1
|
Kouroumalis E, Tsomidis I, Voumvouraki A. HFE-Related Hemochromatosis May Be a Primary Kupffer Cell Disease. Biomedicines 2025; 13:683. [PMID: 40149659 PMCID: PMC11940282 DOI: 10.3390/biomedicines13030683] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2025] [Revised: 02/28/2025] [Accepted: 03/08/2025] [Indexed: 03/29/2025] Open
Abstract
Iron overload can lead to increased deposition of iron and cause organ damage in the liver, the pancreas, the heart and the synovium. Iron overload disorders are due to either genetic or acquired abnormalities such as excess transfusions or chronic liver diseases. The most common genetic disease of iron deposition is classic hemochromatosis (HH) type 1, which is caused by mutations of HFE. Other rare forms of HH include type 2A with mutations at the gene hemojuvelin or type 2B with mutations in HAMP that encodes hepcidin. HH type 3, is caused by mutations of the gene that encodes transferrin receptor 2. Mutations of SLC40A1 which encodes ferroportin cause either HH type 4A or HH type 4B. In the present review, an overview of iron metabolism including absorption by enterocytes and regulation of iron by macrophages, liver sinusoidal endothelial cells (LSECs) and hepatocyte production of hepcidin is presented. Hereditary Hemochromatosis and the current pathogenetic model are analyzed. Finally, a new hypothesis based on published data was suggested. The Kupffer cell is the primary defect in HFE hemochromatosis (and possibly in types 2 and 3), while the hepcidin-relative deficiency, which is the common underlying abnormality in the three types of HH, is a secondary consequence.
Collapse
Affiliation(s)
- Elias Kouroumalis
- Department of Gastroenterology, PAGNI University Hospital, University of Crete Medical School, 71500 Heraklion, Greece
- Laboratory of Gastroenterology and Hepatology, University of Crete Medical School, 71500 Heraklion, Greece;
| | - Ioannis Tsomidis
- Laboratory of Gastroenterology and Hepatology, University of Crete Medical School, 71500 Heraklion, Greece;
| | - Argyro Voumvouraki
- 1st Department of Internal Medicine, AHEPA University Hospital, 54621 Thessaloniki, Greece;
| |
Collapse
|
2
|
Wang Z, Peng J. Impact of serum iron levels on in-hospital mortality and clinical outcomes in patients with ST segment elevation myocardial infarction undergoing emergency percutaneous coronary intervention: a retrospective analysis. Coron Artery Dis 2024; 35:539-546. [PMID: 38809141 PMCID: PMC11426973 DOI: 10.1097/mca.0000000000001393] [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: 09/27/2023] [Accepted: 11/26/2023] [Indexed: 05/30/2024]
Abstract
BACKGROUND Despite advances in percutaneous coronary intervention (PCI) for ST segment elevation myocardial infarction (STEMI), in-hospital mortality remains a concern, highlighting the need for the identification of additional risk factors such as serum iron levels. OBJECTIVE This study aims to assess the relationship between serum iron levels and in-hospital mortality among patients with STEMI undergoing emergency PCI. METHODS A total of 685 patients diagnosed with STEMI, treated with emergency PCI between January 2020 and June 2023, were included in this retrospective observational study. Participants were categorized based on serum iron levels into a low serum iron group (Fe <7.8 μmol/L) and a control group (Fe ≥7.8 μmol/L). Clinical and biochemical variables were compared between the groups. Univariate and multivariate logistic regression analyses were performed to identify independent risk factors for in-hospital mortality. RESULTS The low serum iron group demonstrated significantly higher in-hospital mortality rates (9.3 vs. 1.0%, P < 0.05) compared with the control group. Multivariate logistic regression revealed that a left ventricular ejection fraction less than 40% upon admission [odds ratio (OR), 8.01; 95% confidence interval (CI), 1.230-52.173; P = 0.029], the occurrence of no-reflow during PCI (OR, 7.13; 95% CI, 1.311-38.784; P = 0.023), and serum iron levels below 7.8 μmol/L (OR, 11.32; 95% CI, 2.345-54.640; P = 0.003) were independent risk factors for in-hospital mortality. CONCLUSION Low serum iron levels are associated with increased in-hospital mortality in patients with STEMI undergoing emergency PCI. Serum iron levels may serve as an independent prognostic marker and could inform risk stratification and therapeutic targeting in this patient population.
Collapse
Affiliation(s)
- Zuoyan Wang
- Department of Cardiology, Beijing Shijitan Hospital, Capital Medical University, Beijing, China
| | - Jianjun Peng
- Department of Cardiology, Beijing Shijitan Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
3
|
Gosmanov AR, Gemoets DE, Schumacher KA. Increased risk of erythrocytosis in men with type 2 diabetes treated with combined sodium-glucose cotransporter-2 inhibitor and testosterone replacement therapy. J Endocrinol Invest 2024; 47:2615-2621. [PMID: 38536657 DOI: 10.1007/s40618-024-02350-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Accepted: 02/22/2024] [Indexed: 09/13/2024]
Abstract
PURPOSE In clinical trials, sodium-glucose cotransporter-2 inhibitors (SGLT-2i) and testosterone replacement therapy (TRT) were shown to stimulate red blood cell production. Little is known if combination therapy poses risk of erythrocytosis in real world clinical practice. METHODS This was a retrospective nationwide cohort study of US Veterans with type 2 diabetes (T2D) and baseline hematocrit between 38 and 50% who were prescribed SGLT-2i and/or TRT between 3/2013 and 10/2022 and had adequate adherence based on the proportion of days covered > 80%. Patients were divided into 3 groups: SGLT-2i only, TRT only, or combination therapy. Odds Ratio (OR) of new erythrocytosis defined as hematocrit level > 54% within 365 days of therapy initiation was calculated by logistic regression model adjusted for baseline hematocrit, age, BMI, obstructive sleep apnea, diuretic use, and smoking status. RESULTS Of the entire cohort of 53,971 people with T2D, total of 756 (1.4%) patients developed erythrocytosis. In unadjusted analyses, the OR of new onset erythrocytosis was higher in the combined SGLT-2i and TRT group compared with the SGLT-2i or TRT group alone (4.99, 95% CI (3.10-7.71) and 2.91, 95% CI (1.87-4.31), respectively). In the models adjusted for baseline characteristics, patients on combination therapy had significantly higher odds of erythrocytosis compared to those on SGLT-2i (OR 3.80, 95% CI (2.27-6.11)) or TRT alone (OR 2.49, 95% CI (1.51-3.59)). Testosterone delivery route (topical vs injectable) did not modify increased odds of erythrocytosis. CONCLUSIONS For the first time, we demonstrated that in large cohort of patients combined therapy with SGLT-2i and TRT is associated with increased erythrocytosis risk compared with either treatment alone. Given rising prevalence of SGLT-2i use, providers should consider periodic hematocrit assessment in persons receiving both SGLT-2i and TRT.
Collapse
Affiliation(s)
- A R Gosmanov
- Section of Endocrinology, Stratton VA Medical Center, 113 Holland Ave, Albany, 12208, USA.
- Division of Endocrinology, Department of Medicine, Albany Medical College, Albany, NY, USA.
| | - D E Gemoets
- Department of Research and Development, Stratton VA Medical Center, Albany, NY, USA
| | - K A Schumacher
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA, USA
| |
Collapse
|
4
|
Xu T, Zhang X, Zhao W, Shi J, Wan S, Zhang Y, Hao Y, Sun M, He J, Jiang L, Wang H, Gao H, Luo J, Luo Y, An P. Foxo1 is an iron-responsive transcriptional factor regulating systemic iron homeostasis. Blood 2024; 144:1314-1328. [PMID: 38848533 DOI: 10.1182/blood.2024024293] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2024] [Revised: 05/22/2024] [Accepted: 05/28/2024] [Indexed: 06/09/2024] Open
Abstract
The liver plays a crucial role in maintaining systemic iron homeostasis by secreting hepcidin, which is essential for coordinating iron levels in the body. Imbalances in iron homeostasis are associated with various clinical disorders related to iron deficiency or iron overload. Despite the clinical significance, the mechanisms underlying how hepatocytes sense extracellular iron levels to regulate hepcidin synthesis and iron storage are not fully understood. In this study, we identified Foxo1, a well-known regulator of macronutrient metabolism, which translocates to the nucleus of hepatocytes in response to high-iron feeding, holo-transferrin, and bone morphogenetic protein 6 (BMP6) treatment. Furthermore, Foxo1 plays a crucial role in mediating hepcidin induction in response to both iron and BMP signals by directly interacting with evolutionally conserved Foxo binding sites within the hepcidin promoter region. These binding sites were found to colocalize with Smad-binding sites. To investigate the physiological relevance of Foxo1 in iron metabolism, we generated mice with hepatocyte-specific deletion of Foxo1. These mice exhibited reduced hepatic hepcidin expression and serum hepcidin levels, accompanied by elevated serum iron and liver nonheme iron concentrations. Moreover, high-iron diet further exacerbated these abnormalities in iron metabolism in mice lacking hepatic Foxo1. Conversely, hepatocyte-specific Foxo1 overexpression increased hepatic hepcidin expression and serum hepcidin levels, thereby ameliorating iron overload in a murine model of hereditary hemochromatosis (Hfe-/- mice). In summary, our study identifies Foxo1 as a critical regulator of hepcidin and systemic iron homeostasis. Targeting Foxo1 may offer therapeutic opportunities for managing conditions associated with aberrant iron metabolism.
Collapse
Affiliation(s)
- Teng Xu
- Department of Nutrition and Health, China Agricultural University, Beijing, China
| | - Xu Zhang
- Department of Nutrition and Health, China Agricultural University, Beijing, China
| | - Wenting Zhao
- Department of Nutrition and Health, China Agricultural University, Beijing, China
| | - Jiaxin Shi
- Department of Nutrition and Health, China Agricultural University, Beijing, China
| | - Sitong Wan
- Department of Nutrition and Health, China Agricultural University, Beijing, China
| | - Yan Zhang
- College of Food Science and Engineering, Gansu Agricultural University, Lanzhou, China
| | - Yanling Hao
- Department of Nutrition and Health, China Agricultural University, Beijing, China
| | - Mingyue Sun
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Jingjing He
- Department of Nutrition and Health, China Agricultural University, Beijing, China
| | - Li Jiang
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Hangzhou Normal University, Hangzhou, China
| | - Hao Wang
- School of Public Health, Zhengzhou University, Zhengzhou, China
| | - Hong Gao
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, TaiKang Center for Life and Medical Sciences, Wuhan University, Wuhan, China
| | - Junjie Luo
- Department of Nutrition and Health, China Agricultural University, Beijing, China
| | - Yongting Luo
- Department of Nutrition and Health, China Agricultural University, Beijing, China
| | - Peng An
- Department of Nutrition and Health, China Agricultural University, Beijing, China
| |
Collapse
|
5
|
Pan X, Köberle M, Ghashghaeinia M. Vitamin C-Dependent Uptake of Non-Heme Iron by Enterocytes, Its Impact on Erythropoiesis and Redox Capacity of Human Erythrocytes. Antioxidants (Basel) 2024; 13:968. [PMID: 39199214 PMCID: PMC11352176 DOI: 10.3390/antiox13080968] [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: 07/25/2024] [Revised: 08/06/2024] [Accepted: 08/07/2024] [Indexed: 09/01/2024] Open
Abstract
In the small intestine, nutrients from ingested food are absorbed and broken down by enterocytes, which constitute over 95% of the intestinal epithelium. Enterocytes demonstrate diet- and segment-dependent metabolic flexibility, enabling them to take up large amounts of glutamine and glucose to meet their energy needs and transfer these nutrients into the bloodstream. During glycolysis, ATP, lactate, and H+ ions are produced within the enterocytes. Based on extensive but incomplete glutamine oxidation large amounts of alanine or lactate are produced. Lactate, in turn, promotes hypoxia-inducible factor-1α (Hif-1α) activation and Hif-1α-dependent transcription of various proton channels and exchangers, which extrude cytoplasmic H+-ions into the intestinal lumen. In parallel, the vitamin C-dependent and duodenal cytochrome b-mediated conversion of ferric iron into ferrous iron progresses. Finally, the generated electrochemical gradient is utilized by the divalent metal transporter 1 for H+-coupled uptake of non-heme Fe2+-ions. Iron efflux from enterocytes, subsequent binding to the plasma protein transferrin, and systemic distribution supply a wide range of cells with iron, including erythroid precursors essential for erythropoiesis. In this review, we discuss the impact of vitamin C on the redox capacity of human erythrocytes and connect enterocyte function with iron metabolism, highlighting its effects on erythropoiesis.
Collapse
Affiliation(s)
- Xia Pan
- Physiological Institute, Department of Vegetative and Clinical Physiology, Eberhard Karls University of Tübingen, 72074 Tübingen, Germany
| | - Martin Köberle
- Department of Dermatology and Allergology, School of Medicine and Health, Technical University of Munich, Biedersteinerstr. 29, 80802 München, Germany
| | - Mehrdad Ghashghaeinia
- Physiological Institute, Department of Vegetative and Clinical Physiology, Eberhard Karls University of Tübingen, 72074 Tübingen, Germany
| |
Collapse
|
6
|
Sardo U, Perrier P, Cormier K, Sotin M, Personnaz J, Medjbeur T, Desquesnes A, Cannizzo L, Ruiz-Martinez M, Thevenin J, Billoré B, Jung G, Abboud E, Peyssonnaux C, Nemeth E, Ginzburg YZ, Ganz T, Kautz L. The hepatokine FGL1 regulates hepcidin and iron metabolism during anemia in mice by antagonizing BMP signaling. Blood 2024; 143:1282-1292. [PMID: 38232308 PMCID: PMC11103088 DOI: 10.1182/blood.2023022724] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Revised: 11/20/2023] [Accepted: 12/13/2023] [Indexed: 01/19/2024] Open
Abstract
ABSTRACT As a functional component of erythrocyte hemoglobin, iron is essential for oxygen delivery to all tissues in the body. The liver-derived peptide hepcidin is the master regulator of iron homeostasis. During anemia, the erythroid hormone erythroferrone regulates hepcidin synthesis to ensure the adequate supply of iron to the bone marrow for red blood cell production. However, mounting evidence suggested that another factor may exert a similar function. We identified the hepatokine fibrinogen-like 1 (FGL1) as a previously undescribed suppressor of hepcidin that is induced in the liver in response to hypoxia during the recovery from anemia, and in thalassemic mice. We demonstrated that FGL1 is a potent suppressor of hepcidin in vitro and in vivo. Deletion of Fgl1 in mice results in higher hepcidin levels at baseline and after bleeding. FGL1 exerts its activity by directly binding to bone morphogenetic protein 6 (BMP6), thereby inhibiting the canonical BMP-SMAD signaling cascade that controls hepcidin transcription.
Collapse
Affiliation(s)
- Ugo Sardo
- Institut de Recherche en Santé Digestive, Université de Toulouse, INSERM, INRAE, ENVT, Université Toulouse III Paul Sabatier, Toulouse, France
| | - Prunelle Perrier
- Institut de Recherche en Santé Digestive, Université de Toulouse, INSERM, INRAE, ENVT, Université Toulouse III Paul Sabatier, Toulouse, France
| | - Kevin Cormier
- Institut de Recherche en Santé Digestive, Université de Toulouse, INSERM, INRAE, ENVT, Université Toulouse III Paul Sabatier, Toulouse, France
| | - Manon Sotin
- Institut de Recherche en Santé Digestive, Université de Toulouse, INSERM, INRAE, ENVT, Université Toulouse III Paul Sabatier, Toulouse, France
| | - Jean Personnaz
- Institut de Recherche en Santé Digestive, Université de Toulouse, INSERM, INRAE, ENVT, Université Toulouse III Paul Sabatier, Toulouse, France
| | - Thanina Medjbeur
- Institut de Recherche en Santé Digestive, Université de Toulouse, INSERM, INRAE, ENVT, Université Toulouse III Paul Sabatier, Toulouse, France
| | - Aurore Desquesnes
- Institut de Recherche en Santé Digestive, Université de Toulouse, INSERM, INRAE, ENVT, Université Toulouse III Paul Sabatier, Toulouse, France
| | - Lisa Cannizzo
- Institut de Recherche en Santé Digestive, Université de Toulouse, INSERM, INRAE, ENVT, Université Toulouse III Paul Sabatier, Toulouse, France
| | | | - Julie Thevenin
- Institut de Recherche en Santé Digestive, Université de Toulouse, INSERM, INRAE, ENVT, Université Toulouse III Paul Sabatier, Toulouse, France
| | - Benjamin Billoré
- Institut de Recherche en Santé Digestive, Université de Toulouse, INSERM, INRAE, ENVT, Université Toulouse III Paul Sabatier, Toulouse, France
| | - Grace Jung
- Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA
| | - Elise Abboud
- Institut Cochin, INSERM, Centre National de la Recherche Scientifique, Université de Paris, Paris, France
- Laboratory of Excellence GR-Ex, Paris, France
| | - Carole Peyssonnaux
- Institut Cochin, INSERM, Centre National de la Recherche Scientifique, Université de Paris, Paris, France
- Laboratory of Excellence GR-Ex, Paris, France
| | - Elizabeta Nemeth
- Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA
| | | | - Tomas Ganz
- Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA
- Department of Pathology, David Geffen School of Medicine at UCLA, Los Angeles, CA
| | - Léon Kautz
- Institut de Recherche en Santé Digestive, Université de Toulouse, INSERM, INRAE, ENVT, Université Toulouse III Paul Sabatier, Toulouse, France
| |
Collapse
|
7
|
Sarate N, Sonawane R, Pai V, Karatela S, Mulkalwar A. Iron Deficiency: A Silent Threat in Patients With Heart Failure With Reduced Ejection Fraction. Cureus 2024; 16:e53542. [PMID: 38445122 PMCID: PMC10912968 DOI: 10.7759/cureus.53542] [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: 02/04/2024] [Indexed: 03/07/2024] Open
Abstract
Background Iron deficiency is a prevalent and clinically significant comorbidity in patients with heart failure with reduced ejection fraction (HFrEF). Despite its high prevalence, its impact on clinical outcomes, mortality, and various physiological parameters remains a subject of ongoing investigation. The findings of this study are anticipated to contribute valuable insights into the management and prognosis of patients with HFrEF, potentially informing future interventions and improving patient outcomes. This study aimed to assess the clinical profile of iron deficiency and its implications on morbidity and mortality in patients with HFrEF. Methodology A prospective cohort study was conducted at King Edward Memorial Hospital, India, involving 371 patients with HFrEF. Participants underwent comprehensive clinical and laboratory assessments, evaluating iron deficiency with signs, symptoms, comorbidities, dyspnea, elevated jugular venous pressure (JVP), past medical history, and various hematological and biochemical parameters. Results Overall, 50% of HFrEF participants were iron deficient (n = 185), of whom 80% (n = 148) had anemia against 43% (n = 81) anemics in iron repletes (n = 186). Of the 185 iron-deficient patients, 44 (11.86%) had absolute iron deficiency and 141 (38%) had functional iron deficiency. Iron deficiency significantly correlated with increased mortality in HFrEF patients (χ2 (1, N = 371) = 3.88, p = 0.048). A large positive correlation was observed between absolute iron deficiency and dyspnea severity (r2 = 0.949, p = 0.026). Statistically significant differences were found in hemoglobin (anemia), serum iron, serum ferritin, total iron-binding capacity, and transferrin saturation between iron-deficient and iron-replete patients (p < 0.05). However, no statistically significant difference in left ventricular ejection fraction between iron-deficient and replete patients was noted. Conclusions Iron deficiency emerges as more than a mere comorbidity in heart failure, becoming a prognostic factor with multifaceted outcomes. Its impact extends beyond cardiovascular consequences, encompassing adverse manifestations such as anemia, ascites, edema, dyspnea, elevated JVP, and a heightened risk of mortality. This intricate interplay positions iron deficiency as a critical determinant, significantly influencing the clinical trajectory and outcomes for patients with HFrEF.
Collapse
Affiliation(s)
- Nitin Sarate
- Department of Medicine, Seth Gordhandas Sunderdas Medical College and King Edward Memorial Hospital, Mumbai, IND
| | - Rahul Sonawane
- Department of Medicine, Seth Gordhandas Sunderdas Medical College and King Edward Memorial Hospital, Mumbai, IND
| | - Vinayak Pai
- Department of Medicine, Seth Gordhandas Sunderdas Medical College and King Edward Memorial Hospital, Mumbai, IND
| | - Shifa Karatela
- Department of Medicine, Medical College Baroda and Sir Sayajirao General Hospital, Vadodara, IND
| | - Alhad Mulkalwar
- Department of Pharmacology, Dr. D. Y. Patil Medical College, Hospital & Research Centre, Pune, IND
| |
Collapse
|
8
|
Zheng R, Lin C, Mao Y, Jin F. miR-761-hepcidin/Gpx4 pathway contribute to unexplained liver dysfunction in polycystic ovary syndrome by regulating liver iron overload and ferroptosis. Gynecol Endocrinol 2023; 39:2166483. [PMID: 36657482 DOI: 10.1080/09513590.2023.2166483] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
Aims: To investigate the underling mechanisms of liver dysfunction in patients with polycystic ovary syndrome (PCOS).Materials and methods: PCOS patients were enrolled according to the Amsterdam criteria while PCOS animal model was established by dihydrotestosterone (DHEA) sustained release tablet implantation on its neck. Further liver damage and iron overload were detected by HE and Prussian blue staining. The liver related enzymes, mRNA and protein levels of hepcidin and GPX4 were tested by ELISA, qRT-PCR and Western blot. RNA interference and miR-761 transfection were routinely performed while the regulation of miR-761 on hepcidin and GPX4 was confirmed by luciferase reporter gene analysis.Results: We found that a part of PCOS patients and animal model had unexplained liver damage, which is independent of nonalcoholic fatty liver disease (NAFLD) and accompanied by increased ferrum (Fe) deposition. Besides, the expression of hepcidin and GPX4 that is important effector proteins for ferroptosis was down regulated in liver, showing the importance of iron metabolism in this unexplained liver damage. Based on the miR-761-hepcidin/GPX4 axis, we systematically studied the effects of miR-761 on ferroptosis and Fe deposition, which further influence the phenotype and liver function of PCOS model. From both in vivo and in vitro levels, changes in PCOS disease phenotype and ferroptosis were observed through hierarchical antagonism or overexpression of miR-761, hepcidin and GPX4.Conclusions: our results provide a novel explanation for unexplained liver damage in PCOS and a potential therapeutic target.
Collapse
Affiliation(s)
- Ruoheng Zheng
- School of Medicine, Zhejiang University, Hangzhou, Zhejiang, P. R. China
- School of Clinical Medicine, Hangzhou Medical College, Hangzhou, Zhejiang, P. R. China
| | - Chuanping Lin
- Department of Reproductive Endocrinology, Key Laboratory of Reproductive Genetics of National Ministry of Education, Women's Reproductive Health Laboratory of Zhejiang Province, Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, P. R. China
| | - Yuchan Mao
- Department of Reproductive Endocrinology, Key Laboratory of Reproductive Genetics of National Ministry of Education, Women's Reproductive Health Laboratory of Zhejiang Province, Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, P. R. China
| | - Fan Jin
- Department of Reproductive Endocrinology, Key Laboratory of Reproductive Genetics of National Ministry of Education, Women's Reproductive Health Laboratory of Zhejiang Province, Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, P. R. China
| |
Collapse
|
9
|
Xiao X, Xu Y, Moschetta GA, Yu Y, Fisher AL, Alfaro-Magallanes VM, McMillen S, Phillips S, Wang CY, Christian J, Babitt JL. BMP5 contributes to hepcidin regulation and systemic iron homeostasis in mice. Blood 2023; 142:1312-1322. [PMID: 37478395 PMCID: PMC10613724 DOI: 10.1182/blood.2022019195] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2022] [Revised: 06/14/2023] [Accepted: 06/22/2023] [Indexed: 07/23/2023] Open
Abstract
Hepcidin is the master regulator of systemic iron homeostasis. The bone morphogenetic protein (BMP) signaling pathway is a critical regulator of hepcidin expression in response to iron and erythropoietic drive. Although endothelial-derived BMP6 and BMP2 ligands have key functional roles as endogenous hepcidin regulators, both iron and erythropoietic drives still regulate hepcidin in mice lacking either or both ligands. Here, we used mice with an inactivating Bmp5 mutation (Bmp5se), either alone or together with a global or endothelial Bmp6 knockout, to investigate the functional role of BMP5 in hepcidin and systemic iron homeostasis regulation. We showed that Bmp5se-mutant mice exhibit hepcidin deficiency at age 10 days, blunted hepcidin induction in response to oral iron gavage, and mild liver iron loading when fed on a low- or high-iron diet. Loss of 1 or 2 functional Bmp5 alleles also leads to increased iron loading in Bmp6-heterozygous mice and more profound hemochromatosis in global or endothelial Bmp6-knockout mice. Moreover, double Bmp5- and Bmp6-mutant mice fail to induce hepcidin in response to long-term dietary iron loading. Finally, erythroferrone binds directly to BMP5 and inhibits BMP5 induction of hepcidin in vitro. Although erythropoietin suppresses hepcidin in Bmp5se-mutant mice, it fails to suppress hepcidin in double Bmp5- and Bmp6-mutant males. Together, these data demonstrate that BMP5 plays a functional role in hepcidin and iron homeostasis regulation, particularly under conditions in which BMP6 is limited.
Collapse
Affiliation(s)
- Xia Xiao
- Nephrology Division and Endocrine Unit, Massachusetts General Hospital, Harvard Medical School, Boston, MA
| | - Yang Xu
- Nephrology Division and Endocrine Unit, Massachusetts General Hospital, Harvard Medical School, Boston, MA
| | - Gillian A. Moschetta
- Nephrology Division and Endocrine Unit, Massachusetts General Hospital, Harvard Medical School, Boston, MA
| | - Yang Yu
- Nephrology Division and Endocrine Unit, Massachusetts General Hospital, Harvard Medical School, Boston, MA
| | - Allison L. Fisher
- Nephrology Division and Endocrine Unit, Massachusetts General Hospital, Harvard Medical School, Boston, MA
| | - Víctor M. Alfaro-Magallanes
- Nephrology Division and Endocrine Unit, Massachusetts General Hospital, Harvard Medical School, Boston, MA
- LFE Research Group, Department of Health and Human Performance, Faculty of Physical Activity and Sport Sciences, Universidad Politécnica de Madrid, Madrid, Spain
| | - Shasta McMillen
- Nephrology Division and Endocrine Unit, Massachusetts General Hospital, Harvard Medical School, Boston, MA
| | - Sydney Phillips
- Nephrology Division and Endocrine Unit, Massachusetts General Hospital, Harvard Medical School, Boston, MA
| | - Chia-Yu Wang
- Nephrology Division and Endocrine Unit, Massachusetts General Hospital, Harvard Medical School, Boston, MA
| | - Jan Christian
- Division of Hematology and Hematologic Malignancies, Department of Neurobiology and Internal Medicine, University of Utah, Salt Lake City, UT
| | - Jodie L. Babitt
- Nephrology Division and Endocrine Unit, Massachusetts General Hospital, Harvard Medical School, Boston, MA
| |
Collapse
|
10
|
Rehati A, Abuduaini B, Liang Z, Chen D, He F. Identification of heat shock protein family A member 5 (HSPA5) targets involved in nonalcoholic fatty liver disease. Genes Immun 2023:10.1038/s41435-023-00205-y. [PMID: 37156995 DOI: 10.1038/s41435-023-00205-y] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2022] [Revised: 02/27/2023] [Accepted: 04/17/2023] [Indexed: 05/10/2023]
Abstract
Heat shock protein family A (Hsp70) member 5 (HSPA5) is an endoplasmic reticulum chaperone, which regulates cell metabolism, particularly lipid metabolism. While HSPA5's role in regulating cell function is well described, HSPA5 binding to RNA and its biological function in nonalcoholic fatty liver disease (NAFLD) is still lacking. In the present study, the ability of HSPA5 to modulate alternative splicing (AS) of cellular genes was assessed using Real-Time PCR on 89 NAFLD-associated genes. RNA immunoprecipitation coupled to RNA sequencing (RIP-Seq) assays were also performed to identify cellular mRNAs bound by HSPA5. We obtained the HSPA5-bound RNA profile in HeLa cells and peak calling analysis revealed that HSPA5 binds to coding genes and lncRNAs. Moreover, RIP-Seq assays demonstrated that HSPA5 immunoprecipitates specific cellular mRNAs such as EGFR, NEAT1, LRP1 and TGFß1, which are important in the pathology of NAFLD. Finally, HSPA5 binding sites may be associated with splicing sites. We used the HOMER algorithm to search for motifs enriched in coding sequence (CDs) peaks, which identified over-representation of the AGAG motif in both sets of immunoprecipitated peaks. HSPA5 regulated genes at the 5'UTR alternative splicing and introns and in an AG-rich sequence-dependent manner. We propose that the HSPA5-AGAG interaction might play an important role in regulating alternative splicing of NAFLD-related genes. This report is the first to demonstrate that HSPA5 regulated pre-RNA alternative splicing, stability, or translation and affected target protein(s) via binding to lncRNA and mRNA linked to NAFLD.
Collapse
Affiliation(s)
- Aliya Rehati
- Department of Gastroenterology, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, 830054, Xinjiang, China
| | - Buzukela Abuduaini
- Department of Intensive Care Unit, The First Affiliated Hospital of Xinjiang Medical University, 393 South Li Yu Shan Road, Urumqi, 830054, Xinjiang, China.
| | - Zhao Liang
- Department of General Surgery, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, 830054, Xinjiang, China
| | - Dong Chen
- ABLife BioBigData Institute, Wuhan, 430075, Hubei, China
| | - Fangping He
- Department of Gastroenterology, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, 830054, Xinjiang, China.
| |
Collapse
|
11
|
Li X, Duan X, Tan D, Zhang B, Xu A, Qiu N, Chen Z. Iron deficiency and iron overload in men and woman of reproductive age, and pregnant women. Reprod Toxicol 2023; 118:108381. [PMID: 37023911 DOI: 10.1016/j.reprotox.2023.108381] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Revised: 03/06/2023] [Accepted: 04/02/2023] [Indexed: 04/08/2023]
Abstract
Iron is an essential micronutrient for human biology and health, but high iron levels can be dangerous. Both iron deficiency and iron overload have been linked to reproductive health. This review summarizes the effects of iron deficiency and iron overload on men of reproductive age, women of reproductive age, and pregnant women. In addition, appropriate iron levels and the need for iron and nutritional supplements at different stages of life and pregnancy are discussed. In general, men should be aware of the risk of iron overload at any stage of life; women should take appropriate iron supplements before menopause; postmenopausal women should pay attention to the risk of iron overload; and pregnant women should receive reasonable iron supplementation in middle and late pregnancy. By summarizing evidence on the relationship between iron and reproductive health, this review aims to promote the development of strategies to optimize reproductive capacity from the perspective of nutrition. However, additional detailed experimental investigations and clinical studies are needed to assess the underlying causes and mechanisms of the observed associations between iron and reproductive health.
Collapse
Affiliation(s)
- Xiuyun Li
- Maternal and Child Health Development Research Center, Key Laboratory of Birth Regulation and Control Technology of National Health Commission of China, Shandong Provincial Maternal and Child Health Care Hospital Affiliated to Qingdao University, Jinan, China
| | - Xuexia Duan
- Physical Examination Center, Shandong Provincial Maternal and Child Health Care Hospital Affiliated to Qingdao University, Jinan, China
| | - Dongmei Tan
- Traditional Chinese Medicine Department, Shandong Provincial Maternal and Child Health Care Hospital Affiliated to Qingdao University, Jinan, China
| | - Bin Zhang
- Department of Ophthalmology, Shandong Provincial Maternal and Child Health Care Hospital Affiliated to Qingdao University, Jinan, China
| | - Anran Xu
- Reproductive Medicine Center, Shandong Provincial Maternal and Child Health Care Hospital Affiliated to Qingdao University, Jinan, China
| | - Ningning Qiu
- Department of Anesthesiology, Shandong Provincial Maternal and Child Health Care Hospital Affiliated to Qingdao University, Jinan, China.
| | - Zhaowen Chen
- Obstetrics Department, Shandong Provincial Maternal and Child Health Care Hospital Affiliated to Qingdao University, Jinan, China.
| |
Collapse
|
12
|
Satué K, Fazio E, La Fauci D, Medica P. Changes of Hepcidin, Ferritin and Iron Levels in Cycling Purebred Spanish Mares. Animals (Basel) 2023; 13:ani13071229. [PMID: 37048485 PMCID: PMC10093536 DOI: 10.3390/ani13071229] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Revised: 03/21/2023] [Accepted: 03/30/2023] [Indexed: 04/05/2023] Open
Abstract
Several studies have demonstrated that in woman the sex hormones such as estrogen (E2) and progesterone (P4) influence iron (Fe) regulation, contributing to variations in Fe parameters along the menstrual cycle. These mechanisms based on the regulation of hepcidin (Hepc) which limits Fe availability during the cycle, remain poorly characterized in healthy mares. The objective of this study was to establish the relationship between Hepc, Fe, ferritin (Ferr), and the primary ovarian hormones E2 and P4 in cycling Purebred Spanish mares. Blood samples were taken from 31 Purebred Spanish mares day −5, on day 0, day +5 and day +16 of the cycle. Fe and Ferr significantly increased and Hepc decreased during pre- and ovulatory periods. The secretion peak of estradiol-17β (E2) was reached on day 0 and progesterone (P4) between days +5 and +16. Fe and Ferr were positively correlated (r = 0.57). Fe and Ferr were negatively correlated with Hepc (r = −0.72 and r = −0.02, respectively). E2 and P4 were negatively and positively correlated with Hepc (r = −0.753 and r = 0.54, respectively). In cycling Purebred Spanish mares there is a measurable relationship between steroid hormones and systemic Fe metabolism. Estrogenic dominance in the pre- and ovulatory period allows for a more effective iron status, mediated by hepcidin inhibition. However, P4 during the luteal phase substantially reduces serum Fe and iron stores, possibly related to Hepc stimulation. Future research is required to clarify the relationship between steroid hormones and iron metabolism at the molecular level in equids.
Collapse
Affiliation(s)
- Katiuska Satué
- Department of Animal Medicine and Surgery, Faculty of Veterinary Medicine, CEU-Cardenal Herrera University, 46115 Valencia, Spain
| | - Esterina Fazio
- Department of Veterinary Sciences, Veterinary Physiology Unit, Messina University, Viale Palatucci 13, 98168 Messina, Italy
| | - Deborah La Fauci
- Department of Veterinary Sciences, Veterinary Physiology Unit, Messina University, Viale Palatucci 13, 98168 Messina, Italy
| | - Pietro Medica
- Department of Veterinary Sciences, Veterinary Physiology Unit, Messina University, Viale Palatucci 13, 98168 Messina, Italy
| |
Collapse
|
13
|
Adams AR, Li X, Byanyima JI, Vesslee SA, Nguyen TD, Wang Y, Moon B, Pond T, Kranzler HR, Witschey WR, Shi Z, Wiers CE. Peripheral and Central Iron Measures in Alcohol Use Disorder and Aging: A Quantitative Susceptibility Mapping Pilot Study. Int J Mol Sci 2023; 24:4461. [PMID: 36901892 PMCID: PMC10002495 DOI: 10.3390/ijms24054461] [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: 02/05/2023] [Revised: 02/18/2023] [Accepted: 02/21/2023] [Indexed: 02/26/2023] Open
Abstract
Chronic excessive alcohol use has neurotoxic effects, which may contribute to cognitive decline and the risk of early-onset dementia. Elevated peripheral iron levels have been reported in individuals with alcohol use disorder (AUD), but its association with brain iron loading has not been explored. We evaluated whether (1) serum and brain iron loading are higher in individuals with AUD than non-dependent healthy controls and (2) serum and brain iron loading increase with age. A fasting serum iron panel was obtained and a magnetic resonance imaging scan with quantitative susceptibility mapping (QSM) was used to quantify brain iron concentrations. Although serum ferritin levels were higher in the AUD group than in controls, whole-brain iron susceptibility did not differ between groups. Voxel-wise QSM analyses revealed higher susceptibility in a cluster in the left globus pallidus in individuals with AUD than controls. Whole-brain iron increased with age and voxel-wise QSM indicated higher susceptibility with age in various brain areas including the basal ganglia. This is the first study to analyze both serum and brain iron loading in individuals with AUD. Larger studies are needed to examine the effects of alcohol use on iron loading and its associations with alcohol use severity, structural and functional brain changes, and alcohol-induced cognitive impairments.
Collapse
Affiliation(s)
- Aiden R. Adams
- Center for Studies of Addiction, Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, 3535 Market St Ste 500, Philadelphia, PA 19104, USA
| | - Xinyi Li
- Center for Studies of Addiction, Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, 3535 Market St Ste 500, Philadelphia, PA 19104, USA
| | - Juliana I. Byanyima
- Center for Studies of Addiction, Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, 3535 Market St Ste 500, Philadelphia, PA 19104, USA
| | - Sianneh A. Vesslee
- Center for Studies of Addiction, Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, 3535 Market St Ste 500, Philadelphia, PA 19104, USA
| | - Thanh D. Nguyen
- Department of Radiology, Weill Cornell Medicine, 525 E 68th St, New York, NY 10065, USA
| | - Yi Wang
- Department of Radiology, Weill Cornell Medicine, 525 E 68th St, New York, NY 10065, USA
| | - Brianna Moon
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, 3400 Civic Center Blvd, South Pavilion, Room 11-155, Philadelphia, PA 19104, USA
| | - Timothy Pond
- Center for Studies of Addiction, Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, 3535 Market St Ste 500, Philadelphia, PA 19104, USA
| | - Henry R. Kranzler
- Center for Studies of Addiction, Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, 3535 Market St Ste 500, Philadelphia, PA 19104, USA
| | - Walter R. Witschey
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, 3400 Civic Center Blvd, South Pavilion, Room 11-155, Philadelphia, PA 19104, USA
| | - Zhenhao Shi
- Center for Studies of Addiction, Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, 3535 Market St Ste 500, Philadelphia, PA 19104, USA
| | - Corinde E. Wiers
- Center for Studies of Addiction, Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, 3535 Market St Ste 500, Philadelphia, PA 19104, USA
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, 3400 Civic Center Blvd, South Pavilion, Room 11-155, Philadelphia, PA 19104, USA
| |
Collapse
|
14
|
Fisher AL, Wang CY, Xu Y, Joachim K, Xiao X, Phillips S, Moschetta GA, Alfaro-Magallanes VM, Babitt JL. Functional role of endothelial transferrin receptor 1 in iron sensing and homeostasis. Am J Hematol 2022; 97:1548-1559. [PMID: 36069607 PMCID: PMC9662186 DOI: 10.1002/ajh.26716] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Revised: 08/09/2022] [Accepted: 09/01/2022] [Indexed: 01/31/2023]
Abstract
Systemic iron homeostasis is regulated by the hepatic hormone hepcidin to balance meeting iron requirements while limiting toxicity from iron excess. Iron-mediated induction of bone morphogenetic protein (BMP) 6 is a central mechanism for regulating hepcidin production. Liver endothelial cells (LECs) are the main source of endogenous BMP6, but how they sense iron to modulate BMP6 transcription and thereby hepcidin is uncertain. Here, we investigate the role of endothelial cell transferrin receptor 1 (TFR1) in iron uptake, BMP6 regulation, and systemic iron homeostasis using primary LEC cultures and endothelial Tfrc (encoding TFR1) knockout mice. We show that intracellular iron regulates Bmp6 expression in a cell-autonomous manner, and TFR1 mediates iron uptake and Bmp6 expression by holo-transferrin in primary LEC cultures. In addition, endothelial Tfrc knockout mice exhibit altered iron homeostasis compared with littermate controls when fed a limited iron diet, as evidenced by increased liver iron and inappropriately low Bmp6 and hepcidin expression relative to liver iron. However, endothelial Tfrc knockout mice have a similar iron phenotype compared to littermate controls when fed an iron-rich standard diet. Finally, ferritin and non-transferrin bound iron (NTBI) are additional sources of iron that mediate Bmp6 induction in primary LEC cultures via TFR1-independent mechanisms. Together, our data demonstrate a minor functional role for endothelial cell TFR1 in iron uptake, BMP6 regulation, and hepatocyte hepcidin regulation under iron limiting conditions, and suggest that ferritin and/or NTBI uptake by other transporters have a dominant role when iron availability is high.
Collapse
Affiliation(s)
- Allison L Fisher
- Nephrology Division and Endocrine Unit, Massachusetts General Hospital, Harvard Medical School, Boston, MA
| | - Chia-yu Wang
- Nephrology Division and Endocrine Unit, Massachusetts General Hospital, Harvard Medical School, Boston, MA
| | - Yang Xu
- Nephrology Division and Endocrine Unit, Massachusetts General Hospital, Harvard Medical School, Boston, MA
| | - Kole Joachim
- Nephrology Division and Endocrine Unit, Massachusetts General Hospital, Harvard Medical School, Boston, MA
| | - Xia Xiao
- Nephrology Division and Endocrine Unit, Massachusetts General Hospital, Harvard Medical School, Boston, MA
| | - Sydney Phillips
- Nephrology Division and Endocrine Unit, Massachusetts General Hospital, Harvard Medical School, Boston, MA
| | - Gillian A Moschetta
- Nephrology Division and Endocrine Unit, Massachusetts General Hospital, Harvard Medical School, Boston, MA
| | - Victor M Alfaro-Magallanes
- Nephrology Division and Endocrine Unit, Massachusetts General Hospital, Harvard Medical School, Boston, MA
- LFE Research Group, Department of Health and Human Performance, Faculty of Physical Activity and Sport Sciences, Universidad Politécnica de Madrid, Madrid, Spain
| | - Jodie L Babitt
- Nephrology Division and Endocrine Unit, Massachusetts General Hospital, Harvard Medical School, Boston, MA
| |
Collapse
|
15
|
Luiz MM, Schneider IJC, Kuriki HU, Fattori A, Corrêa VP, Steptoe A, Alexandre TDS, de Oliveira C. The combined effect of anemia and dynapenia on mortality risk in older adults: 10-Year evidence from the ELSA cohort study. Arch Gerontol Geriatr 2022; 102:104739. [DOI: 10.1016/j.archger.2022.104739] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Revised: 05/26/2022] [Accepted: 05/29/2022] [Indexed: 11/25/2022]
|
16
|
Zaman BA, Ibrahim SA. Hepcidin-to-Ferritin Ratio as an Early Diagnostic Index of Iron Overload in β-Thalassemia Major Patients. Hemoglobin 2022; 46:106-113. [PMID: 35930276 DOI: 10.1080/03630269.2022.2083969] [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: 01/19/2023]
Abstract
Hepcidin (HEPC) hormone production is expected to be elevated in cases accompanying iron overload, but the opposite impact of ineffective erythropoiesis in β-thalassemia major (β-TM) patients overrides this effect. The role of the HEPC-to-ferritin (FER) ratio and its components in iron metabolism along with their diagnostic cutoff values, sensitivity, specificity, and accuracy in β-TM patients with iron overload, were examined in this study. This was a 1:1 case-control study with 120 participants, ages ranging from 2 to 30 years of both sexes, who were assigned into two groups: 60 β-TM patients with iron overload, and a control group, comprising 60 healthy individuals matched by gender and age. In the present study, we found slightly elevated serum HEPC concentration (21.9 ng/mL) compared to the controls (9.9 ng/mL), which was not statistically significant (p =0.1), and the median HEPC-to-FER ratio of the cases was significantly lower than the controls, with the median case-control difference of (-0.366; p < 0.001). Our results revealed a statistically significant impact (p < 0.001) of mean age on the serum HEPC level with the inverse linear correlation of (-0.487, p < 0.001). The area under the curve of the HEPC-to-FER ratio was 0.999 and the optimum cutoff value was 0.046 ng/mL (p < 0.001) with 100.0% sensitivity and 98.3% specificity. In conclusion, we found that serum HEPC-to-FER ratio, with an accuracy of 99.2%, may serve as an excellent index for the diagnosis of iron overload in β-TM patients differentiating them from nonthalassemic controls.
Collapse
Affiliation(s)
- Burhan A Zaman
- Department of Basic Sciences, College of Pharmacy, University of Duhok, Duhok City, Iraq
| | - Shereen A Ibrahim
- Department of Physiology and Pharmacology, College of Medicine, University of Duhok, Duhok City, Iraq
| |
Collapse
|
17
|
Badenhorst CE, Forsyth AK, Govus AD. A contemporary understanding of iron metabolism in active premenopausal females. Front Sports Act Living 2022; 4:903937. [PMID: 35966107 PMCID: PMC9366739 DOI: 10.3389/fspor.2022.903937] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Accepted: 06/27/2022] [Indexed: 11/13/2022] Open
Abstract
Iron metabolism research in the past decade has identified menstrual blood loss as a key contributor to the prevalence of iron deficiency in premenopausal females. The reproductive hormones estrogen and progesterone influence iron regulation and contribute to variations in iron parameters throughout the menstrual cycle. Despite the high prevalence of iron deficiency in premenopausal females, scant research has investigated female-specific causes and treatments for iron deficiency. In this review, we provide a comprehensive discussion of factors that influence iron status in active premenopausal females, with a focus on the menstrual cycle. We also outline several practical guidelines for monitoring, diagnosing, and treating iron deficiency in premenopausal females. Finally, we highlight several areas for further research to enhance the understanding of iron metabolism in this at-risk population.
Collapse
Affiliation(s)
- Claire E. Badenhorst
- School of Sport, Exercise, and Nutrition, College of Health, Massey University, Auckland, New Zealand
- *Correspondence: Claire E. Badenhorst
| | - Adrienne K. Forsyth
- School of Behavioural and Health Science, Australian Catholic University, Fitzroy, VIC, Australia
| | - Andrew D. Govus
- Discipline of Sport and Exercise, Department of Sport, Exercise, and Nutrition Science, La Trobe University, Melbourne, VIC, Australia
| |
Collapse
|
18
|
Hoving V, Korman SE, Antonopoulos P, Donker AE, Schols SEM, Swinkels DW. IRIDA Phenotype in TMPRSS6 Monoallelic-Affected Patients: Toward a Better Understanding of the Pathophysiology. Genes (Basel) 2022; 13:genes13081309. [PMID: 35893046 PMCID: PMC9331965 DOI: 10.3390/genes13081309] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2022] [Revised: 07/19/2022] [Accepted: 07/21/2022] [Indexed: 12/19/2022] Open
Abstract
Iron-refractory iron deficiency anemia (IRIDA) is an autosomal recessive inherited form of iron deficiency anemia characterized by discrepantly high hepcidin levels relative to body iron status. However, patients with monoallelic exonic TMPRSS6 variants have also been reported to express the IRIDA phenotype. The pathogenesis of an IRIDA phenotype in these patients is unknown and causes diagnostic uncertainty. Therefore, we retrospectively summarized the data of 16 patients (4 men, 12 women) who expressed the IRIDA phenotype in the presence of only a monoallelic TMPRSS6 variant. Eight unaffected relatives with identical exonic TMPRSS6 variants were used as controls. Haplotype analysis was performed to assess the (intra)genetic differences between patients and relatives. The expression and severity of the IRIDA phenotype were highly variable. Compared with their relatives, patients showed lower Hb, MCV, and TSAT/hepcidin ratios and inherited a different wild-type allele. We conclude that IRIDA in monoallelic TMPRSS6-affected patients is a phenotypically and genotypically heterogeneous disease that is more common in female patients. We hypothesize that allelic imbalance, polygenetic inheritance, or modulating environmental factors and their complex interplay are possible causes. This explorative study is the first step toward improved insights into the pathophysiology and improved diagnostic accuracy for patients presenting with IRIDA and a monoallelic exonic TMPRSS6 variant.
Collapse
Affiliation(s)
- Vera Hoving
- Department of Hematology, Radboud University Medical Center, Geert Grooteplein Zuid 10, 6525 GE Nijmegen, The Netherlands;
| | - Scott E. Korman
- Translational Metabolic Laboratory, Department of Laboratory Medicine, Radboud University Medical Center, Geert Grooteplein Zuid 10, 6525 GE Nijmegen, The Netherlands; (S.E.K.); (P.A.); (D.W.S.)
| | - Petros Antonopoulos
- Translational Metabolic Laboratory, Department of Laboratory Medicine, Radboud University Medical Center, Geert Grooteplein Zuid 10, 6525 GE Nijmegen, The Netherlands; (S.E.K.); (P.A.); (D.W.S.)
| | - Albertine E. Donker
- Department of Pediatrics, Máxima Medical Center, De Run 4600, 5504 NB Veldhoven, The Netherlands;
| | - Saskia E. M. Schols
- Department of Hematology, Radboud University Medical Center, Geert Grooteplein Zuid 10, 6525 GE Nijmegen, The Netherlands;
- Correspondence:
| | - Dorine W. Swinkels
- Translational Metabolic Laboratory, Department of Laboratory Medicine, Radboud University Medical Center, Geert Grooteplein Zuid 10, 6525 GE Nijmegen, The Netherlands; (S.E.K.); (P.A.); (D.W.S.)
- Sanquin Blood Bank, Sanquin Diagnostics BV, Plesmanlaan 125, 1066 NH Amsterdam, The Netherlands
| |
Collapse
|
19
|
Pradhan-Sundd T, Kato GJ, Novelli EM. Molecular Mechanisms of Hepatic Dysfunction in Sickle Cell Disease: Lessons From The Townes Mouse Model. Am J Physiol Cell Physiol 2022; 323:C494-C504. [PMID: 35759437 PMCID: PMC9359658 DOI: 10.1152/ajpcell.00175.2022] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Sickle cell disease (SCD) is an autosomal-recessive-genetic disorder that affects ~100,000 Americans and millions of people worldwide. Erythrocyte sickling, vaso-occlusion, sterile inflammation and hemolysis are the major pathophysiological pathways leading to liver injury in SCD. Although hepatic dysfunction affects up to 10-40% of SCD patients, therapeutic approaches to prevent liver injury in SCD are not known, and the molecular mechanisms promoting progressive liver injury in SCD remain poorly understood. Animal models have been beneficial in bridging the gap between preclinical and translational research in SCD. Recent advances in methodology have allowed the development of several humanized mouse models to address various aspects of SCD related liver diseases. This review provides an overview of current knowledge of the molecular mechanisms and potential therapeutic options of SCD associated liver dysfunction using the Townes mouse model.
Collapse
Affiliation(s)
- Tirthadipa Pradhan-Sundd
- Pittsburgh Heart, Liver and Blood Vascular Medicine Institute, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States.,Division of Hematology/Oncology, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| | | | - Enrico M Novelli
- Pittsburgh Heart, Liver and Blood Vascular Medicine Institute, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States.,Division of Hematology/Oncology, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| |
Collapse
|
20
|
Shahandeh A, Bui BV, Finkelstein DI, Nguyen CTO. Effects of Excess Iron on the Retina: Insights From Clinical Cases and Animal Models of Iron Disorders. Front Neurosci 2022; 15:794809. [PMID: 35185447 PMCID: PMC8851357 DOI: 10.3389/fnins.2021.794809] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2021] [Accepted: 12/17/2021] [Indexed: 01/19/2023] Open
Abstract
Iron plays an important role in a wide range of metabolic pathways that are important for neuronal health. Excessive levels of iron, however, can promote toxicity and cell death. An example of an iron overload disorder is hemochromatosis (HH) which is a genetic disorder of iron metabolism in which the body’s ability to regulate iron absorption is altered, resulting in iron build-up and injury in several organs. The retina was traditionally assumed to be protected from high levels of systemic iron overload by the blood-retina barrier. However, recent data shows that expression of genes that are associated with HH can disrupt retinal iron metabolism. Thus, the effects of iron overload on the retina have become an area of research interest, as excessively high levels of iron are implicated in several retinal disorders, most notably age–related macular degeneration. This review is an effort to highlight risk factors for excessive levels of systemic iron build-up in the retina and its potential impact on the eye health. Information is integrated across clinical and preclinical animal studies to provide insights into the effects of systemic iron loading on the retina.
Collapse
Affiliation(s)
- Ali Shahandeh
- Department of Optometry and Vision Sciences, Faculty of Medicine, Dentistry and Health Sciences, University of Melbourne, Parkville, VIC, Australia
| | - Bang V. Bui
- Department of Optometry and Vision Sciences, Faculty of Medicine, Dentistry and Health Sciences, University of Melbourne, Parkville, VIC, Australia
| | - David I. Finkelstein
- Florey Department of Neuroscience and Mental Health, University of Melbourne, Parkville, VIC, Australia
| | - Christine T. O. Nguyen
- Department of Optometry and Vision Sciences, Faculty of Medicine, Dentistry and Health Sciences, University of Melbourne, Parkville, VIC, Australia
- *Correspondence: Christine T. O. Nguyen,
| |
Collapse
|
21
|
Fisher AL, Babitt JL. Coordination of iron homeostasis by bone morphogenetic proteins: Current understanding and unanswered questions. Dev Dyn 2022; 251:26-46. [PMID: 33993583 PMCID: PMC8594283 DOI: 10.1002/dvdy.372] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Revised: 04/15/2021] [Accepted: 05/07/2021] [Indexed: 01/19/2023] Open
Abstract
Iron homeostasis is tightly regulated to balance the iron requirement for erythropoiesis and other vital cellular functions, while preventing cellular injury from iron excess. The liver hormone hepcidin is the master regulator of systemic iron balance by controlling the degradation and function of the sole known mammalian iron exporter ferroportin. Liver hepcidin expression is coordinately regulated by several signals that indicate the need for more or less iron, including plasma and tissue iron levels, inflammation, and erythropoietic drive. Most of these signals regulate hepcidin expression by modulating the activity of the bone morphogenetic protein (BMP)-SMAD pathway, which controls hepcidin transcription. Genetic disorders of iron overload and iron deficiency have identified several hepatocyte membrane proteins that play a critical role in mediating the BMP-SMAD and hepcidin regulatory response to iron. However, the precise molecular mechanisms by which serum and tissue iron levels are sensed to regulate BMP ligand production and promote the physical and/or functional interaction of these proteins to modulate SMAD signaling and hepcidin expression remain uncertain. This critical commentary will focus on the current understanding and key unanswered questions regarding how the liver senses iron levels to regulate BMP-SMAD signaling and thereby hepcidin expression to control systemic iron homeostasis.
Collapse
Affiliation(s)
| | - Jodie L Babitt
- Corresponding author: Jodie L Babitt, Division of Nephrology, Program in Membrane Biology, Massachusetts General Hospital, Harvard Medical School, Boston, MA. Mailing address: 185 Cambridge St., CPZN-8208, Boston, MA 02114. Telephone: +1 (617) 643-3181.
| |
Collapse
|
22
|
Schumacher KA, Gosmanov AR. Hemochromatosis Gene Mutation in Persons Developing Erythrocytosis on Combined Testosterone and SGLT-2 Inhibitor Therapy. J Investig Med High Impact Case Rep 2022; 10:23247096221111774. [PMID: 35848311 PMCID: PMC9290160 DOI: 10.1177/23247096221111774] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Revised: 06/03/2022] [Accepted: 06/18/2022] [Indexed: 01/10/2023] Open
Abstract
In clinical trials, sodium-glucose cotransporter-2 inhibitors (SGLT-2i) use alone in persons with type 2 diabetes (T2D) or testosterone replacement therapy (TRT) prescription alone in men with hypogonadism was shown to lead to a modest but significant increase in red blood cell mass. Recent evidence indicates that combined use of TRT and SGLT-2i in persons with T2D may be associated with risk of erythrocytosis. However, factor(s) that may lead to the development of erythrocytosis in these patients is unknown. We describe here 5 consecutive patients with hypogonadism on chronic TRT who developed erythrocytosis following addition of SGLT-2i empagliflozin for optimization of T2D management. In addition to the careful review of medical history, all patients underwent genetic screening for hereditary hemochromatosis. We have found that none of the patients had C282Y mutation in the HFE (Homeostatic Iron Regulator) gene and 4 out of 5 patients had heterozygosity in the H63D allele. Upon TRT discontinuation or its dose reduction or referral for scheduled phlebotomy, patients showed resolution of erythrocytosis. Our study reaffirms that practitioners should monitor for changes in hematocrit following the initiation of SGLT-2i in persons with T2D and hypogonadism on chronic TRT. Also, for the first time, we showed that in some of the patients receiving combined TRT and SGLT-2i H63D heterozygosity in the HFE gene may mediate the development of new-onset erythrocytosis.
Collapse
Affiliation(s)
| | - Aidar R. Gosmanov
- Albany Medical College, NY, USA
- Albany Stratton VA Medical Center, NY, USA
| |
Collapse
|
23
|
Lanser L, Fuchs D, Kurz K, Weiss G. Physiology and Inflammation Driven Pathophysiology of Iron Homeostasis-Mechanistic Insights into Anemia of Inflammation and Its Treatment. Nutrients 2021; 13:3732. [PMID: 34835988 PMCID: PMC8619077 DOI: 10.3390/nu13113732] [Citation(s) in RCA: 66] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2021] [Revised: 10/18/2021] [Accepted: 10/19/2021] [Indexed: 02/07/2023] Open
Abstract
Anemia is very common in patients with inflammatory disorders. Its prevalence is associated with severity of the underlying disease, and it negatively affects quality of life and cardio-vascular performance of patients. Anemia of inflammation (AI) is caused by disturbances of iron metabolism resulting in iron retention within macrophages, a reduced erythrocyte half-life, and cytokine mediated inhibition of erythropoietin function and erythroid progenitor cell differentiation. AI is mostly mild to moderate, normochromic and normocytic, and characterized by low circulating iron, but normal and increased levels of the storage protein ferritin and the iron hormone hepcidin. The primary therapeutic approach for AI is treatment of the underlying inflammatory disease which mostly results in normalization of hemoglobin levels over time unless other pathologies such as vitamin deficiencies, true iron deficiency on the basis of bleeding episodes, or renal insufficiency are present. If the underlying disease and/or anemia are not resolved, iron supplementation therapy and/or treatment with erythropoietin stimulating agents may be considered whereas blood transfusions are an emergency treatment for life-threatening anemia. New treatments with hepcidin-modifying strategies and stabilizers of hypoxia inducible factors emerge but their therapeutic efficacy for treatment of AI in ill patients needs to be evaluated in clinical trials.
Collapse
Affiliation(s)
- Lukas Lanser
- Department of Internal Medicine II, Medical University of Innsbruck, 6020 Innsbruck, Austria; (L.L.); (K.K.)
| | - Dietmar Fuchs
- Division of Biological Chemistry, Biocenter, Medical University of Innsbruck, 6020 Innsbruck, Austria;
| | - Katharina Kurz
- Department of Internal Medicine II, Medical University of Innsbruck, 6020 Innsbruck, Austria; (L.L.); (K.K.)
| | - Günter Weiss
- Department of Internal Medicine II, Medical University of Innsbruck, 6020 Innsbruck, Austria; (L.L.); (K.K.)
- Christian Doppler Laboratory for Iron Metabolism and Anemia Research, Medical University of Innsbruck, 6020 Innsbruck, Austria
| |
Collapse
|
24
|
Xu Y, Alfaro-Magallanes VM, Babitt JL. Physiological and pathophysiological mechanisms of hepcidin regulation: clinical implications for iron disorders. Br J Haematol 2021; 193:882-893. [PMID: 33316086 PMCID: PMC8164969 DOI: 10.1111/bjh.17252] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Accepted: 11/04/2020] [Indexed: 02/06/2023]
Abstract
The discovery of hepcidin has provided a solid foundation for understanding the mechanisms of systemic iron homeostasis and the aetiologies of iron disorders. Hepcidin assures the balance of circulating and stored iron levels for multiple physiological processes including oxygen transport and erythropoiesis, while limiting the toxicity of excess iron. The liver is the major site where regulatory signals from iron, erythropoietic drive and inflammation are integrated to control hepcidin production. Pathologically, hepcidin dysregulation by genetic inactivation, ineffective erythropoiesis, or inflammation leads to diseases of iron deficiency or overload such as iron-refractory iron-deficiency anaemia, anaemia of inflammation, iron-loading anaemias and hereditary haemochromatosis. In the present review, we discuss recent insights into the molecular mechanisms governing hepcidin regulation, how these pathways are disrupted in iron disorders, and how this knowledge is being used to develop novel diagnostic and therapeutic strategies.
Collapse
Affiliation(s)
- Yang Xu
- Division of Nephrology, Program in Membrane Biology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Víctor M. Alfaro-Magallanes
- Division of Nephrology, Program in Membrane Biology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- LFE Research Group, Department of Health and Human Performance, Faculty of Physical Activity and Sport Sciences, Universidad Politécnica de Madrid (UPM), Madrid, Spain
| | - Jodie L. Babitt
- Division of Nephrology, Program in Membrane Biology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
25
|
Kowdley KV, Gochanour EM, Sundaram V, Shah RA, Handa P. Hepcidin Signaling in Health and Disease: Ironing Out the Details. Hepatol Commun 2021; 5:723-735. [PMID: 34027264 PMCID: PMC8122377 DOI: 10.1002/hep4.1717] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Revised: 02/22/2021] [Accepted: 02/23/2021] [Indexed: 12/19/2022] Open
Abstract
Hepcidin, a peptide hormone produced by hepatocytes, is the central regulator of systemic iron homeostasis through its interaction with ferroportin, the major cellular iron export protein. Hepcidin binding to ferroportin results in reduced iron export from macrophages and intestinal absorptive cells, leading to decreased serum iron levels. Hepcidin expression is influenced by several factors that include serum and liver iron stores, erythropoiesis, hypoxia, inflammation, and infection. Erythropoietic drive and hypoxia suppress hepcidin expression and promote red cell production. In contrast, inflammation and infection are associated with increased hepcidin production to sequester iron intracellularly as a means of depriving microorganisms of iron. Chronic inflammation may up-regulate hepcidin expression through the interleukin-6 (IL-6)-Janus kinase 2 (JAK2)-signal transducer and activator of transcription 3 (STAT3) pathway. The bone morphogenetic protein (BMP)-mothers against decapentaplegic homolog (SMAD) pathway is a major positive driver of hepcidin expression in response to either increased circulating iron in the form of transferrin or iron loading in organs. Hereditary hemochromatosis (HH) consists of several inherited disorders that cause inappropriately reduced hepcidin expression in response to body iron stores, leading to increased iron absorption from a normal diet. The most common form of HH is due to a mutation in the HFE gene, which causes a failure in the hepatocyte iron-sensing mechanism, leading to reduced hepcidin expression; the clinical manifestations of HFE-HH include increased serum transferrin-iron saturation and progressive iron loading in the liver and other tissues over time among patients who express the disease phenotype. In this article, we review the physiologic mechanisms and cellular pathways by which hepcidin expression is regulated, and the different forms of HH resulting from various mutations that cause hepcidin deficiency. We also review other drivers of hepcidin expression and the associated pathophysiologic consequences.
Collapse
Affiliation(s)
- Kris V. Kowdley
- Liver Institute Northwest and Elson S. Floyd College of MedicineWashington State UniversitySpokaneWAUSA
- Liver Care Network and Organ Care ResearchSwedish Medical CenterSeattleWAUSA
| | - Eric M. Gochanour
- Liver Institute Northwest and Elson S. Floyd College of MedicineWashington State UniversitySpokaneWAUSA
- Liver Care Network and Organ Care ResearchSwedish Medical CenterSeattleWAUSA
| | - Vinay Sundaram
- Division of Gastroenterology and Comprehensive Transplant CenterLos AngelesCAUSA
| | - Raj A. Shah
- Liver Care Network and Organ Care ResearchSwedish Medical CenterSeattleWAUSA
| | - Priya Handa
- Liver Care Network and Organ Care ResearchSwedish Medical CenterSeattleWAUSA
| |
Collapse
|
26
|
Milanese C, Gabriels S, Barnhoorn S, Cerri S, Ulusoy A, Gornati SV, Wallace DF, Blandini F, Di Monte DA, Subramaniam VN, Mastroberardino PG. Gender biased neuroprotective effect of Transferrin Receptor 2 deletion in multiple models of Parkinson's disease. Cell Death Differ 2021; 28:1720-1732. [PMID: 33323945 PMCID: PMC8166951 DOI: 10.1038/s41418-020-00698-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Revised: 11/16/2020] [Accepted: 11/24/2020] [Indexed: 01/28/2023] Open
Abstract
Alterations in the metabolism of iron and its accumulation in the substantia nigra pars compacta accompany the pathogenesis of Parkinson's disease (PD). Changes in iron homeostasis also occur during aging, which constitutes a PD major risk factor. As such, mitigation of iron overload via chelation strategies has been considered a plausible disease modifying approach. Iron chelation, however, is imperfect because of general undesired side effects and lack of specificity; more effective approaches would rely on targeting distinctive pathways responsible for iron overload in brain regions relevant to PD and, in particular, the substantia nigra. We have previously demonstrated that the Transferrin/Transferrin Receptor 2 (TfR2) iron import mechanism functions in nigral dopaminergic neurons, is perturbed in PD models and patients, and therefore constitutes a potential therapeutic target to halt iron accumulation. To validate this hypothesis, we generated mice with targeted deletion of TfR2 in dopaminergic neurons. In these animals, we modeled PD with multiple approaches, based either on neurotoxin exposure or alpha-synuclein proteotoxic mechanisms. We found that TfR2 deletion can provide neuroprotection against dopaminergic degeneration, and against PD- and aging-related iron overload. The effects, however, were significantly more pronounced in females rather than in males. Our data indicate that the TfR2 iron import pathway represents an amenable strategy to hamper PD progression. Data also suggest, however, that therapeutic strategies targeting TfR2 should consider a potential sexual dimorphism in neuroprotective response.
Collapse
Affiliation(s)
- Chiara Milanese
- Department of Molecular Genetics, Rotterdam, the Netherlands ,grid.7678.e0000 0004 1757 7797IFOM-The FIRC Institute of Molecular Oncology, Milan, Italy
| | - Sylvia Gabriels
- Department of Molecular Genetics, Rotterdam, the Netherlands
| | | | | | - Ayse Ulusoy
- grid.424247.30000 0004 0438 0426German Centre for Neurodegenerative Diseases (DZNE), 53175 Bonn, Germany
| | - S. V. Gornati
- grid.5645.2000000040459992XDepartment of Neuroscience Erasmus MC, Rotterdam, the Netherlands
| | - Daniel F. Wallace
- grid.1024.70000000089150953School of Biomedical Sciences, Institute of Health and Biomedical Innovation, Queensland University of Technology (QUT), Brisbane, QLD Australia
| | - Fabio Blandini
- IRCCS Mondino Foundation, 27100 Pavia, Italy ,grid.8982.b0000 0004 1762 5736Department of Brain and Behavioral Sciences, University of Pavia, Pavia, Italy
| | - Donato A. Di Monte
- grid.424247.30000 0004 0438 0426German Centre for Neurodegenerative Diseases (DZNE), 53175 Bonn, Germany
| | - V. Nathan Subramaniam
- grid.1024.70000000089150953School of Biomedical Sciences, Institute of Health and Biomedical Innovation, Queensland University of Technology (QUT), Brisbane, QLD Australia
| | - Pier G. Mastroberardino
- Department of Molecular Genetics, Rotterdam, the Netherlands ,grid.7678.e0000 0004 1757 7797IFOM-The FIRC Institute of Molecular Oncology, Milan, Italy ,grid.158820.60000 0004 1757 2611Department of Life, Health, and Environmental Sciences, University of L’Aquila, L’Aquila, Italy
| |
Collapse
|
27
|
Anderson GJ, Bardou-Jacquet E. Revisiting hemochromatosis: genetic vs. phenotypic manifestations. ANNALS OF TRANSLATIONAL MEDICINE 2021; 9:731. [PMID: 33987429 PMCID: PMC8106074 DOI: 10.21037/atm-20-5512] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Iron overload disorders represent an important class of human diseases. Of the primary iron overload conditions, by far the most common and best studied is HFE-related hemochromatosis, which results from homozygosity for a mutation leading to the C282Y substitution in the HFE protein. This disease is characterized by reduced expression of the iron-regulatory hormone hepcidin, leading to increased dietary iron absorption and iron deposition in multiple tissues including the liver, pancreas, joints, heart and pituitary. The phenotype of HFE-related hemochromatosis is quite variable, with some individuals showing little or no evidence of increased body iron, yet others showing severe iron loading, tissue damage and clinical sequelae. The majority of genetically predisposed individuals show at least some evidence of iron loading (increased transferrin saturation and serum ferritin), but a minority show clinical symptoms and severe consequences are rare. Thus, the disorder has a high biochemical penetrance, but a low clinical prevalence. Nevertheless, it is such a common condition in Caucasian populations (1:100–200) that it remains an important clinical entity. The phenotypic variability can largely be explained by a range of environmental, genetic and physiological factors. Men are far more likely to manifest significant disease than women, with the latter losing iron through menstrual blood loss and childbirth. Other forms of blood loss, immune system influences, the amount of bioavailable iron in the diet and lifestyle factors such as high alcohol intake can also contribute to iron loading and disease expression. Polymorphisms in a range of genes have been linked to variations in body iron levels, both in the general population and in hemochromatosis. Some of the genes identified play well known roles in iron homeostasis, yet others are novel. Other factors, including both co-morbidities and genetic polymorphisms, do not affect iron levels per se, but determine the propensity for tissue pathology.
Collapse
Affiliation(s)
- Gregory J Anderson
- Iron Metabolism Laboratory, QIMR Berghofer Medical Research Institute and School of Chemistry and Molecular Bioscience, University of Queensland, Brisbane, Queensland, Australia
| | - Edouard Bardou-Jacquet
- Liver Disease Department, University of Rennes and French Reference Center for Hemochromatosis and Iron Metabolism Disease, Rennes, France
| |
Collapse
|
28
|
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
|
29
|
Tkaczyszyn M, Drozd M, Węgrzynowska-Teodorczyk K, Bojarczuk J, Majda J, Banasiak W, Ponikowski P, Jankowska EA. Iron status, catabolic/anabolic balance, and skeletal muscle performance in men with heart failure with reduced ejection fraction. Cardiol J 2020; 28:391-401. [PMID: 33140393 DOI: 10.5603/cj.a2020.0138] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Revised: 10/26/2020] [Accepted: 10/13/2020] [Indexed: 11/25/2022] Open
Abstract
BACKGROUND Metabolic derangements related to tissue energetics constitute an important pathophysiological feature of heart failure. We investigated whether iron deficiency and catabolic/anabolic imbalance contribute to decreased skeletal muscle performance in men with heart failure with reduced ejection fraction (HFrEF), and whether these pathologies are related to each other. METHODS We comprehensively examined 23 men with stable HFrEF (median age [interquartile range]: 63 [59-66] years; left ventricular ejection fraction: 28 [25-35]%; New York Heart Association class I/II/III: 17/43/39%). We analyzed clinical characteristics, iron status, hormones, strength and fatigability of forearm flexors and quadriceps (surface electromyography), and exercise capacity (6-minute walking test). RESULTS None of the patients had anemia whereas 8 were iron-deficient. Flexor carpi radialis fatigability correlated with lower reticulocyte hemoglobin content (CHR, p < 0.05), and there was a trend towards greater fatigability in patients with higher body mass index and lower serum ferritin (both p < 0.1). Flexor carpi ulnaris fatigability correlated with lower serum iron and CHR (both p < 0.05). Vastus medialis fatigability was related to lower free and bioavailable testosterone (FT and BT, respectively, both p < 0.05), and 6-minute walking test distance was shorter in patients with higher cortisol/FT and cortisol/BT ratio (both p < 0.05). Lower ferritin and transferrin saturation correlated with lower percentage of FT and BT. Men with HFrEF and iron deficiency had higher total testosterone, but lower percentage of FT and BT. CONCLUSIONS Iron deficiency correlates with lower bioactive testosterone in men with HFrEF. These two pathologies can both contribute to decreased skeletal muscle performance in such patients.
Collapse
|
30
|
Corradini E, Buzzetti E, Pietrangelo A. Genetic iron overload disorders. Mol Aspects Med 2020; 75:100896. [PMID: 32912773 DOI: 10.1016/j.mam.2020.100896] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Revised: 08/11/2020] [Accepted: 08/17/2020] [Indexed: 02/06/2023]
Abstract
Due to its pivotal role in orchestrating vital cellular functions and metabolic processes, iron is an essential component of the human body and a main micronutrient in the human diet. However, excess iron causes an increased production of reactive oxygen species leading to cell dysfunction or death, tissue damage and organ disease. Iron overload disorders encompass a wide spectrum of pathological conditions of hereditary or acquired origin. A number of 'iron genes' have been identified as being associated with hereditary iron overload syndromes, the most common of which is hemochromatosis. Although linked to at least five different genes, hemochromatosis is recognized as a unique syndromic entity based on a common pathogenetic mechanism leading to excessive entry of unneeded iron into the bloodstream. In this review, we focus on the pathophysiologic basis and clinical aspects of the most common genetic iron overload syndromes in humans.
Collapse
Affiliation(s)
- Elena Corradini
- Department of Medical and Surgical Sciences, University of Modena and Reggio Emilia; Internal Medicine and Centre for Hemochromatosis and Heredometabolic Liver Diseases, ERN -EuroBloodNet Center, Azienda Ospedaliero-Universitaria di Modena, Policlinico, Modena, Italy
| | - Elena Buzzetti
- Department of Medical and Surgical Sciences, University of Modena and Reggio Emilia; Internal Medicine and Centre for Hemochromatosis and Heredometabolic Liver Diseases, ERN -EuroBloodNet Center, Azienda Ospedaliero-Universitaria di Modena, Policlinico, Modena, Italy
| | - Antonello Pietrangelo
- Department of Medical and Surgical Sciences, University of Modena and Reggio Emilia; Internal Medicine and Centre for Hemochromatosis and Heredometabolic Liver Diseases, ERN -EuroBloodNet Center, Azienda Ospedaliero-Universitaria di Modena, Policlinico, Modena, Italy.
| |
Collapse
|
31
|
Altamura S, Marques O, Colucci S, Mertens C, Alikhanyan K, Muckenthaler MU. Regulation of iron homeostasis: Lessons from mouse models. Mol Aspects Med 2020; 75:100872. [DOI: 10.1016/j.mam.2020.100872] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2020] [Revised: 06/28/2020] [Accepted: 07/02/2020] [Indexed: 12/13/2022]
|
32
|
Xiao X, Alfaro-Magallanes VM, Babitt JL. Bone morphogenic proteins in iron homeostasis. Bone 2020; 138:115495. [PMID: 32585319 PMCID: PMC7453787 DOI: 10.1016/j.bone.2020.115495] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/15/2020] [Revised: 06/12/2020] [Accepted: 06/15/2020] [Indexed: 02/07/2023]
Abstract
The bone morphogenetic protein (BMP)-SMAD signaling pathway plays a central role in regulating hepcidin, which is the master hormone governing systemic iron homeostasis. Hepcidin is produced by the liver and acts on the iron exporter ferroportin to control iron absorption from the diet and iron release from body stores, thereby providing adequate iron for red blood cell production, while limiting the toxic effects of excess iron. BMP6 and BMP2 ligands produced by liver endothelial cells bind to BMP receptors and the coreceptor hemojuvelin (HJV) on hepatocytes to activate SMAD1/5/8 signaling, which directly upregulates hepcidin transcription. Most major signals that influence hepcidin production, including iron, erythropoietic drive, and inflammation, intersect with the BMP-SMAD pathway to regulate hepcidin transcription. Mutation or inactivation of BMP ligands, BMP receptors, HJV, SMADs or other proteins that modulate the BMP-SMAD pathway result in hepcidin dysregulation, leading to iron-related disorders, such as hemochromatosis and iron refractory iron deficiency anemia. Pharmacologic modulators of the BMP-SMAD pathway have shown efficacy in pre-clinical models to regulate hepcidin expression and treat iron-related disorders. This review will discuss recent insights into the role of the BMP-SMAD pathway in regulating hepcidin to control systemic iron homeostasis.
Collapse
Affiliation(s)
- Xia Xiao
- Division of Nephrology, Program in Membrane Biology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Víctor M Alfaro-Magallanes
- Division of Nephrology, Program in Membrane Biology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA; LFE Research Group, Department of Health and Human Performance, Faculty of Physical Activity and Sport Sciences, Universidad Politécnica de Madrid (UPM), Madrid, Spain
| | - Jodie L Babitt
- Division of Nephrology, Program in Membrane Biology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
33
|
Xiao X, Dev S, Canali S, Bayer A, Xu Y, Agarwal A, Wang CY, Babitt JL. Endothelial Bone Morphogenetic Protein 2 (Bmp2) Knockout Exacerbates Hemochromatosis in Homeostatic Iron Regulator (Hfe) Knockout Mice but not Bmp6 Knockout Mice. Hepatology 2020; 72:642-655. [PMID: 31778583 PMCID: PMC7253321 DOI: 10.1002/hep.31048] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/21/2018] [Accepted: 11/13/2019] [Indexed: 12/15/2022]
Abstract
BACKGROUND AND AIMS Bone morphogenetic proteins BMP2 and BMP6 play key roles in systemic iron homeostasis by regulating production of the iron hormone hepcidin. The homeostatic iron regulator (HFE) also regulates hepcidin through a mechanism that intersects with the BMP-mothers against decapentaplegic homolog 1/5/8 (SMAD1/5/8) pathway. However, the relative roles of BMP2 compared with BMP6 and whether HFE regulates hepcidin through a BMP2-dependent mechanism remain uncertain. APPROACH AND RESULTS We therefore examined the iron phenotype of mice deficient for both Bmp2 and Bmp6 or both Bmp2 and Hfe compared with single knockout (KO) mice and littermate controls. Eight-week-old double endothelial Bmp6/Bmp2 KO mice exhibited a similar degree of hepcidin deficiency, serum iron overload, and tissue iron overload compared with single KO mice. Notably, dietary iron loading still induced liver SMAD5 phosphorylation and hepcidin in double Bmp6/endothelial Bmp2 KO mice, although no other BMP ligand mRNAs were increased in the livers of double KO mice, and only Bmp6 and Bmp2 mRNA were induced by dietary iron loading in wild-type mice. In contrast, double Hfe/endothelial Bmp2 KO mice exhibited reduced hepcidin and increased extrahepatic iron loading compared to single Hfe or endothelial Bmp2 KO mice. Liver phosphorylated SMAD5 and the SMAD1/5/8 target inhibitor of DNA binding 1 (Id1) mRNA were also reduced in double Hfe/endothelial Bmp2 KO compared with single endothelial Bmp2 KO female mice. Finally, hepcidin and Id1 mRNA induction by homodimeric BMP2, homodimeric BMP6, and heterodimeric BMP2/6 were blunted in Hfe KO primary hepatocytes. CONCLUSIONS These data suggest that BMP2 and BMP6 work collaboratively to regulate hepcidin expression, that BMP2-independent and BMP6-independent SMAD1/5/8 signaling contributes a nonredundant role to hepcidin regulation by iron, and that HFE regulates hepcidin at least in part through a BMP2-independent but SMAD1/5/8-dependent mechanism.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Jodie L. Babitt
- Contact Information Jodie L. Babitt MD, Massachusetts General Hospital, 185 Cambridge St., CPZN-8208, Boston, MA 02114, Phone: (617)-643-3181, Fax: (617)-643-3182,
| |
Collapse
|
34
|
Yang L, Wang H, Yang X, Wu Q, An P, Jin X, Liu W, Huang X, Li Y, Yan S, Shen S, Liang T, Min J, Wang F. Auranofin mitigates systemic iron overload and induces ferroptosis via distinct mechanisms. Signal Transduct Target Ther 2020; 5:138. [PMID: 32732975 PMCID: PMC7393508 DOI: 10.1038/s41392-020-00253-0] [Citation(s) in RCA: 178] [Impact Index Per Article: 35.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Revised: 06/21/2020] [Accepted: 06/29/2020] [Indexed: 02/07/2023] Open
Abstract
Iron homeostasis is essential for health; moreover, hepcidin-deficiency results in iron overload in both hereditary hemochromatosis and iron-loading anemia. Here, we identified iron modulators by functionally screening hepcidin agonists using a library of 640 FDA-approved drugs in human hepatic Huh7 cells. We validated the results in C57BL/6J mice and a mouse model of hemochromatosis (Hfe−/− mice). Our screen revealed that the anti-rheumatoid arthritis drug auranofin (AUR) potently upregulates hepcidin expression. Interestingly, we found that canonical signaling pathways that regulate iron, including the Bmp/Smad and IL-6/Jak2/Stat3 pathways, play indispensable roles in mediating AUR’s effects. In addition, AUR induces IL-6 via the NF-κB pathway. In C57BL/6J mice, acute treatment with 5 mg/kg AUR activated hepatic IL-6/hepcidin signaling and decreased serum iron and transferrin saturation. Whereas chronically treating male Hfe−/− mice with 5 mg/kg AUR activated hepatic IL-6/hepcidin signaling, decreasing systemic iron overload, but less effective in females. Further analyses revealed that estrogen reduced the ability of AUR to induce IL-6/hepcidin signaling in Huh7 cells, providing a mechanistic explanation for ineffectiveness of AUR in female Hfe−/− mice. Notably, high-dose AUR (25 mg/kg) induces ferroptosis and causes lipid peroxidation through inhibition of thioredoxin reductase (TXNRD) activity. We demonstrate the ferroptosis inhibitor ferrostatin significantly protects liver toxicity induced by high-dose AUR without comprising its beneficial effect on iron metabolism. In conclusion, our findings provide compelling evidence that TXNRD is a key regulator of ferroptosis, and AUR is a novel activator of hepcidin and ferroptosis via distinct mechanisms, suggesting a promising approach for treating hemochromatosis and hepcidin-deficiency related disorders.
Collapse
Affiliation(s)
- Lei Yang
- Department of Nutrition, Precision Nutrition Innovation Center, School of Public Health, Zhengzhou University, 450001, Zhengzhou, China.,The First Affiliated Hospital, School of Public Health, Institute of Translational Medicine, Zhejiang University School of Medicine, 310058, Hangzhou, China.,Department of Nutrition and Health, Beijing Advanced Innovation Center for Food Nutrition and Human Health, China Agricultural University, 100193, Beijing, China.,School of Nursing, Xinxiang Medical University, 453003, Xinxiang, China
| | - Hao Wang
- Department of Nutrition, Precision Nutrition Innovation Center, School of Public Health, Zhengzhou University, 450001, Zhengzhou, China.,The First Affiliated Hospital, School of Public Health, Institute of Translational Medicine, Zhejiang University School of Medicine, 310058, Hangzhou, China.,Department of Nutrition and Health, Beijing Advanced Innovation Center for Food Nutrition and Human Health, China Agricultural University, 100193, Beijing, China
| | - Xiang Yang
- The First Affiliated Hospital, School of Public Health, Institute of Translational Medicine, Zhejiang University School of Medicine, 310058, Hangzhou, China
| | - Qian Wu
- The First Affiliated Hospital, School of Public Health, Institute of Translational Medicine, Zhejiang University School of Medicine, 310058, Hangzhou, China
| | - Peng An
- Department of Nutrition and Health, Beijing Advanced Innovation Center for Food Nutrition and Human Health, China Agricultural University, 100193, Beijing, China
| | - Xi Jin
- The First Affiliated Hospital, School of Public Health, Institute of Translational Medicine, Zhejiang University School of Medicine, 310058, Hangzhou, China
| | - Weiwei Liu
- Department of Nutrition, Precision Nutrition Innovation Center, School of Public Health, Zhengzhou University, 450001, Zhengzhou, China
| | - Xin Huang
- Department of Nutrition, Precision Nutrition Innovation Center, School of Public Health, Zhengzhou University, 450001, Zhengzhou, China
| | - Yuzhu Li
- The First Affiliated Hospital, School of Public Health, Institute of Translational Medicine, Zhejiang University School of Medicine, 310058, Hangzhou, China
| | - Shiyu Yan
- The First Affiliated Hospital, School of Public Health, Institute of Translational Medicine, Zhejiang University School of Medicine, 310058, Hangzhou, China
| | - Shuying Shen
- The First Affiliated Hospital, School of Public Health, Institute of Translational Medicine, Zhejiang University School of Medicine, 310058, Hangzhou, China
| | - Tingbo Liang
- The First Affiliated Hospital, School of Public Health, Institute of Translational Medicine, Zhejiang University School of Medicine, 310058, Hangzhou, China.
| | - Junxia Min
- The First Affiliated Hospital, School of Public Health, Institute of Translational Medicine, Zhejiang University School of Medicine, 310058, Hangzhou, China.
| | - Fudi Wang
- Department of Nutrition, Precision Nutrition Innovation Center, School of Public Health, Zhengzhou University, 450001, Zhengzhou, China. .,The First Affiliated Hospital, School of Public Health, Institute of Translational Medicine, Zhejiang University School of Medicine, 310058, Hangzhou, China. .,Department of Nutrition and Health, Beijing Advanced Innovation Center for Food Nutrition and Human Health, China Agricultural University, 100193, Beijing, China.
| |
Collapse
|
35
|
Guo W, Abou Ghayda R, Schmidt PJ, Fleming MD, Bhasin S. The role of iron in mediating testosterone's effects on erythropoiesis in mice. FASEB J 2020; 34:11672-11684. [PMID: 32667087 DOI: 10.1096/fj.202000920rr] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2020] [Revised: 06/15/2020] [Accepted: 06/16/2020] [Indexed: 11/11/2022]
Abstract
Testosterone stimulates iron-dependent erythropoiesis and suppresses hepcidin. To clarify the role of iron in mediating testosterone's effects on erythropoiesis, we induced iron deficiency in mice by feeding low iron diet. Iron-replete and iron-deficient mice were treated weekly with testosterone propionate or vehicle for 3 weeks. Testosterone treatment increased red cell count in iron-replete mice, but, surprisingly, testosterone reduced red cell count in iron-deficient mice. Splenic stress erythropoiesis was stimulated in iron-deficient mice relative to iron-replete mice, and further increased by testosterone treatment, as indicated by the increase in red pulp area, the number of nucleated erythroblasts, and expression levels of TfR1, GATA1, and other erythroid genes. Testosterone treatment of iron-deficient mice increased the ratio of early-to-late erythroblasts in the spleen and bone marrow, and serum LDH level, consistent with ineffective erythropoiesis. In iron-deficient mice, erythropoietin levels were higher but erythropoietin-regulated genes were generally downregulated relative to iron-replete mice, suggesting erythropoietin resistance. Conclusion: Testosterone treatment stimulates splenic stress erythropoiesis in iron-replete as well as iron-deficient mice. However, testosterone worsens anemia in iron-deficient mice because of ineffective erythropoiesis possibly due to erythropoietin resistance associated with iron deficiency. Iron plays an important role in mediating testosterone's effects on erythropoiesis.
Collapse
Affiliation(s)
- Wen Guo
- Research Program in Men's Health: Aging and Metabolism, The Boston Claude D. Pepper Older Americans Independence Center, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Ramy Abou Ghayda
- Research Program in Men's Health: Aging and Metabolism, The Boston Claude D. Pepper Older Americans Independence Center, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Paul J Schmidt
- Department of Pathology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Mark D Fleming
- Department of Pathology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Shalender Bhasin
- Research Program in Men's Health: Aging and Metabolism, The Boston Claude D. Pepper Older Americans Independence Center, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
36
|
Kurz K, Lanser L, Seifert M, Kocher F, Pölzl G, Weiss G. Anaemia, iron status, and gender predict the outcome in patients with chronic heart failure. ESC Heart Fail 2020; 7:1880-1890. [PMID: 32458571 PMCID: PMC7373900 DOI: 10.1002/ehf2.12755] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2020] [Revised: 04/20/2020] [Accepted: 04/27/2020] [Indexed: 12/11/2022] Open
Abstract
Aims Anaemia and iron deficiency (ID) are frequently found in patients with chronic heart failure (CHF) and associated with adverse outcome. However, it is unclear whether absolute [transferrin saturation (TSAT) <20%, ferritin <100 μg/L] or inflammation‐driven functional ID (TSAT <20%, ferritin >100 μg/L) with and without anaemia had similar or different consequences for such patients. Methods and results Within this retrospective cohort study, 2223 patients (1601 men and 622 women) with CHF, referred to our department, between 2000 and 2018, were followed for a median time of 84 months. Anaemia was found in 393 patients and was an independent predictor for an adverse outcome [HR 2.164 (95% CI 1.865–2.512), P < 0.001]. In 674 patients with available parameters of iron metabolism, ID was present in 228 patients and was associated with an unfavourable outcome [HR 1.499 (95% CI 1.158–1.940), P = 0.002]. ID was best predicting an adverse outcome in men ≤59 years, with heart failure with reduced ejection fraction, preserved kidney function, no inflammation, and a body mass index (BMI) ≥25.5 kg/m2. Functional ID in women and absolute ID in men were associated with poor prognosis. Of note, TSAT <20% but not low ferritin levels were predictive for an adverse outcome. Anaemic patients with high ferritin levels, advanced inflammation, older age, low BMI, male gender, and reduced glomerular filtration rate had the worst prognosis. Conclusions Anaemia and low tissue iron availability as reflected by TSAT <20% are negative predictors of outcome in patients with CHF. Systemic inflammation, renal function, BMI, age, and gender are important contributors for the clinical course.
Collapse
Affiliation(s)
- Katharina Kurz
- Department of Internal Medicine II, Innsbruck Medical University, Anichstraße 35, 6020, Innsbruck, Austria
| | - Lukas Lanser
- Department of Internal Medicine II, Innsbruck Medical University, Anichstraße 35, 6020, Innsbruck, Austria.,Christian Doppler Laboratory for Iron Metabolism and Anaemia Research, Innsbruck Medical University, Innsbruck, Austria
| | - Markus Seifert
- Department of Internal Medicine II, Innsbruck Medical University, Anichstraße 35, 6020, Innsbruck, Austria.,Christian Doppler Laboratory for Iron Metabolism and Anaemia Research, Innsbruck Medical University, Innsbruck, Austria
| | - Florian Kocher
- Department of Internal Medicine V, Innsbruck Medical University, Innsbruck, Austria
| | - Gerhard Pölzl
- Department of Internal Medicine III, Innsbruck Medical University, Innsbruck, Austria
| | - Günter Weiss
- Department of Internal Medicine II, Innsbruck Medical University, Anichstraße 35, 6020, Innsbruck, Austria.,Christian Doppler Laboratory for Iron Metabolism and Anaemia Research, Innsbruck Medical University, Innsbruck, Austria
| |
Collapse
|
37
|
McManus JF, Nguyen NYN, Davey RA, MacLean HE, Pomilio G, McCormack MP, Chiu WS, Wei AH, Zajac JD, Curtis DJ. Androgens stimulate erythropoiesis through the DNA-binding activity of the androgen receptor in non-hematopoietic cells. Eur J Haematol 2020; 105:247-254. [PMID: 32311143 DOI: 10.1111/ejh.13431] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2020] [Revised: 04/10/2020] [Accepted: 04/13/2020] [Indexed: 12/27/2022]
Abstract
BACKGROUND Androgens function through DNA and non-DNA binding-dependent signalling of the androgen receptor (AR). How androgens promote erythropoiesis is not fully understood. DESIGN AND METHODS To identify the androgen signalling pathway, we treated male mice lacking the second zinc finger of the DNA-binding domain of the AR (ARΔZF2 ) with non-aromatizable 5α-dihydrotestosterone (5α-DHT) or aromatizable testosterone. To distinguish direct hematopoietic and non-hematopoietic mechanisms, we performed bone marrow reconstitution experiments. RESULTS In wild-type mice, 5α-DHT had greater erythroid activity than testosterone, which can be aromatized to estradiol. The erythroid response in wild-type mice following 5α-DHT treatment was associated with increased serum erythropoietin (EPO) and its downstream target erythroferrone, and hepcidin suppression. 5α-DHT had no erythroid activity in ARΔZF2 mice, proving the importance of DNA binding by the AR. Paradoxically, testosterone, but not 5α-DHT, suppressed EPO levels in ARΔZF2 mice, suggesting testosterone following aromatization may oppose the erythroid-stimulating effects of androgens. Female wild-type mice reconstituted with ARΔZF2 bone marrow cells remained responsive to 5α-DHT. In contrast, ARΔZF2 mice reconstituted with female wild-type bone marrow cells showed no response to 5α-DHT. CONCLUSION Erythroid promoting effects of androgens are mediated through DNA binding-dependent actions of the AR in non-hematopoietic cells, including stimulating EPO expression.
Collapse
Affiliation(s)
- Julie F McManus
- Central Clinical School, Australian Centre for Blood Diseases, Monash University, Melbourne, Vic., Australia.,Human Molecular Pathology, Alfred Pathology Service, Alfred Health, Melbourne, Vic., Australia
| | - Nhu-Y N Nguyen
- Cartherics Pty Ltd, Melbourne, Vic., Australia.,Hudson Institute of Medical Research, Melbourne, Vic., Australia
| | - Rachel A Davey
- Department of Medicine, Austin Health, The University of Melbourne, Melbourne, Vic., Australia
| | - Helen E MacLean
- Department of Medicine, Austin Health, The University of Melbourne, Melbourne, Vic., Australia
| | - Giovanna Pomilio
- Central Clinical School, Australian Centre for Blood Diseases, Monash University, Melbourne, Vic., Australia.,Department of Clinical Haematology, Alfred Health, Melbourne, Vic., Australia
| | - Matthew P McCormack
- Central Clinical School, Australian Centre for Blood Diseases, Monash University, Melbourne, Vic., Australia
| | - Wan Sze Chiu
- Department of Medicine, Austin Health, The University of Melbourne, Melbourne, Vic., Australia
| | - Andrew H Wei
- Central Clinical School, Australian Centre for Blood Diseases, Monash University, Melbourne, Vic., Australia.,Department of Clinical Haematology, Alfred Health, Melbourne, Vic., Australia
| | - Jeffrey D Zajac
- Department of Medicine, Austin Health, The University of Melbourne, Melbourne, Vic., Australia
| | - David J Curtis
- Central Clinical School, Australian Centre for Blood Diseases, Monash University, Melbourne, Vic., Australia.,Department of Clinical Haematology, Alfred Health, Melbourne, Vic., Australia
| |
Collapse
|
38
|
Qi W, Wang B, Yang M, Zhu L, Hu S, Sun H. The implementation of drug reposition for alcoholic hepatitis based on a sub-pathway integration strategy. ANNALS OF TRANSLATIONAL MEDICINE 2020; 8:208. [PMID: 32309355 PMCID: PMC7154418 DOI: 10.21037/atm.2020.01.36] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Background Alcoholic hepatitis (AH) is one of the most severe forms of liver disease. The therapies which are currently available are not entirely effective, and thus novel therapies are urgently needed. However, the development of these novel therapies is limited due to incomplete information about the molecular mechanisms involved in AH. Methods The microarray data (GSE28619) was downloaded from the Gene Expression Omnibus (GEO) database. The differentially expressed genes (DEGs) between the AH and the control samples were identified using the significant analysis of microarrays (SAM) method. Metabolic sub-pathways were identified using the SubpathwayMiner R package. Cell Counting Kit-8 (CCK-8) was used to evaluate the cell viability of AML-12 cells treated with different concentrations of ethanol or riboflavin. Alanine aminotransferase (ALT) and aspartate aminotransferase (AST) were employed to show the hepatocyte function. Results A total of 1,041 genes were determined to be expressed differentially. We then identified 11 metabolic sub-pathways that could be involved in the development of AH. This was followed by a final integrated analysis of the sub-pathways involved in AH as well as the sub-pathways involved in the drug-affected cases. The final integration results led to the identification of 64 small molecular drugs. A potential novel drug (riboflavin) involved in the fatty acid metabolism pathway was identified for further investigation. Riboflavin at the 60 nM for 24 h could reverse ethanol-induced AML-12 cell injury and could markedly decrease ALT and AST activity. The decrease in the activities of these two enzymes was observed in a dose-dependent manner when it was compared to ethanol alone, which suggests that riboflavin has a protective effect against liver cell injury caused by alcoholism. Conclusions To summarize, the candidate agents which are identified in the present study might give practitioners insight into the development of novel therapeutic approaches for AH.
Collapse
Affiliation(s)
- Wei Qi
- College of Pharmacy, College of Pharmacy, Harbin Medical University, Harbin 150001, China.,Department of Inorganic Chemistry and Physical Chemistry, College of Pharmacy, Harbin Medical University, Harbin 150001, China
| | - Bing Wang
- Qingdao Women and Children Hospital, Qingdao 266011, China
| | - Ming Yang
- College of Pharmacy, College of Pharmacy, Harbin Medical University, Harbin 150001, China
| | - Lin Zhu
- The Second Affiliated Hospital of Harbin Medical University, Harbin 150001, China
| | - Sen Hu
- College of Pharmacy, College of Pharmacy, Harbin Medical University, Harbin 150001, China
| | - Hui Sun
- College of Pharmacy, College of Pharmacy, Harbin Medical University, Harbin 150001, China.,Pharmaceutical Experiment Teaching Center, College of Pharmacy, Harbin Medical University, Harbin 150001, China
| |
Collapse
|
39
|
Wu FJ, Li IH, Chien WC, Shih JH, Lin YC, Chuang CM, Cheng YD, Kao LT. Androgen deprivation therapy and the risk of iron-deficiency anaemia among patients with prostate cancer: a population-based cohort study. BMJ Open 2020; 10:e034202. [PMID: 32213519 PMCID: PMC7170598 DOI: 10.1136/bmjopen-2019-034202] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
OBJECTIVES The administration of androgen deprivation therapy (ADT) to patients with metastatic prostate cancer might be associated with some adverse effects such as anaemia; however, few studies have been performed in East Asian populations. This study aimed to investigate the association between ADT and iron-deficiency anaemia (IDA) among patients with prostate cancer in a population-based nationwide cohort. DESIGN Cohort study. SETTING Taiwan. PARTICIPANTS Data for the cohort study were retrieved from the Taiwan National Health Insurance Research Database. Propensity score matching was used to select 7262 patients with prostate cancer who received ADT as the study group and 3631 patients who did not receive ADT as the control group. PRIMARY AND SECONDARY OUTCOME MEASURES This study individually tracked patients over a 3-year study period and identified those who were subsequently diagnosed with IDA following the index date. RESULTS The incidence rates of IDA in the study and control groups were 1.66 (95% CI CI 1.45 to 1.86) and 1.01 per 100 person-years (95% CI 0.78 to 1.25), respectively. Furthermore, proportional Cox regression revealed an HR of 1.62 (95% CI 1.24 to 2.12) for IDA in the study group after adjusting for patients' age, monthly income, geographic location, residential urbanisation level and incidence of hyperlipidaemia, diabetes, hypertension, coronary heart disease, inflammatory bowel disease, other cancers and gastrointestinal bleeding. CONCLUSION Compared with its non-use among patients with prostate cancer, ADT use was associated with a higher risk of IDA.
Collapse
Affiliation(s)
- Fang-Jen Wu
- Department of Pharmacy, West Garden Hospital, Taipei, Taiwan
| | - I-Hsun Li
- Department of Pharmacy Practice, Tri-Service General Hospital, Taipei, Taiwan
- School of Pharmacy, National Defense Medical Center, Taipei, Taiwan
- Department of Pharmacology, National Defense Medical Center, Taipei, Taiwan
| | - Wu-Chien Chien
- Graduate Institute of Life Sciences, National Defense Medical Center, Taipei, Taiwan
- Department of Medical Research, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
- School of Public Health, National Defense Medical Center, Taipei, Taiwan
| | - Jui-Hu Shih
- Department of Pharmacy Practice, Tri-Service General Hospital, Taipei, Taiwan
- School of Pharmacy, National Defense Medical Center, Taipei, Taiwan
| | - Yi-Chun Lin
- Research Center of Biostatistics, Taipei Medical University, Taipei, Taiwan
| | - Chin-Min Chuang
- Emergency Department, China Medical University Hospital, Taichung City, Taiwan
| | - Yih-Dih Cheng
- Department of Pharmacy, China Medical University Hospital, Taichung City, Taiwan
| | - Li-Ting Kao
- Department of Pharmacy Practice, Tri-Service General Hospital, Taipei, Taiwan
- School of Pharmacy, National Defense Medical Center, Taipei, Taiwan
- Graduate Institute of Life Sciences, National Defense Medical Center, Taipei, Taiwan
- School of Public Health, National Defense Medical Center, Taipei, Taiwan
| |
Collapse
|
40
|
Wang CY, Xiao X, Bayer A, Xu Y, Dev S, Canali S, Nair AV, Masia R, Babitt JL. Ablation of Hepatocyte Smad1, Smad5, and Smad8 Causes Severe Tissue Iron Loading and Liver Fibrosis in Mice. Hepatology 2019; 70:1986-2002. [PMID: 31127639 PMCID: PMC6874904 DOI: 10.1002/hep.30780] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/04/2018] [Accepted: 05/17/2019] [Indexed: 12/15/2022]
Abstract
A failure of iron to appropriately regulate liver hepcidin production is central to the pathogenesis of hereditary hemochromatosis. SMAD1/5 transcription factors, activated by bone morphogenetic protein (BMP) signaling, are major regulators of hepcidin production in response to iron; however, the role of SMAD8 and the contribution of SMADs to hepcidin production by other systemic cues remain uncertain. Here, we generated hepatocyte Smad8 single (Smad8fl/fl ;Alb-Cre+ ), Smad1/5/8 triple (Smad158;Alb-Cre+ ), and littermate Smad1/5 double (Smad15;Alb-Cre+ ) knockout mice to investigate the role of SMAD8 in hepcidin and iron homeostasis regulation and liver injury. We found that Smad8;Alb-Cre+ mice exhibited no iron phenotype, whereas Smad158;Alb-Cre+ mice had greater iron overload than Smad15;Alb-Cre+ mice. In contrast to the sexual dimorphism reported for wild-type mice and other hemochromatosis models, hepcidin deficiency and extrahepatic iron loading were similarly severe in Smad15;Alb-Cre+ and Smad158;Alb-Cre+ female compared with male mice. Moreover, epidermal growth factor (EGF) failed to suppress hepcidin in Smad15;Alb-Cre+ hepatocytes. Conversely, hepcidin was still increased by lipopolysaccharide in Smad158;Alb-Cre+ mice, although lower basal hepcidin resulted in lower maximal hepcidin. Finally, unlike most mouse hemochromatosis models, Smad158;Alb-Cre+ developed liver injury and fibrosis at 8 weeks. Liver injury and fibrosis were prevented in Smad158;Alb-Cre+ mice by a low-iron diet and were minimal in iron-loaded Cre- mice. Conclusion: Hepatocyte Smad1/5/8 knockout mice are a model of hemochromatosis that encompasses liver injury and fibrosis seen in human disease. These mice reveal the redundant but critical role of SMAD8 in hepcidin and iron homeostasis regulation, establish a requirement for SMAD1/5/8 in hepcidin regulation by testosterone and EGF but not inflammation, and suggest a pathogenic role for both iron loading and SMAD1/5/8 deficiency in liver injury and fibrosis.
Collapse
Affiliation(s)
- Chia-Yu Wang
- Program in Anemia Signaling Research, Division of Nephrology, Program in Membrane Biology, Center for Systems Biology,Address correspondence to: Chia-Yu Wang, Massachusetts General Hospital, 185 Cambridge St., CPZN-8150, Boston, MA 02114, Phone: (617)-724-9078, Fax: (617)-643-3182,
| | - Xia Xiao
- Program in Anemia Signaling Research, Division of Nephrology, Program in Membrane Biology, Center for Systems Biology
| | - Abraham Bayer
- Program in Anemia Signaling Research, Division of Nephrology, Program in Membrane Biology, Center for Systems Biology
| | - Yang Xu
- Program in Anemia Signaling Research, Division of Nephrology, Program in Membrane Biology, Center for Systems Biology
| | - Som Dev
- Program in Anemia Signaling Research, Division of Nephrology, Program in Membrane Biology, Center for Systems Biology
| | - Susanna Canali
- Program in Anemia Signaling Research, Division of Nephrology, Program in Membrane Biology, Center for Systems Biology
| | - Anil V. Nair
- Program in Anemia Signaling Research, Division of Nephrology, Program in Membrane Biology, Center for Systems Biology
| | - Ricard Masia
- Department of Pathology, Massachusetts General Hospital, Harvard Medical School, Boston, MA
| | - Jodie L. Babitt
- Program in Anemia Signaling Research, Division of Nephrology, Program in Membrane Biology, Center for Systems Biology
| |
Collapse
|
41
|
Armitage AE, Agbla SC, Betts M, Sise EA, Jallow MW, Sambou E, Darboe B, Worwui A, Weinstock GM, Antonio M, Pasricha SR, Prentice AM, Drakesmith H, Darboe MK, Kwambana-Adams BA. Rapid growth is a dominant predictor of hepcidin suppression and declining ferritin in Gambian infants. Haematologica 2019; 104:1542-1553. [PMID: 30733275 PMCID: PMC6669141 DOI: 10.3324/haematol.2018.210146] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2018] [Accepted: 01/31/2019] [Indexed: 12/18/2022] Open
Abstract
Iron deficiency and iron deficiency anemia are highly prevalent in low-income countries, especially among young children. Hepcidin is the major regulator of systemic iron homeostasis. It controls dietary iron absorption, dictates whether absorbed iron is made available in circulation for erythropoiesis and other iron-demanding processes, and predicts response to oral iron supplementation. Understanding how hepcidin is itself regulated is therefore important, especially in young children. We investigated how changes in iron-related parameters, inflammation and infection status, seasonality, and growth influenced plasma hepcidin and ferritin concentrations during infancy using longitudinal data from two birth cohorts of infants in rural Gambia (n=114 and n=193). This setting is characterized by extreme seasonality, prevalent childhood anemia, undernutrition, and frequent infection. Plasma was collected from infants at birth and at regular intervals, up to 12 months of age. Hepcidin, ferritin and plasma iron concentrations declined markedly during infancy, with reciprocal increases in soluble transferrin receptor and transferrin concentrations, indicating declining iron stores and increasing tissue iron demand. In cross-sectional analyses at 5 and 12 months of age, we identified expected relationships of hepcidin with iron and inflammatory markers, but also observed significant negative associations between hepcidin and antecedent weight gain. Correspondingly, longitudinal fixed effects modeling demonstrated weight gain to be the most notable dynamic predictor of decreasing hepcidin and ferritin through infancy across both cohorts. Infants who grow rapidly in this setting are at particular risk of depletion of iron stores, but since hepcidin concentrations decrease with weight gain, they may also be the most responsive to oral iron interventions.
Collapse
Affiliation(s)
- Andrew E Armitage
- MRC Human Immunology Unit, MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, UK
| | - Schadrac C Agbla
- Faculty of Epidemiology and Population Health, London School of Hygiene and Tropical Medicine, London, UK
| | - Modupeh Betts
- MRC Unit The Gambia at the London School of Hygiene and Tropical Medicine, Banjul, The Gambia, Africa
| | - Ebrima A Sise
- MRC Unit The Gambia at the London School of Hygiene and Tropical Medicine, Banjul, The Gambia, Africa
| | - Momodou W Jallow
- MRC Unit The Gambia at the London School of Hygiene and Tropical Medicine, Banjul, The Gambia, Africa
| | - Ellen Sambou
- WHO Collaborating Center for New Vaccines Surveillance, MRC Unit The Gambia at the London School of Hygiene and Tropical Medicine, Banjul, The Gambia, Africa
| | - Bakary Darboe
- MRC Unit The Gambia at the London School of Hygiene and Tropical Medicine, Banjul, The Gambia, Africa
| | - Archibald Worwui
- MRC Unit The Gambia at the London School of Hygiene and Tropical Medicine, Banjul, The Gambia, Africa
| | | | - Martin Antonio
- WHO Collaborating Center for New Vaccines Surveillance, MRC Unit The Gambia at the London School of Hygiene and Tropical Medicine, Banjul, The Gambia, Africa
| | - Sant-Rayn Pasricha
- MRC Human Immunology Unit, MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, UK
- Walter and Eliza Hall Institute for Medical Research, Melbourne, VIC, Australia
- Department of Medical Biology, The University of Melbourne, VIC, Melbourne, Australia
| | - Andrew M Prentice
- MRC Unit The Gambia at the London School of Hygiene and Tropical Medicine, Banjul, The Gambia, Africa
| | - Hal Drakesmith
- MRC Human Immunology Unit, MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, UK
- Haematology Theme, Oxford Biomedical Research Centre, Oxford, UK
| | - Momodou K Darboe
- MRC Unit The Gambia at the London School of Hygiene and Tropical Medicine, Banjul, The Gambia, Africa
| | - Brenda Anna Kwambana-Adams
- WHO Collaborating Center for New Vaccines Surveillance, MRC Unit The Gambia at the London School of Hygiene and Tropical Medicine, Banjul, The Gambia, Africa
- NIHR Global Health Research Unit on Mucosal Pathogens, Division of Infection and Immunity, University College London, London, UK
| |
Collapse
|
42
|
Cavey T, Latour C, Island ML, Leroyer P, Guggenbuhl P, Coppin H, Roth MP, Bendavid C, Brissot P, Ropert M, Loréal O. Spleen iron, molybdenum, and manganese concentrations are coregulated in hepcidin‐deficient and secondary iron overload models in mice. FASEB J 2019; 33:11072-11081. [DOI: 10.1096/fj.201801381rr] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Affiliation(s)
- Thibault Cavey
- INSERMUniversité RennesINRAUMR 1241Analyse Elémentaire et Métabolisme des Métaux (AEM2) PlatformCHU Pontchaillou, NutritionMétabolismes et Cancer Institute (NuMeCan)RennesFrance
| | - Chloé Latour
- Institut de Recherche en Santé Digestive (IRSD)INSERMINRAÉcole Nationale Vétérinaire de Toulouse (ENVT)Université Paul Sabatier (UPS)-Université de ToulouseToulouseFrance
| | - Marie-Laure Island
- INSERMUniversité RennesINRAUMR 1241Analyse Elémentaire et Métabolisme des Métaux (AEM2) PlatformCHU Pontchaillou, NutritionMétabolismes et Cancer Institute (NuMeCan)RennesFrance
| | - Patricia Leroyer
- INSERMUniversité RennesINRAUMR 1241Analyse Elémentaire et Métabolisme des Métaux (AEM2) PlatformCHU Pontchaillou, NutritionMétabolismes et Cancer Institute (NuMeCan)RennesFrance
| | - Pascal Guggenbuhl
- INSERMUniversité RennesINRAUMR 1241Analyse Elémentaire et Métabolisme des Métaux (AEM2) PlatformCHU Pontchaillou, NutritionMétabolismes et Cancer Institute (NuMeCan)RennesFrance
| | - Hélène Coppin
- Institut de Recherche en Santé Digestive (IRSD)INSERMINRAÉcole Nationale Vétérinaire de Toulouse (ENVT)Université Paul Sabatier (UPS)-Université de ToulouseToulouseFrance
| | - Marie-Paule Roth
- Institut de Recherche en Santé Digestive (IRSD)INSERMINRAÉcole Nationale Vétérinaire de Toulouse (ENVT)Université Paul Sabatier (UPS)-Université de ToulouseToulouseFrance
| | - Claude Bendavid
- INSERMUniversité RennesINRAUMR 1241Analyse Elémentaire et Métabolisme des Métaux (AEM2) PlatformCHU Pontchaillou, NutritionMétabolismes et Cancer Institute (NuMeCan)RennesFrance
| | - Pierre Brissot
- INSERMUniversité RennesINRAUMR 1241Analyse Elémentaire et Métabolisme des Métaux (AEM2) PlatformCHU Pontchaillou, NutritionMétabolismes et Cancer Institute (NuMeCan)RennesFrance
| | - Martine Ropert
- INSERMUniversité RennesINRAUMR 1241Analyse Elémentaire et Métabolisme des Métaux (AEM2) PlatformCHU Pontchaillou, NutritionMétabolismes et Cancer Institute (NuMeCan)RennesFrance
| | - Olivier Loréal
- INSERMUniversité RennesINRAUMR 1241Analyse Elémentaire et Métabolisme des Métaux (AEM2) PlatformCHU Pontchaillou, NutritionMétabolismes et Cancer Institute (NuMeCan)RennesFrance
| |
Collapse
|
43
|
Iron considerations for the athlete: a narrative review. Eur J Appl Physiol 2019; 119:1463-1478. [PMID: 31055680 DOI: 10.1007/s00421-019-04157-y] [Citation(s) in RCA: 151] [Impact Index Per Article: 25.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2019] [Accepted: 05/02/2019] [Indexed: 02/07/2023]
Abstract
Iron plays a significant role in the body, and is specifically important to athletes, since it is a dominant feature in processes such as oxygen transport and energy metabolism. Despite its importance, athlete populations, especially females and endurance athletes, are commonly diagnosed with iron deficiency, suggesting an association between sport performance and iron regulation. Although iron deficiency is most common in female athletes (~ 15-35% athlete cohorts deficient), approximately 5-11% of male athlete cohorts also present with this issue. Furthermore, interest has grown in the mechanisms that influence iron absorption in athletes over the last decade, with the link between iron regulation and exercise becoming a research focus. Specifically, exercise-induced increases in the master iron regulatory hormone, hepcidin, has been highlighted as a contributing factor towards altered iron metabolism in athletes. To date, a plethora of research has been conducted, including investigation into the impact that sex hormones, diet (e.g. macronutrient manipulation), training and environmental stress (e.g. hypoxia due to altitude training) have on an athlete's iron status, with numerous recommendations proposed for consideration. This review summarises the current state of research with respect to the aforementioned factors, drawing conclusions and recommendations for future work.
Collapse
|
44
|
Guo W, Schmidt PJ, Fleming MD, Bhasin S. Hepcidin is not essential for mediating testosterone's effects on erythropoiesis. Andrology 2019; 8:82-90. [DOI: 10.1111/andr.12622] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2019] [Revised: 03/06/2019] [Accepted: 03/07/2019] [Indexed: 12/21/2022]
Affiliation(s)
- W. Guo
- Research Program in Men's Health: Aging and Metabolism The Boston Claude D. Pepper Older Americans Independence Center Brigham and Women's Hospital, and Harvard Medical School Boston MA USA
| | - P. J. Schmidt
- Department of Pathology Boston Children's Hospital and Harvard Medical School Boston MA USA
| | - M. D. Fleming
- Department of Pathology Boston Children's Hospital and Harvard Medical School Boston MA USA
| | - S. Bhasin
- Research Program in Men's Health: Aging and Metabolism The Boston Claude D. Pepper Older Americans Independence Center Brigham and Women's Hospital, and Harvard Medical School Boston MA USA
| |
Collapse
|
45
|
Abstract
Hepcidin, the main regulator of iron metabolism, is synthesized and released by hepatocytes in response to increased body iron concentration and inflammation. Deregulation of hepcidin expression is a common feature of genetic and acquired iron disorders: in Hereditary Hemochromatosis (HH) and iron-loading anemias low hepcidin causes iron overload, while in Iron Refractory Iron Deficiency Anemia (IRIDA) and anemia of inflammation (AI), high hepcidin levels induce iron-restricted erythropoiesis. Hepcidin expression in the liver is mainly controlled by the BMP-SMAD pathway, activated in a paracrine manner by BMP2 and BMP6 produced by liver sinusoidal endothelial cells. The BMP type I receptors ALK2 and ALK3 are responsible for iron-dependent hepcidin upregulation and basal hepcidin expression, respectively. Characterization of animal models with genetic inactivation of the key components of the pathway has suggested the existence of two BMP/SMAD pathway branches: the first ALK3 and HH proteins dependent, responsive to BMP2 for basal hepcidin activation, and the second ALK2 dependent, activated by BMP6 in response to increased tissue iron. The erythroid inhibitor of hepcidin Erythroferrone also impacts on the liver BMP-SMAD pathway although its effect is blunted by pathway hyper-activation. The liver BMP-SMAD pathway is required also in inflammation to cooperate with JAK2/STAT3 signaling for full hepcidin activation. Pharmacologic targeting of BMP-SMAD pathway components or regulators may improve the outcome of both genetic and acquired disorders of iron overload and deficiency by increasing or inhibiting hepcidin expression.
Collapse
|
46
|
Abstract
Iron, an essential nutrient, is required for many biological processes but is also toxic in excess. The lack of a mechanism to excrete excess iron makes it crucial for the body to regulate the amount of iron absorbed from the diet. This regulation is mediated by the hepatic hormone hepcidin. Hepcidin also controls iron release from macrophages that recycle iron and from hepatocytes that store iron. Hepcidin binds to the only known iron export protein, ferroportin, inducing its internalization and degradation and thus limiting the amount of iron released into the plasma. Important regulators of hepcidin, and therefore of systemic iron homeostasis, include plasma iron concentrations, body iron stores, infection and inflammation, hypoxia and erythropoiesis, and, to a lesser extent, testosterone. Dysregulation of hepcidin production contributes to the pathogenesis of many iron disorders: hepcidin deficiency causes iron overload in hereditary hemochromatosis and non-transfused β-thalassemia, whereas overproduction of hepcidin is associated with iron-restricted anemias seen in patients with chronic inflammatory diseases and inherited iron-refractory iron-deficiency anemia. The present review summarizes our current understanding of the molecular mechanisms and signaling pathways contributing to hepcidin regulation by these factors and highlights the issues that still need clarification.
Collapse
Affiliation(s)
- Marie-Paule Roth
- Institut de Recherche en Santé Digestive (IRSD), Université de Toulouse, INSERM, INRA, ENVT, UPS, Toulouse, France.
| | - Delphine Meynard
- Institut de Recherche en Santé Digestive (IRSD), Université de Toulouse, INSERM, INRA, ENVT, UPS, Toulouse, France
| | - Hélène Coppin
- Institut de Recherche en Santé Digestive (IRSD), Université de Toulouse, INSERM, INRA, ENVT, UPS, Toulouse, France
| |
Collapse
|
47
|
Traeger L, Ellermann I, Wiethoff H, Ihbe J, Gallitz I, Eveslage M, Moritz R, Herrmann E, Schrader AJ, Steinbicker AU. Serum Hepcidin and GDF-15 levels as prognostic markers in urothelial carcinoma of the upper urinary tract and renal cell carcinoma. BMC Cancer 2019; 19:74. [PMID: 30646851 PMCID: PMC6334404 DOI: 10.1186/s12885-019-5278-0] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2018] [Accepted: 01/04/2019] [Indexed: 01/07/2023] Open
Abstract
Background Cancer is a life-threatening disease that causes every fourth death. It is often hard to determine the time point of progression. Therefore, biomarkers for cancer entities that indicate disease progression or aggressiveness and thereby guide therapeutic decisions are required. Unfortunately, reliable biomarkers are rare. In this study, the potential of serum hepcidin and serum GDF-15 as biomarkers that correlate with patient’s survival in the two entities upper urinary tract urothelial carcinomas (UUTUC) and renal cell carcinoma (RCC) were analyzed. Methods In this retrospective study n = 38 patients suffering from UUTUC, n = 94 patients suffering from RCC and n = 21 patients without infections or cancer, all hospitalized at the University Hospital Muenster, were included. Serum samples of patients were retrospectively analyzed. Serum hepcidin and GDF-15 levels were measured and correlated to aggressiveness and progression of the disease as well as patient’s outcome. Results For both entities, UUTUC and RCC, serum hepcidin levels as well as serum GDF-15 levels were increased compared to sera of controls. High serum hepcidin and GDF-15 levels were associated with metastases and cancer relapse. Also, in both entities, the overall survival was decreased in patients with increased serum hepcidin and GDF-15 levels. Hence, high serum hepcidin and GDF-15 levels correlated with patient’s outcome. Conclusion To conclude, the data of this study show a correlation of high serum hepcidin and GDF-15 levels with aggressiveness and progression of the disease and demonstrate potential prognostic properties of serum hepcidin and GDF-15 levels. The data support the further assessment of serum hepcidin and GDF-15 as prognostic markers in RCC and UUTUC.
Collapse
Affiliation(s)
- Lisa Traeger
- Department of Anesthesiology, Intensive Care and Pain Medicine, University Hospital Muenster, University of Muenster, Albert-Schweitzer Campus 1, Building A1, 48149, Muenster, Germany
| | - Ines Ellermann
- Department of Pharmacy, University Hospital Muenster, University of Muenster, Muenster, Germany
| | - Helene Wiethoff
- Department of Anesthesiology, Intensive Care and Pain Medicine, University Hospital Muenster, University of Muenster, Albert-Schweitzer Campus 1, Building A1, 48149, Muenster, Germany
| | - Janina Ihbe
- Department of Anesthesiology, Intensive Care and Pain Medicine, University Hospital Muenster, University of Muenster, Albert-Schweitzer Campus 1, Building A1, 48149, Muenster, Germany
| | - Inka Gallitz
- Department of Anesthesiology, Intensive Care and Pain Medicine, University Hospital Muenster, University of Muenster, Albert-Schweitzer Campus 1, Building A1, 48149, Muenster, Germany
| | - Maria Eveslage
- Institute of Biostatistics and Clinical Research, University Hospital Muenster, University of Muenster, Münster, Germany
| | - Rudolf Moritz
- Department of Urology, University Hospital Muenster, University of Muenster, Muenster, Germany.,Present Address: Department of Urology, St. Antonius Hospital, Gronau, Germany
| | - Edwin Herrmann
- Department of Urology, University Hospital Muenster, University of Muenster, Muenster, Germany.,Present Address: Department of Urology, Prosper Hospital, Recklinghausen, Germany
| | - Andres Jan Schrader
- Department of Urology, University Hospital Muenster, University of Muenster, Muenster, Germany
| | - Andrea U Steinbicker
- Department of Anesthesiology, Intensive Care and Pain Medicine, University Hospital Muenster, University of Muenster, Albert-Schweitzer Campus 1, Building A1, 48149, Muenster, Germany.
| |
Collapse
|
48
|
Song M, Vos MB, McClain CJ. Copper-Fructose Interactions: A Novel Mechanism in the Pathogenesis of NAFLD. Nutrients 2018; 10:E1815. [PMID: 30469339 PMCID: PMC6266129 DOI: 10.3390/nu10111815] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2018] [Revised: 11/08/2018] [Accepted: 11/16/2018] [Indexed: 12/13/2022] Open
Abstract
Compelling epidemiologic data support the critical role of dietary fructose in the epidemic of obesity, metabolic syndrome and nonalcoholic fatty liver disease (NAFLD). The metabolic effects of fructose on the development of metabolic syndrome and NAFLD are not completely understood. High fructose intake impairs copper status, and copper-fructose interactions have been well documented in rats. Altered copper-fructose metabolism leads to exacerbated experimental metabolic syndrome and NAFLD. A growing body of evidence has demonstrated that copper levels are low in NAFLD patients. Moreover, hepatic and serum copper levels are inversely correlated with the severity of NAFLD. Thus, high fructose consumption and low copper availability are considered two important risk factors in NAFLD. However, the causal effect of copper-fructose interactions as well as the effects of fructose intake on copper status remain to be evaluated in humans. The aim of this review is to summarize the role of copper-fructose interactions in the pathogenesis of the metabolic syndrome and discuss the potential underlying mechanisms. This review will shed light on the role of copper homeostasis and high fructose intake and point to copper-fructose interactions as novel mechanisms in the fructose induced NAFLD.
Collapse
Affiliation(s)
- Ming Song
- Department of Medicine, Division of Gastroenterology, Hepatology and Nutrition, University of Louisville School of Medicine, Louisville, KY 40202, USA.
- Hepatobiology&Toxicology Center, University of Louisville School of Medicine, Louisville, KY 40202, USA.
| | - Miriam B Vos
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA 30307, USA.
- Children's Healthcare of Atlanta, Atlanta, GA 30322, USA.
| | - Craig J McClain
- Department of Medicine, Division of Gastroenterology, Hepatology and Nutrition, University of Louisville School of Medicine, Louisville, KY 40202, USA.
- Hepatobiology&Toxicology Center, University of Louisville School of Medicine, Louisville, KY 40202, USA.
- Department of Pharmacology and Toxicology, University of Louisville School of Medicine, Louisville, KY 40202, USA.
- University of Louisville Alcohol Research Center, University of Louisville School of Medicine, Louisville, KY 40202, USA.
- Robley Rex Veterans Affairs Medical Center, Louisville, KY 40206, USA.
| |
Collapse
|
49
|
Hepcidin Therapeutics. Pharmaceuticals (Basel) 2018; 11:ph11040127. [PMID: 30469435 PMCID: PMC6316648 DOI: 10.3390/ph11040127] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2018] [Revised: 11/15/2018] [Accepted: 11/19/2018] [Indexed: 12/12/2022] Open
Abstract
Hepcidin is a key hormonal regulator of systemic iron homeostasis and its expression is induced by iron or inflammatory stimuli. Genetic defects in iron signaling to hepcidin lead to “hepcidinopathies” ranging from hereditary hemochromatosis to iron-refractory iron deficiency anemia, which are disorders caused by hepcidin deficiency or excess, respectively. Moreover, dysregulation of hepcidin is a pathogenic cofactor in iron-loading anemias with ineffective erythropoiesis and in anemia of inflammation. Experiments with preclinical animal models provided evidence that restoration of appropriate hepcidin levels can be used for the treatment of these conditions. This fueled the rapidly growing field of hepcidin therapeutics. Several hepcidin agonists and antagonists, as well as inducers and inhibitors of hepcidin expression have been identified to date. Some of them were further developed and are currently being evaluated in clinical trials. This review summarizes the state of the art.
Collapse
|
50
|
Camaschella C, Pagani A. Advances in understanding iron metabolism and its crosstalk with erythropoiesis. Br J Haematol 2018; 182:481-494. [PMID: 29938779 DOI: 10.1111/bjh.15403] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Recent years have witnessed impressive advances in our understanding of iron metabolism. A number of studies of iron disorders and of their animal models have provided landmark insights into the mechanisms of iron trafficking, distribution and homeostatic regulation, the latter essential to prevent both iron deficiency and iron excess. Our perception of iron metabolism has been completely changed by an improved definition of cellular and systemic iron homeostasis, of the molecular pathogenesis of iron disorders, the fine tuning of the iron hormone hepcidin by activators and inhibitors and the dissection of the components of the hepcidin regulatory pathway. Important for haematology, the crosstalk of erythropoiesis, the most important iron consumer, and the hepcidin pathway has been at least partially clarified. Novel potential biomarkers are available and novel therapeutic targets for iron-related disorders have been tested in murine models. These preclinical studies provided proofs of principle and are laying the ground for clinical trials. Understanding iron control in tissues other than erythropoiesis remains a challenge for the future.
Collapse
Affiliation(s)
- Clara Camaschella
- Division of Genetics and Cell Biology, San Raffaele Scientific Institute and Vita Salute University, Milano, Italy
| | - Alessia Pagani
- Division of Genetics and Cell Biology, San Raffaele Scientific Institute and Vita Salute University, Milano, Italy
| |
Collapse
|