methods Intra- and extracellular iron was measured in cell-line-derived and in freshly isolated sputum macrophages under various experimental conditions including treatment with exogenous IL-6 and hepcidin. Bacterial uptake and killing were compared in the presence or absence of iron-loading. A prospective cohort of COPD patients with defined sputum hemosiderin indices were monitored to determine the annual rate of severe infectious exacerbations. Results Gene expression studies suggest that airway macrophages have the requisite apparatus of the hepcidin-ferroportin axis. IL-6 and hepcidin play roles in pulmonary iron sequestration, though IL-6 appears to exert its effect via a hepcidin-independent mechanism. Iron-loaded macrophages had reduced uptake of COPD-relevant organisms and were associated with higher growth rates. Infectious exacerbations were predicted by sputum hemosiderin index (β = 0.035, p = 0.035). Conclusions We demonstrate in-vitro and population-level evidence that excess iron in pulmonary macrophages may contribute to recurrent airway infection in COPD. Specifically, IL-6-dependent iron sequestration by sputum macrophages may result in immune cell dysfunction and ultimately lead to increased frequency of infective exacerbation. Supplementary Information The online version contains supplementary material available at 10.1186/s12931-022-01929-7.
Collapse
Affiliation(s)
- Terence Ho
- Department of Medicine, McMaster University, Hamilton, Canada. .,Firestone Institute for Respiratory Health, St. Joseph's Healthcare Hamilton, 50 Charlton Avenue East, Hamilton, ON, L8N 4A6, Canada.
| | - Matthew Nichols
- Department of Pathology and Laboratory Medicine, Western University, London, Canada
| | - Gayatri Nair
- Department of Medicine, McMaster University, Hamilton, Canada
| | | | | | - Chynna Huang
- St. Joseph's Healthcare Hamilton, Hamilton, Canada
| | - Anurag Bhalla
- Department of Medicine, McMaster University, Hamilton, Canada.,Firestone Institute for Respiratory Health, St. Joseph's Healthcare Hamilton, 50 Charlton Avenue East, Hamilton, ON, L8N 4A6, Canada
| | | | | | - Michael Surette
- Department of Medicine, McMaster University, Hamilton, Canada
| | - Joseph Macri
- Department of Pathology and Molecular Medicine, McMaster University, Hamilton, Canada
| | - Parameswaran Nair
- Department of Medicine, McMaster University, Hamilton, Canada.,Firestone Institute for Respiratory Health, St. Joseph's Healthcare Hamilton, 50 Charlton Avenue East, Hamilton, ON, L8N 4A6, Canada
| |
Collapse
|
8
|
Li S, Zhang H, Chang J, Li D, Cao P. Iron overload and mitochondrial dysfunction orchestrate pulmonary fibrosis. Eur J Pharmacol 2021; 912:174613. [PMID: 34740581 DOI: 10.1016/j.ejphar.2021.174613] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2021] [Revised: 09/06/2021] [Accepted: 09/11/2021] [Indexed: 12/26/2022]
Abstract
Pulmonary fibrosis (PF) is a chronic, progressive heterogeneous disease of lung tissues with poor lung function caused by scar tissue. Due to our limited understanding of its mechanism, there is currently no treatment strategy that can prevent the development of PF. In recent years, iron accumulation and mitochondrial damage have been reported to participate in PF, and drugs that reduce iron content and improve mitochondrial function have shown significant efficacy in animal experimental models. Excessive iron leads to mitochondrial impairment, which may be the key cause that results in the dysfunction of various kinds of pulmonary cells and further promotes PF. As an emerging research hotspot, there are few targeted effective therapeutic strategies at present due to limited mechanistic understanding. In this review, the roles of iron homeostasis imbalance and mitochondrial damage in PF are summarized and discussed, highlighting a promising direction for finding truly effective therapeutics for PF.
Collapse
Affiliation(s)
- Shuxin Li
- Ministry of Education Key Laboratory of Molecular and Cellular Biology, Key Laboratory of Animal Physiology, Biochemistry and Molecular Biology of Hebei Province, College of Life Sciences, Hebei Normal University, Shijiazhuang, Hebei, 050024, People's Republic of China
| | - Hongmin Zhang
- Ministry of Education Key Laboratory of Molecular and Cellular Biology, Key Laboratory of Animal Physiology, Biochemistry and Molecular Biology of Hebei Province, College of Life Sciences, Hebei Normal University, Shijiazhuang, Hebei, 050024, People's Republic of China
| | - Jing Chang
- Ministry of Education Key Laboratory of Molecular and Cellular Biology, Key Laboratory of Animal Physiology, Biochemistry and Molecular Biology of Hebei Province, College of Life Sciences, Hebei Normal University, Shijiazhuang, Hebei, 050024, People's Republic of China
| | - Dongming Li
- Ministry of Education Key Laboratory of Molecular and Cellular Biology, Key Laboratory of Animal Physiology, Biochemistry and Molecular Biology of Hebei Province, College of Life Sciences, Hebei Normal University, Shijiazhuang, Hebei, 050024, People's Republic of China.
| | - Pengxiu Cao
- Ministry of Education Key Laboratory of Molecular and Cellular Biology, Key Laboratory of Animal Physiology, Biochemistry and Molecular Biology of Hebei Province, College of Life Sciences, Hebei Normal University, Shijiazhuang, Hebei, 050024, People's Republic of China.
| |
Collapse
|
9
|
Han J, Wang K. Clinical significance of serum hepcidin in the diagnosis and treatment of patients with anemia of chronic disease: a meta-analysis. Biomarkers 2021; 26:296-301. [PMID: 33653208 DOI: 10.1080/1354750x.2021.1893812] [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: 07/14/2020] [Accepted: 01/28/2021] [Indexed: 10/22/2022]
Abstract
OBJECTIVE To systematically evaluate the value of serum hepcidin in the diagnosis of Anaemia of Chronic Disease (ACD) in order to provide appropriate treatment. METHODS Literature search was performed in PubMed, EMbase, Cochrane Library, CNKI, CBMdisc and CSPD till Jan, 2020. Studies using hepcidin assay for the diagnosis of ACD were included. Two researchers selected the literature according to the pre-defined inclusion and exclusion criteria. Meta-analysis was performed using Stata 15.0. RESULTS A total of 10 studies were included, including 739 patients with 402 ACD patients. Heterogeneity test results suggest that there is no statistical heterogeneity between the included studies and Meta- analysis was performed using a fixed-effects model. Results showed that serum hepcidin levels in patients with ACD combined with SEN, SPE, PLR, NLR, and Diagnostic OR were 0.94 [95% CI (0.90, 0.96)], 0.85 [95% CI (0.81, 0.88)], 6.1 [95% CI (4.8, 7.6)], 0.08 [95% CI (0.05, 0.12)] and 81 [95% CI (47, 139)] respectively. The area under the SROC curve (AUC) value was 0.91. CONCLUSION Serum hepcidin assay is a valuable method to diagnose ACD in patients. However, due to the limitations of the quantity and quality of the research, the above conclusions need more research to verify.
Collapse
Affiliation(s)
- Jie Han
- Department of Hepatology, Qilu Hospital of Shandong University and Hepatology Institute of Shandong University, Jinan, China
| | - Kai Wang
- Department of Hepatology, Qilu Hospital of Shandong University and Hepatology Institute of Shandong University, Jinan, China
| |
Collapse
|
10
|
Jo JR, Lee SE, An S, Nedumaran B, Ghosh S, Park KG, Kim YD. Gluconeogenic signals regulate hepcidin gene expression via a CRBN-KLF15 axis. BMB Rep 2021. [PMID: 33795032 PMCID: PMC8093939 DOI: 10.5483/bmbrep.2021.54.4.215] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Abstract
Hepcidin (HAMP) is synthesized in the liver. It is a key iron-regulatory hormone that controls systemic iron homeostasis. Cereblon (CRBN) and Kruppel-like factor 15 (KLF15) are known to regulate diverse physiological functions. In this study, we investigated the role of CRBN on hepatic hepcidin gene expression and production under gluconeogenic stimuli. Fasted mice as well as forskolin (FSK)- and glucagon (GLU)-treated mice had reduced serum iron levels but increased expression levels of hepatic Crbn and Klf15 and hepcidin secretion. MicroRNA (miRNA) expression analysis of fasted and Ad-Crbn-infected mice revealed significant reduction of microRNA-639 (miR-639). Hepatic overexpression of Crbn elevated hepcidin expression and production along with Klf15 gene expression, whereas knockdown of Crbn and Klf15 markedly decreased FSK- and fasting-mediated induction of hepcidin gene expression and its biosynthesis in mouse livers and primary hepatocytes. Moreover, expression of KLF15 significantly increased the activity of hepcidin reporter gene. It was exclusively dependent on the KLF15-binding site identified within the hepcidin gene promoter. Overall, this study demonstrates that CRBN and KLF15 are novel mediators of gluconeogenic signal-induced hepcidin gene expression and production. Thus, CRBN and KLF15 might be novel potential therapeutic targets to intervene metabolic dysfunction.
Collapse
Affiliation(s)
- Jeong-Rang Jo
- Research Institute of Aging and Metabolism, Kyungpook National University, Daegu 41566, Korea
- Department of Internal Medicine, School of Medicine, Kyungpook National University, Kyungpook National University Hospital, Daegu 41944, Korea
| | - Sung-Eun Lee
- Department of Applied Biosciences, Kyungpook National University, Daegu 41566, Korea
| | - Seungwon An
- Department of Ophthalmology and Visual Sciences, University of Illinois at Chicago, Chicago, Illinois 60612, USA
| | - Balachandar Nedumaran
- Barbara Davis Center for Diabetes, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado 80045, USA
| | - Swati Ghosh
- Department of Pediatrics, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado 80045, USA
| | - Keun-Gyu Park
- Research Institute of Aging and Metabolism, Kyungpook National University, Daegu 41566, Korea
- Department of Internal Medicine, School of Medicine, Kyungpook National University, Kyungpook National University Hospital, Daegu 41944, Korea
| | - Yong Deuk Kim
- Research Institute of Aging and Metabolism, Kyungpook National University, Daegu 41566, Korea
- Department of Internal Medicine, School of Medicine, Kyungpook National University, Kyungpook National University Hospital, Daegu 41944, Korea
- Department of Applied Biosciences, Kyungpook National University, Daegu 41566, Korea
| |
Collapse
|
11
|
Liu Q, Wu J, Zhang X, Wu X, Zhao Y, Ren J. Iron homeostasis and disorders revisited in the sepsis. Free Radic Biol Med 2021; 165:1-13. [PMID: 33486088 DOI: 10.1016/j.freeradbiomed.2021.01.025] [Citation(s) in RCA: 59] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/20/2020] [Revised: 12/31/2020] [Accepted: 01/11/2021] [Indexed: 12/26/2022]
Abstract
Sepsis is a life-threatening condition caused by a dysregulated host-response to inflammation, although it currently lacks a fully elucidated pathobiology. Iron is a crucial trace element that is essential for fundamental processes in both humans and bacteria. During sepsis, iron metabolism is altered, including increased iron transport and uptake into cells and decreased iron export. The intracellular sequestration of iron limits its availability to circulating pathogens, which serves as a conservative strategy against the pathogens. Although iron retention has been showed to have protective protect effects, an increase in labile iron may cause oxidative injury and cell death (e.g., pyroptosis, ferroptosis) as the condition progresses. Moreover, iron disorders are substantial and correlate with the severity of sepsis. This also suggests that iron may be useful as a diagnostic marker for evaluating the severity and predicting the outcome of the disease. Further knowledge about these disorders could help in evaluating how drugs targeting iron homeostasis can be optimally applied to improve the treatment of patients with sepsis. Here, we present a comprehensive review of recent advances in the understanding of iron metabolism, focusing on the regulatory mechanisms and iron-mediated injury in sepsis.
Collapse
Affiliation(s)
- Qinjie Liu
- Research Institute of General Surgery, Jinling Hospital, Medical School of Nanjing University, Nanjing, 210002, PR China.
| | - Jie Wu
- Department of General Surgery, BenQ Medical Center, The Affiliated BenQ Hospital of Nanjing Medical University, Nanjing, 210002, PR China.
| | - Xufei Zhang
- Research Institute of General Surgery, Jinling Hospital, Nanjing Medical University, Nanjing, 210002, PR China.
| | - Xiuwen Wu
- Research Institute of General Surgery, Jinling Hospital, Nanjing, 210002, PR China.
| | - Yun Zhao
- Department of General Surgery, BenQ Medical Center, The Affiliated BenQ Hospital of Nanjing Medical University, Nanjing, 210002, PR China.
| | - Jianan Ren
- Research Institute of General Surgery, Jinling Hospital, Medical School of Nanjing University, Nanjing, 210002, PR China; Department of General Surgery, BenQ Medical Center, The Affiliated BenQ Hospital of Nanjing Medical University, Nanjing, 210002, PR China; Research Institute of General Surgery, Jinling Hospital, Nanjing Medical University, Nanjing, 210002, PR China.
| |
Collapse
|
12
|
Marques O, Neves J, Horvat NK, Altamura S, Muckenthaler MU. Mild Attenuation of the Pulmonary Inflammatory Response in a Mouse Model of Hereditary Hemochromatosis Type 4. Front Physiol 2021; 11:589351. [PMID: 33519502 PMCID: PMC7838636 DOI: 10.3389/fphys.2020.589351] [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: 07/30/2020] [Accepted: 12/18/2020] [Indexed: 11/13/2022] Open
Abstract
The respiratory tract is constantly exposed to pathogens that require iron for proliferation and virulence. Pulmonary iron levels are increased in several lung diseases and associated with increased susceptibility to infections. However, regulation of lung iron homeostasis and its cross talk to pulmonary immune responses are largely unexplored. Here we investigated how increased lung iron levels affect the early pulmonary inflammatory response. We induced acute local pulmonary inflammation via aerosolized LPS in a mouse model of hereditary hemochromatosis type 4 (Slc40a1 C326S/C326S), which is hallmarked by systemic and pulmonary iron accumulation, specifically in alveolar macrophages. We show that Slc40a1 C326S/C326S mice display a mild attenuation in the LPS-induced pulmonary inflammatory response, with a reduced upregulation of some pro-inflammatory cytokines and chemokines. Despite mildly reduced cytokine levels, there is no short-term impairment in the recruitment of neutrophils into the bronchoalveolar space. These data suggest that increased pulmonary iron levels do not strongly alter the acute inflammatory response of the lung.
Collapse
Affiliation(s)
- Oriana Marques
- Department of Pediatric Oncology, Hematology, Immunology and Pulmonology, University Hospital Heidelberg, Heidelberg, Germany.,Molecular Medicine Partnership Unit, University of Heidelberg, Heidelberg, Germany
| | - Joana Neves
- Department of Pediatric Oncology, Hematology, Immunology and Pulmonology, University Hospital Heidelberg, Heidelberg, Germany.,Translational Lung Research Center Heidelberg (TLRC), German Center for Lung Research (DZL), University of Heidelberg, Heidelberg, Germany
| | - Natalie K Horvat
- Department of Pediatric Oncology, Hematology, Immunology and Pulmonology, University Hospital Heidelberg, Heidelberg, Germany.,Molecular Medicine Partnership Unit, University of Heidelberg, Heidelberg, Germany.,Translational Lung Research Center Heidelberg (TLRC), German Center for Lung Research (DZL), University of Heidelberg, Heidelberg, Germany.,European Molecular Biology Laboratory, Heidelberg, Germany
| | - Sandro Altamura
- Department of Pediatric Oncology, Hematology, Immunology and Pulmonology, University Hospital Heidelberg, Heidelberg, Germany.,Molecular Medicine Partnership Unit, University of Heidelberg, Heidelberg, Germany
| | - Martina U Muckenthaler
- Department of Pediatric Oncology, Hematology, Immunology and Pulmonology, University Hospital Heidelberg, Heidelberg, Germany.,Molecular Medicine Partnership Unit, University of Heidelberg, Heidelberg, Germany.,Translational Lung Research Center Heidelberg (TLRC), German Center for Lung Research (DZL), University of Heidelberg, Heidelberg, Germany
| |
Collapse
|
13
|
Mleczko‐Sanecka K, Silvestri L. Cell-type-specific insights into iron regulatory processes. Am J Hematol 2021; 96:110-127. [PMID: 32945012 DOI: 10.1002/ajh.26001] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Revised: 08/20/2020] [Accepted: 09/14/2020] [Indexed: 12/16/2022]
Abstract
Despite its essential role in many biological processes, iron is toxic when in excess due to its propensity to generate reactive oxygen species. To prevent diseases associated with iron deficiency or iron loading, iron homeostasis must be tightly controlled. Intracellular iron content is regulated by the Iron Regulatory Element-Iron Regulatory Protein (IRE-IRP) system, whereas systemic iron availability is adjusted to body iron needs chiefly by the hepcidin-ferroportin (FPN) axis. Here, we aimed to review advances in the field that shed light on cell-type-specific regulatory mechanisms that control or modify systemic and local iron balance, and how shifts in cellular iron levels may affect specialized cell functions.
Collapse
Affiliation(s)
| | - Laura Silvestri
- Regulation of Iron Metabolism Unit, Division of Genetics and Cell Biology IRCCS San Raffaele Scientific Institute Milan Italy
- Vita‐Salute San Raffaele University Milan Italy
| |
Collapse
|
14
|
Perez E, Baker JR, Di Giandomenico S, Kermani P, Parker J, Kim K, Yang J, Barnes PJ, Vaulont S, Scandura JM, Donnelly LE, Stout-Delgado H, Cloonan SM. Hepcidin Is Essential for Alveolar Macrophage Function and Is Disrupted by Smoke in a Murine Chronic Obstructive Pulmonary Disease Model. THE JOURNAL OF IMMUNOLOGY 2020; 205:2489-2498. [PMID: 32958690 DOI: 10.4049/jimmunol.1901284] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/23/2019] [Accepted: 08/31/2020] [Indexed: 12/21/2022]
Abstract
Chronic obstructive pulmonary disease (COPD) is a debilitating lung disease associated with cigarette smoking. Alterations in local lung and systemic iron regulation are associated with disease progression and pathogenesis. Hepcidin, an iron regulatory peptide hormone, is altered in subjects with COPD; however, the molecular role of hepcidin in COPD pathogenesis remains to be determined. In this study, using a murine model of smoke-induced COPD, we demonstrate that lung and circulating hepcidin levels are inhibited by cigarette smoke. We show that cigarette smoke exposure increases erythropoietin and bone marrow-derived erythroferrone and leads to expanded but inefficient erythropoiesis in murine bone marrow and an increase in ferroportin on alveolar macrophages (AMs). AMs from smokers and subjects with COPD display increased expression of ferroportin as well as hepcidin. Notably, murine AMs exposed to smoke fail to increase hepcidin in response to Gram-negative or Gram-positive infection. Loss of hepcidin in vivo results in blunted functional responses of AMs and exaggerated responses to Streptococcus pneumoniae infection.
Collapse
Affiliation(s)
- Elizabeth Perez
- Division of Pulmonary and Critical Care Medicine, Joan and Sanford I. Weill Department of Medicine, Weill Cornell Medicine, New York, NY 10065
| | - Jonathan R Baker
- Airway Disease Section, National Heart and Lung Institute, Imperial College London and Royal Brompton Hospital, London SW3 6NP, United Kingdom
| | - Silvana Di Giandomenico
- Division of Hematology and Oncology, Joan and Sanford I. Weill Department of Medicine, Weill Cornell Medicine, New York, NY 10065
| | - Pouneh Kermani
- Division of Hematology and Oncology, Joan and Sanford I. Weill Department of Medicine, Weill Cornell Medicine, New York, NY 10065
| | - Jacqueline Parker
- Division of Pulmonary and Critical Care Medicine, Joan and Sanford I. Weill Department of Medicine, Weill Cornell Medicine, New York, NY 10065.,New York-Presbyterian Hospital, Weill Cornell Medicine, New York, NY 10065
| | - Kihwan Kim
- Division of Pulmonary and Critical Care Medicine, Joan and Sanford I. Weill Department of Medicine, Weill Cornell Medicine, New York, NY 10065
| | - Jianjun Yang
- Division of Pulmonary and Critical Care Medicine, Joan and Sanford I. Weill Department of Medicine, Weill Cornell Medicine, New York, NY 10065
| | - Peter J Barnes
- Airway Disease Section, National Heart and Lung Institute, Imperial College London and Royal Brompton Hospital, London SW3 6NP, United Kingdom
| | - Sophie Vaulont
- Université de Paris, INSERM U1016, Institut Cochin, CNRS UMR8104, 75014 Paris, France.,Laboratory of Excellence GR-Ex, 75015 Paris, France; and
| | - Joseph M Scandura
- Division of Hematology and Oncology, Joan and Sanford I. Weill Department of Medicine, Weill Cornell Medicine, New York, NY 10065.,New York-Presbyterian Hospital, Weill Cornell Medicine, New York, NY 10065
| | - Louise E Donnelly
- Airway Disease Section, National Heart and Lung Institute, Imperial College London and Royal Brompton Hospital, London SW3 6NP, United Kingdom
| | - Heather Stout-Delgado
- Division of Pulmonary and Critical Care Medicine, Joan and Sanford I. Weill Department of Medicine, Weill Cornell Medicine, New York, NY 10065
| | - Suzanne M Cloonan
- Division of Pulmonary and Critical Care Medicine, Joan and Sanford I. Weill Department of Medicine, Weill Cornell Medicine, New York, NY 10065; .,School of Medicine, Trinity College Dublin and Tallaght University Hospital, Dublin D24 NR04, Ireland
| |
Collapse
|
15
|
Zhang V, Ganz T, Nemeth E, Kim A. Iron overload causes a mild and transient increase in acute lung injury. Physiol Rep 2020; 8:e14470. [PMID: 32596989 PMCID: PMC7322498 DOI: 10.14814/phy2.14470] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2020] [Revised: 05/05/2020] [Accepted: 05/07/2020] [Indexed: 12/27/2022] Open
Abstract
Recent studies have demonstrated a strong link between acute respiratory distress syndrome (ARDS) and the levels of iron and iron-related proteins in the lungs. However, the role of iron overload in ARDS development has yet to be characterized. In this study, we compared the highly iron-overloaded hepcidin knockout mice (HKO) to their iron-sufficient wild-type (WT) littermates in a model of sterile acute lung injury (ALI) induced by treatment with oropharyngeal (OP) LPS. There were no major differences in systemic inflammatory response or airway neutrophil infiltration between the two groups at the time of maximal injury (days 2 and 3) or during the recovery phase (day 7). Hepcidin knockout mice had transiently increased bronchoalveolar lavage fluid (BALF) protein and MPO activity in the lung and BALF on day 3, indicating worse vascular leakage and increased neutrophil activity, respectively. The increased ALI severity in iron-overloaded mice may be a result of increased apoptosis of lung tissue, as evidenced by an increase in cleaved capsase-3 protein in lung homogenates from HKO mice versus WT mice on day 3. Altogether, our data suggest that even severe iron overload has a relatively minor and transient effect in LPS-induced ALI.
Collapse
Affiliation(s)
- Vida Zhang
- Department of MedicineDavid Geffen School of MedicineUCLALos AngelesCAUSA
- Department of Molecular and Medical PharmacologyUCLALos AngelesCAUSA
| | - Tomas Ganz
- Department of MedicineDavid Geffen School of MedicineUCLALos AngelesCAUSA
| | - Elizabeta Nemeth
- Department of MedicineDavid Geffen School of MedicineUCLALos AngelesCAUSA
| | - Airie Kim
- Department of MedicineDavid Geffen School of MedicineUCLALos AngelesCAUSA
| |
Collapse
|
16
|
Ali MK, Kim RY, Brown AC, Mayall JR, Karim R, Pinkerton JW, Liu G, Martin KL, Starkey MR, Pillar AL, Donovan C, Pathinayake PS, Carroll OR, Trinder D, Tay HL, Badi YE, Kermani NZ, Guo YK, Aryal R, Mumby S, Pavlidis S, Adcock IM, Weaver J, Xenaki D, Oliver BG, Holliday EG, Foster PS, Wark PA, Johnstone DM, Milward EA, Hansbro PM, Horvat JC. Crucial role for lung iron level and regulation in the pathogenesis and severity of asthma. Eur Respir J 2020; 55:13993003.01340-2019. [PMID: 32184317 DOI: 10.1183/13993003.01340-2019] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2019] [Accepted: 01/28/2020] [Indexed: 01/08/2023]
Abstract
Accumulating evidence highlights links between iron regulation and respiratory disease. Here, we assessed the relationship between iron levels and regulatory responses in clinical and experimental asthma.We show that cell-free iron levels are reduced in the bronchoalveolar lavage (BAL) supernatant of severe or mild-moderate asthma patients and correlate with lower forced expiratory volume in 1 s (FEV1). Conversely, iron-loaded cell numbers were increased in BAL in these patients and with lower FEV1/forced vital capacity (FVC) ratio. The airway tissue expression of the iron sequestration molecules divalent metal transporter 1 (DMT1) and transferrin receptor 1 (TFR1) are increased in asthma, with TFR1 expression correlating with reduced lung function and increased Type-2 (T2) inflammatory responses in the airways. Furthermore, pulmonary iron levels are increased in a house dust mite (HDM)-induced model of experimental asthma in association with augmented Tfr1 expression in airway tissue, similar to human disease. We show that macrophages are the predominant source of increased Tfr1 and Tfr1+ macrophages have increased Il13 expression. We also show that increased iron levels induce increased pro-inflammatory cytokine and/or extracellular matrix (ECM) responses in human airway smooth muscle (ASM) cells and fibroblasts ex vivo and induce key features of asthma in vivo, including airway hyper-responsiveness (AHR) and fibrosis, and T2 inflammatory responses.Together these complementary clinical and experimental data highlight the importance of altered pulmonary iron levels and regulation in asthma, and the need for a greater focus on the role and potential therapeutic targeting of iron in the pathogenesis and severity of disease.
Collapse
Affiliation(s)
- Md Khadem Ali
- Division of Pulmonary and Critical Care Medicine, Stanford University, Stanford, CA, USA.,Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute and School of Biomedical Sciences and Pharmacy, University of Newcastle, Newcastle, Australia
| | - Richard Y Kim
- Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute and School of Biomedical Sciences and Pharmacy, University of Newcastle, Newcastle, Australia.,Centre for Inflammation, Centenary Institute and University of Technology Sydney, Sydney, Australia
| | - Alexandra C Brown
- Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute and School of Biomedical Sciences and Pharmacy, University of Newcastle, Newcastle, Australia
| | - Jemma R Mayall
- Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute and School of Biomedical Sciences and Pharmacy, University of Newcastle, Newcastle, Australia
| | - Rafia Karim
- Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute and School of Biomedical Sciences and Pharmacy, University of Newcastle, Newcastle, Australia
| | - James W Pinkerton
- Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute and School of Biomedical Sciences and Pharmacy, University of Newcastle, Newcastle, Australia.,Respiratory Pharmacology and Toxicology Group, National Heart and Lung Institute, Imperial College London, London, UK
| | - Gang Liu
- Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute and School of Biomedical Sciences and Pharmacy, University of Newcastle, Newcastle, Australia.,Centre for Inflammation, Centenary Institute and University of Technology Sydney, Sydney, Australia
| | - Kristy L Martin
- School of Biomedical Sciences and Pharmacy, University of Newcastle, Newcastle, Australia
| | - Malcolm R Starkey
- Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute and School of Biomedical Sciences and Pharmacy, University of Newcastle, Newcastle, Australia.,Dept of Immunology and Pathology, Central Clinical School, Monash University, Melbourne, Australia
| | - Amber L Pillar
- Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute and School of Biomedical Sciences and Pharmacy, University of Newcastle, Newcastle, Australia
| | - Chantal Donovan
- Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute and School of Biomedical Sciences and Pharmacy, University of Newcastle, Newcastle, Australia.,Centre for Inflammation, Centenary Institute and University of Technology Sydney, Sydney, Australia
| | - Prabuddha S Pathinayake
- Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute and School of Medicine and Public Health, University of Newcastle, Newcastle, Australia
| | - Olivia R Carroll
- Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute and School of Biomedical Sciences and Pharmacy, University of Newcastle, Newcastle, Australia
| | - Debbie Trinder
- Medical School, Harry Perkins Medical Research Institute, University of Western Australia, Fiona Stanley Hospital, Perth, Australia
| | - Hock L Tay
- Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute and School of Biomedical Sciences and Pharmacy, University of Newcastle, Newcastle, Australia
| | - Yusef E Badi
- Airway Disease Section, National Heart and Lung Institute, Imperial College London, London, UK
| | - Nazanin Z Kermani
- Data Science Institute, Dept of Computing, Imperial College London, London, UK
| | - Yi-Ke Guo
- Data Science Institute, Dept of Computing, Imperial College London, London, UK
| | - Ritambhara Aryal
- School of Biomedical Sciences and Pharmacy, University of Newcastle, Newcastle, Australia
| | - Sharon Mumby
- Airway Disease Section, National Heart and Lung Institute, Imperial College London, London, UK
| | - Stelios Pavlidis
- Airway Disease Section, National Heart and Lung Institute, Imperial College London, London, UK
| | - Ian M Adcock
- Airway Disease Section, National Heart and Lung Institute, Imperial College London, London, UK
| | - Jessica Weaver
- Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute and School of Biomedical Sciences and Pharmacy, University of Newcastle, Newcastle, Australia
| | - Dikaia Xenaki
- Woolcock Institute of Medical Research, University of Sydney and School of Life Sciences, University of Technology Sydney, Sydney, Australia
| | - Brian G Oliver
- Woolcock Institute of Medical Research, University of Sydney and School of Life Sciences, University of Technology Sydney, Sydney, Australia
| | - Elizabeth G Holliday
- Hunter Medical Research Institute, New Lambton, Australia.,School of Medicine and Public Health, University of Newcastle, Callaghan, Australia
| | - Paul S Foster
- Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute and School of Biomedical Sciences and Pharmacy, University of Newcastle, Newcastle, Australia
| | - Peter A Wark
- Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute and School of Biomedical Sciences and Pharmacy, University of Newcastle, Newcastle, Australia.,Dept of Respiratory and Sleep Medicine, John Hunter Hospital, Newcastle, Australia
| | - Daniel M Johnstone
- Discipline of Physiology and Bosch Institute, University of Sydney, Sydney, Australia
| | - Elizabeth A Milward
- School of Biomedical Sciences and Pharmacy, University of Newcastle, Newcastle, Australia
| | - Philip M Hansbro
- Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute and School of Biomedical Sciences and Pharmacy, University of Newcastle, Newcastle, Australia.,Centre for Inflammation, Centenary Institute and University of Technology Sydney, Sydney, Australia.,These authors contributed equally
| | - Jay C Horvat
- Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute and School of Biomedical Sciences and Pharmacy, University of Newcastle, Newcastle, Australia.,These authors contributed equally
| |
Collapse
|
17
|
Winn NC, Volk KM, Hasty AH. Regulation of tissue iron homeostasis: the macrophage "ferrostat". JCI Insight 2020; 5:132964. [PMID: 31996481 DOI: 10.1172/jci.insight.132964] [Citation(s) in RCA: 110] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Iron is an essential element for multiple fundamental biological processes required for life; yet iron overload can be cytotoxic. Consequently, iron concentrations at the cellular and tissue level must be exquisitely governed by mechanisms that complement and fine-tune systemic control. It is well appreciated that macrophages are vital for systemic iron homeostasis, supplying or sequestering iron as needed for erythropoiesis or bacteriostasis, respectively. Indeed, recycling of iron through erythrophagocytosis by splenic macrophages is a major contributor to systemic iron homeostasis. However, accumulating evidence suggests that tissue-resident macrophages regulate local iron availability and modulate the tissue microenvironment, contributing to cellular and tissue function. Here, we summarize the significance of tissue-specific regulation of iron availability and highlight how resident macrophages are critical for this process. This tissue-dependent regulation has broad implications for understanding both resident macrophage function and tissue iron homeostasis in health and disease.
Collapse
Affiliation(s)
- Nathan C Winn
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| | - Katrina M Volk
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| | - Alyssa H Hasty
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, Tennessee, USA.,VA Tennessee Valley Healthcare System, Nashville, Tennessee, USA
| |
Collapse
|
18
|
Daher R, Lefebvre T, Puy H, Karim Z. Extrahepatic hepcidin production: The intriguing outcomes of recent years. World J Clin Cases 2019; 7:1926-1936. [PMID: 31423425 PMCID: PMC6695539 DOI: 10.12998/wjcc.v7.i15.1926] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/15/2019] [Revised: 06/18/2019] [Accepted: 06/27/2019] [Indexed: 02/05/2023] Open
Abstract
Hepcidin is the hyposideremic hormone regulating iron metabolism. It is a defensin-like disulfide-bonded peptide with antimicrobial activity. The main site of hepcidin production is the liver where its synthesis is modulated by iron, inflammation and erythropoietic signaling. However, hepcidin locally produced in several peripheral organs seems to be an important actor for the maintenance of iron homeostasis in these organs. This review highlights the presence of peripheral hepcidin and its potential functions. Understanding the role of extrahepatic hepcidin could be of great physiological and therapeutic importance for several specific pathologies.
Collapse
Affiliation(s)
- Raêd Daher
- Université Paris Diderot, Bichat site, Paris 75018, France
- Inflammation Research Center (CRI), INSERM U1149/ERL CNRS 8252, Paris 75018, France
- Laboratory of Excellence, GR-Ex, Paris 75018, France
| | - Thibaud Lefebvre
- Université Paris Diderot, Bichat site, Paris 75018, France
- Inflammation Research Center (CRI), INSERM U1149/ERL CNRS 8252, Paris 75018, France
- Laboratory of Excellence, GR-Ex, Paris 75018, France
| | - Hervé Puy
- Université Paris Diderot, Bichat site, Paris 75018, France
- Inflammation Research Center (CRI), INSERM U1149/ERL CNRS 8252, Paris 75018, France
- Laboratory of Excellence, GR-Ex, Paris 75018, France
| | - Zoubida Karim
- Université Paris Diderot, Bichat site, Paris 75018, France
- Inflammation Research Center (CRI), INSERM U1149/ERL CNRS 8252, Paris 75018, France
- Laboratory of Excellence, GR-Ex, Paris 75018, France
| |
Collapse
|
19
|
Kolloli A, Singh P, Rodriguez GM, Subbian S. Effect of Iron Supplementation on the Outcome of Non-Progressive Pulmonary Mycobacterium tuberculosis Infection. J Clin Med 2019; 8:jcm8081155. [PMID: 31382404 PMCID: PMC6722820 DOI: 10.3390/jcm8081155] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2019] [Revised: 07/30/2019] [Accepted: 07/31/2019] [Indexed: 12/13/2022] Open
Abstract
The human response to Mycobacterium tuberculosis (Mtb) infection is affected by the availability of iron (Fe), which is necessary for proper immune cell function and is essential for the growth and virulence of bacteria. Increase in host Fe levels promotes Mtb growth and tuberculosis (TB) pathogenesis, while Fe-supplementation to latently infected, asymptomatic individuals is a significant risk factor for disease reactivation. However, the effect of Fe-supplementation on the host immunity during latent Mtb infection remains unclear, due partly to the paucity in availability of animal models that recapitulate key pathophysiological features seen in humans. We have demonstrated that rabbits can develop non-progressive latency similar to infected humans. In this study, using this model we have evaluated the effect of Fe-supplementation on the bacterial growth, disease pathology, and immune response. Systemic and lung Fe parameters, gene expression profile, lung bacterial burden, and disease pathology were determined in the Mtb-infected/Fe- or placebo-supplemented rabbits. Results show that Fe-supplementation to Mtb-infected rabbits did not significantly change the hematocrit and Hb levels, although it elevated total Fe in the lungs. Expression of selected host iron- and immune-response genes in the blood and lungs was perturbed in Mtb-infected/Fe-supplemented rabbits. Iron-supplementation during acute or chronic stages of Mtb infection did not significantly affect the bacterial burden or disease pathology in the lungs. Data presented in this study is of significant relevance for current public health policies on Fe-supplementation therapy given to anemic patients with latent Mtb infection.
Collapse
Affiliation(s)
- Afsal Kolloli
- The Public Health Research Institute Center of New Jersey Medical School, Rutgers University, Newark, NJ 07103, USA
| | - Pooja Singh
- The Public Health Research Institute Center of New Jersey Medical School, Rutgers University, Newark, NJ 07103, USA
| | - G Marcela Rodriguez
- The Public Health Research Institute Center of New Jersey Medical School, Rutgers University, Newark, NJ 07103, USA
| | - Selvakumar Subbian
- The Public Health Research Institute Center of New Jersey Medical School, Rutgers University, Newark, NJ 07103, USA.
| |
Collapse
|
20
|
Iron in Lung Pathology. Pharmaceuticals (Basel) 2019; 12:ph12010030. [PMID: 30781366 PMCID: PMC6469192 DOI: 10.3390/ph12010030] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2018] [Revised: 01/30/2019] [Accepted: 02/10/2019] [Indexed: 12/21/2022] Open
Abstract
The lung presents a unique challenge for iron homeostasis. The entire airway is in direct contact with the environment and its iron particulate matter and iron-utilizing microbes. However, the homeostatic and adaptive mechanisms of pulmonary iron regulation are poorly understood. This review provides an overview of systemic and local lung iron regulation, as well as the roles of iron in the development of lung infections, airway disease, and lung injury. These mechanisms provide an important foundation for the ongoing development of therapeutic applications.
Collapse
|
21
|
Iron Homeostasis in the Lungs-A Balance between Health and Disease. Pharmaceuticals (Basel) 2019; 12:ph12010005. [PMID: 30609678 PMCID: PMC6469191 DOI: 10.3390/ph12010005] [Citation(s) in RCA: 57] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2018] [Revised: 12/23/2018] [Accepted: 12/25/2018] [Indexed: 12/15/2022] Open
Abstract
A strong mechanistic link between the regulation of iron homeostasis and oxygen sensing is evident in the lung, where both systems must be properly controlled to maintain lung function. Imbalances in pulmonary iron homeostasis are frequently associated with respiratory diseases, such as chronic obstructive pulmonary disease and with lung cancer. However, the underlying mechanisms causing alterations in iron levels and the involvement of iron in the development of lung disorders are incompletely understood. Here, we review current knowledge about the regulation of pulmonary iron homeostasis, its functional importance, and the link between dysregulated iron levels and lung diseases. Gaining greater knowledge on how iron contributes to the pathogenesis of these diseases holds promise for future iron-related therapeutic strategies.
Collapse
|
22
|
Zhang H, Zhabyeyev P, Wang S, Oudit GY. Role of iron metabolism in heart failure: From iron deficiency to iron overload. Biochim Biophys Acta Mol Basis Dis 2018; 1865:1925-1937. [PMID: 31109456 DOI: 10.1016/j.bbadis.2018.08.030] [Citation(s) in RCA: 95] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2018] [Revised: 07/25/2018] [Accepted: 08/22/2018] [Indexed: 12/11/2022]
Abstract
Iron metabolism is a balancing act, and biological systems have evolved exquisite regulatory mechanisms to maintain iron homeostasis. Iron metabolism disorders are widespread health problems on a global scale and range from iron deficiency to iron-overload. Both types of iron disorders are linked to heart failure. Iron play a fundamental role in mitochondrial function and various enzyme functions and iron deficiency has a particular negative impact on mitochondria function. Given the high-energy demand of the heart, iron deficiency has a particularly negative impact on heart function and exacerbates heart failure. Iron-overload can result from excessive gut absorption of iron or frequent use of blood transfusions and is typically seen in patients with congenital anemias, sickle cell anemia and beta-thalassemia major, or in patients with primary hemochromatosis. This review provides an overview of normal iron metabolism, mechanisms underlying development of iron disorders in relation to heart failure, including iron-overload cardiomyopathy, and clinical perspective on the treatment options for iron metabolism disorders.
Collapse
Affiliation(s)
- Hao Zhang
- Division of Cardiology, Department of Medicine, Canada; Mazankowski Alberta Heart Institute, Canada
| | - Pavel Zhabyeyev
- Division of Cardiology, Department of Medicine, Canada; Mazankowski Alberta Heart Institute, Canada
| | - Shaohua Wang
- Mazankowski Alberta Heart Institute, Canada; Division of Cardiac Surgery, Department of Surgery, University of Alberta, Edmonton, Canada
| | - Gavin Y Oudit
- Division of Cardiology, Department of Medicine, Canada; Mazankowski Alberta Heart Institute, Canada.
| |
Collapse
|
23
|
Liu YS, Huang H, Zhou SM, Tian HJ, Li P. Excessive Iron Availability Caused by Disorders of Interleukin-10 and Interleukin-22 Contributes to High Altitude Polycythemia. Front Physiol 2018; 9:548. [PMID: 29872401 PMCID: PMC5972294 DOI: 10.3389/fphys.2018.00548] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2018] [Accepted: 04/30/2018] [Indexed: 12/12/2022] Open
Abstract
Background: Because the pathogenesis of high altitude polycythemia (HAPC) is unclear, the aim of the present study was to explore whether abnormal iron metabolism is involved in the pathogenesis of HAPC and the possible cause. Methods: We examined the serum levels of iron, total iron binding capacity, soluble transferrin receptor (sTfR), ferritin, and hepcidin as well as erythropoietin (EPO) and inflammation-related cytokines in 20 healthy volunteers at sea level, 36 healthy high-altitude migrants, and 33 patients with HAPC. Mice that were exposed to a simulated hypoxic environment at an altitude of 5,000 m for 4 weeks received exogenous iron or intervention on cytokines, and the iron-related and hematological indices of peripheral blood and bone marrow were detected. The in vitro effects of some cytokines on hematopoietic cells were also observed. Results: Iron mobilization and utilization were enhanced in people who had lived at high altitudes for a long time. Notably, both the iron storage in ferritin and the available iron in the blood were elevated in patients with HAPC compared with the healthy high-altitude migrants. The correlation analysis indicated that the decreased hepcidin may have contributed to enhanced iron availability in HAPC, and decreased interleukin (IL)-10 and IL-22 were significantly associated with decreased hepcidin. The results of the animal experiments confirmed that a certain degree of iron redundancy may promote bone marrow erythropoiesis and peripheral red blood cell production in hypoxic mice and that decreased IL-10 and IL-22 stimulated iron mobilization during hypoxia by affecting hepcidin expression. Conclusion: These data demonstrated, for the first time, that an excess of obtainable iron caused by disordered IL-10 and IL-22 was involved in the pathogenesis of some HAPC patients. The potential benefits of iron removal and immunoregulation for the prevention and treatment of HAPC deserve further research.
Collapse
Affiliation(s)
- Yun-Sheng Liu
- Department of High Altitude Hygiene, College of High Altitude Military Medicine, Third Military Medical University, Chongqing, China.,Department of Medical Geography, College of High Altitude Military Medicine, Third Military Medical University, Chongqing, China.,Key Laboratory of High Altitude Environmental Medicine, Ministry of Education, Chongqing, China.,Key Laboratory of High Altitude Physiology and High Altitude Disease, Chinese People's Liberation Army, Chongqing, China
| | - He Huang
- Department of High Altitude Hygiene, College of High Altitude Military Medicine, Third Military Medical University, Chongqing, China.,Key Laboratory of High Altitude Environmental Medicine, Ministry of Education, Chongqing, China.,Key Laboratory of High Altitude Physiology and High Altitude Disease, Chinese People's Liberation Army, Chongqing, China
| | - Si-Min Zhou
- Department of High Altitude Hygiene, College of High Altitude Military Medicine, Third Military Medical University, Chongqing, China.,Key Laboratory of High Altitude Environmental Medicine, Ministry of Education, Chongqing, China.,Key Laboratory of High Altitude Physiology and High Altitude Disease, Chinese People's Liberation Army, Chongqing, China
| | - Huai-Jun Tian
- Department of High Altitude Hygiene, College of High Altitude Military Medicine, Third Military Medical University, Chongqing, China.,Key Laboratory of High Altitude Environmental Medicine, Ministry of Education, Chongqing, China.,Key Laboratory of High Altitude Physiology and High Altitude Disease, Chinese People's Liberation Army, Chongqing, China
| | - Peng Li
- Department of High Altitude Hygiene, College of High Altitude Military Medicine, Third Military Medical University, Chongqing, China.,Key Laboratory of High Altitude Environmental Medicine, Ministry of Education, Chongqing, China.,Key Laboratory of High Altitude Physiology and High Altitude Disease, Chinese People's Liberation Army, Chongqing, China
| |
Collapse
|
24
|
Harrington-Kandt R, Stylianou E, Eddowes LA, Lim PJ, Stockdale L, Pinpathomrat N, Bull N, Pasricha J, Ulaszewska M, Beglov Y, Vaulont S, Drakesmith H, McShane H. Hepcidin deficiency and iron deficiency do not alter tuberculosis susceptibility in a murine M.tb infection model. PLoS One 2018; 13:e0191038. [PMID: 29324800 PMCID: PMC5764373 DOI: 10.1371/journal.pone.0191038] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2017] [Accepted: 12/27/2017] [Indexed: 12/20/2022] Open
Abstract
Tuberculosis (TB), caused by the macrophage-tropic pathogen Mycobacterium tuberculosis (M.tb) is a highly prevalent infectious disease. Since an immune correlate of protection or effective vaccine have yet to be found, continued research into host-pathogen interactions is important. Previous literature reports links between host iron status and disease outcome for many infections, including TB. For some extracellular bacteria, the iron regulatory hormone hepcidin is essential for protection against infection. Here, we investigated hepcidin (encoded by Hamp1) in the context of murine M.tb infection. Female C57BL/6 mice were infected with M.tb Erdman via aerosol. Hepatic expression of iron-responsive genes was measured by qRT-PCR and bacterial burden determined in organ homogenates. We found that hepatic Hamp1 mRNA levels decreased post-infection, and correlated with a marker of BMP/SMAD signalling pathways. Next, we tested the effect of Hamp1 deletion, and low iron diets, on M.tb infection. Hamp1 knockout mice did not have a significantly altered M.tb mycobacterial load in either the lungs or spleen. Up to 10 weeks of dietary iron restriction did not robustly affect disease outcome despite causing iron deficiency anaemia. Taken together, our data indicate that unlike with many other infections, hepcidin is decreased following M.tb infection, and show that hepcidin ablation does not influence M.tb growth in vivo. Furthermore, because even severe iron deficiency did not affect M.tb mycobacterial load, we suggest that the mechanisms M.tb uses to scavenge iron from the host must be extremely efficient, and may therefore represent potential targets for drugs and vaccines.
Collapse
Affiliation(s)
| | - Elena Stylianou
- Jenner Institute, University of Oxford, Oxford, United Kingdom
| | - Lucy A. Eddowes
- MRC Human Immunology Unit, Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, United Kingdom
| | - Pei Jin Lim
- MRC Human Immunology Unit, Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, United Kingdom
| | - Lisa Stockdale
- Department of Immunology and Infection, London School of Hygiene & Tropical Medicine, London, United Kingdom
| | | | - Naomi Bull
- Jenner Institute, University of Oxford, Oxford, United Kingdom
| | - Janet Pasricha
- Jenner Institute, University of Oxford, Oxford, United Kingdom
| | | | - Yulia Beglov
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, United Kingdom
| | - Sophie Vaulont
- Institut Cochin, INSERM 567, CNRS 8104, Université Paris 5, Paris, France
| | - Hal Drakesmith
- MRC Human Immunology Unit, Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, United Kingdom
- * E-mail: (HD); (HMcS)
| | - Helen McShane
- Jenner Institute, University of Oxford, Oxford, United Kingdom
- * E-mail: (HD); (HMcS)
| |
Collapse
|