151
|
Rizzo C, Carbonara R, Ruggieri R, Passantino A, Scrutinio D. Iron Deficiency: A New Target for Patients With Heart Failure. Front Cardiovasc Med 2021; 8:709872. [PMID: 34447793 PMCID: PMC8383833 DOI: 10.3389/fcvm.2021.709872] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Accepted: 07/19/2021] [Indexed: 12/28/2022] Open
Abstract
Iron deficiency (ID) is one of the most frequent comorbidities in patients with heart failure (HF). ID is estimated to be present in up to 50% of outpatients and is a strong independent predictor of HF outcomes. ID has been shown to reduce quality of life, exercise capacity and survival, in both the presence and absence of anemia. The most recent 2016 guidelines recommend starting replacement treatment at ferritin cutoff value <100 mcg/l or between 100 and 299 mcg/l when the transferrin saturation is <20%. Beyond its effect on hemoglobin, iron plays an important role in oxygen transport and in the metabolism of cardiac and skeletal muscles. Mitochondria are the most important sites of iron utilization and energy production. These factors clearly have roles in the diminished exercise capacity in HF. Oral iron administration is usually the first route used for iron repletion in patients. However, the data from the IRONOUT HF study do not support the use of oral iron supplementation in patients with HF and a reduced ejection fraction, because this treatment does not affect peak VO2 (the primary endpoint of the study) or increase serum ferritin levels. The FAIR-HF and CONFIRM-HF studies have shown improvements in symptoms, quality of life and functional capacity in patients with stable, symptomatic, iron-deficient HF after the administration of intravenous iron (i.e., FCM). Moreover, they have shown a decreased risk of first hospitalization for worsening of HF, as later confirmed in a subsequent meta-analysis. In addition, the EFFECT-HF study has shown an improvement in peak oxygen consumption at CPET (a parameter generally considered the gold standard of exercise capacity and a predictor of outcome in HF) in patients randomized to receive ferric carboxymaltose. Finally, the AFFIRM AHF trial evaluating the effects of FCM administration on the outcomes of patients hospitalized for acute HF has found significantly fewer hospital readmissions due to HF among patients treated with FCM rather than placebo.
Collapse
Affiliation(s)
- Caterina Rizzo
- Department of Cardiology, Istituti Clinici Scientifici Maugeri, IRCCS, Bari, Italy
| | - Rosa Carbonara
- Department of Cardiology, Istituti Clinici Scientifici Maugeri, IRCCS, Bari, Italy
| | - Roberta Ruggieri
- Department of Cardiology, Istituti Clinici Scientifici Maugeri, IRCCS, Bari, Italy
| | - Andrea Passantino
- Department of Cardiology, Istituti Clinici Scientifici Maugeri, IRCCS, Bari, Italy
| | - Domenico Scrutinio
- Department of Cardiology, Istituti Clinici Scientifici Maugeri, IRCCS, Bari, Italy
| |
Collapse
|
152
|
Petrillo S, Manco M, Altruda F, Fagoonee S, Tolosano E. Liver Sinusoidal Endothelial Cells at the Crossroad of Iron Overload and Liver Fibrosis. Antioxid Redox Signal 2021; 35:474-486. [PMID: 32689808 DOI: 10.1089/ars.2020.8168] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Significance: Liver fibrosis results from different etiologies and represents one of the most serious health issues worldwide. Fibrosis is the outcome of chronic insults on the liver and is associated with several factors, including abnormal iron metabolism. Recent Advances: Multiple mechanisms underlying the profibrogenic role of iron have been proposed. The pivotal role of liver sinusoidal endothelial cells (LSECs) in iron-level regulation, as well as their morphological and molecular dedifferentiation occurring in liver fibrosis, has encouraged research on LSECs as prime regulators of very early fibrotic events. Importantly, normal differentiated LSECs may act as gatekeepers of fibrogenesis by maintaining the quiescence of hepatic stellate cells, while LSECs capillarization precedes the onset of liver fibrosis. Critical Issues: In the present review, the morphological and molecular alterations occurring in LSECs after liver injury are addressed in an attempt to highlight how vascular dysfunction promotes fibrogenesis. In particular, we discuss in depth how a vicious loop can be established in which iron dysregulation and LSEC dedifferentiation synergize to exacerbate and promote the progression of liver fibrosis. Future Directions: LSECs, due to their pivotal role in early liver fibrosis and iron homeostasis, show great promises as a therapeutic target. In particular, new strategies can be devised for restoring LSECs differentiation and thus their role as regulators of iron homeostasis, hence preventing the progression of liver fibrosis or, even better, promoting its regression. Antioxid. Redox Signal. 35, 474-486.
Collapse
Affiliation(s)
- Sara Petrillo
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Torino, Torino, Italy
| | - Marta Manco
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Torino, Torino, Italy
| | - Fiorella Altruda
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Torino, Torino, Italy
| | - Sharmila Fagoonee
- Institute of Biostructure and Bioimaging, CNR c/o Molecular Biotechnology Center, Torino, Italy
| | - Emanuela Tolosano
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Torino, Torino, Italy
| |
Collapse
|
153
|
Karami H, Khalilzadeh Arjmandi H, Salehifar E, Darvishi-Khezri H, Dabirian M, Kosaryan M, Aliasgharian A, Akbarzadeh R, Naeimayi Aali R, Nasirzadeh A. A double-blind, controlled, crossover trial of amlodipine on iron overload status in transfusion dependent β-thalassemia patients. Int J Clin Pract 2021; 75:e14337. [PMID: 33969592 DOI: 10.1111/ijcp.14337] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/20/2021] [Accepted: 05/04/2021] [Indexed: 11/30/2022] Open
Abstract
BACKGROUND AND AIM This study examined whether administration of amlodipine could improve myocardial iron loading status in patients with transfusion dependent β-thalassemia (TDT), through a placebo-controlled, crossover study. METHODS Amlodipine (5 mg, daily) or placebo were prescribed to all patients (n = 19) for 6 months, and after a 2-week washout period, patients were crossed over to the other group. The efficacy of amlodipine on iron loading was assessed by measuring myocardial T2*-weighted magnetic resonance imaging (MRI T2*, millisecond [ms]) and serum ferritin (ng/mL). RESULTS Seventeen patients completed the study. The mean ± standard deviation [SD] of myocardial MRI T2* at baseline was 9.83 ± 2.67 ms Myocardial MRI T2* value rose to 11.44 ± 4.14 ms post amlodipine treatment in all patients. After placebo, myocardial MRI T2* value reached 10.29 ± 4.01 ms After controlling the baseline measures, Hedges's g for ferritin and myocardial MRI T2* outcomes were estimated 3.84 (95% confidence interval [CI] 2.68 to 4.97) and -1.80 (95% CI -2.58 to -0.10), respectively. CONCLUSION Amlodipine might improve myocardial MRI T2* and serum ferritin level compared to placebo. However, larger clinical studies are needed to confirm the results.
Collapse
Affiliation(s)
- Hossein Karami
- Thalassemia Research Center (TRC), Hemoglobinopathy Institute, Mazandaran University of Medical Sciences, Sari, Iran
| | - Hadiseh Khalilzadeh Arjmandi
- Student Research Committee, Phamaceutical Science Research Center, Hemoglobinopathy Institute, Mazandaran University of Medical Sciences, Sari, Iran
| | - Ebrahim Salehifar
- Phamaceutical Science Research Center, Hemoglobinopathy Institute, Mazandaran University of Medical Sciences, Sari, Iran
| | - Hadi Darvishi-Khezri
- Thalassemia Research Center (TRC), Hemoglobinopathy Institute, Mazandaran University of Medical Sciences, Sari, Iran
| | - Mojdeh Dabirian
- Department of Cardiology, Cardiovascular Research Center, Mazandaran University of Medical Sciences, Sari, Iran
| | - Mehrnoush Kosaryan
- Thalassemia Research Center (TRC), Hemoglobinopathy Institute, Mazandaran University of Medical Sciences, Sari, Iran
| | - Aily Aliasgharian
- Medical Microbiology, Thalassemia Research Center (TRC), Hemoglobinopathy Institute, Mazandaran University of Medical Sciences, Sari, Iran
| | - Rosetta Akbarzadeh
- Thalassemia Research Center (TRC), Hemoglobinopathy Institute, Mazandaran University of Medical Sciences, Sari, Iran
| | | | - Amirreza Nasirzadeh
- Student Research Committee, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
154
|
Girelli D, Marchi G, Busti F, Vianello A. Iron metabolism in infections: Focus on COVID-19. Semin Hematol 2021; 58:182-187. [PMID: 34389110 PMCID: PMC8305218 DOI: 10.1053/j.seminhematol.2021.07.001] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Accepted: 07/12/2021] [Indexed: 12/15/2022]
Abstract
Iron is a micronutrient essential for a wide range of metabolic processes in virtually all living organisms. During infections, a battle for iron takes place between the human host and the invading pathogens. The liver peptide hepcidin, which is phylogenetically and structurally linked to defensins (antimicrobial peptides of the innate immunity), plays a pivotal role by subtracting iron to pathogens through its sequestration into host cells, mainly macrophages. While this phenomenon is well studied in certain bacterial infections, much less is known regarding viral infections. Iron metabolism also has implications on the functionality of cells of the immune system. Once primed by the contact with antigen presenting cells, lymphocytes need iron to sustain the metabolic burst required for mounting an effective cellular and humoral response. The COVID-19 pandemic has boosted an amount of clinical and translational research over the possible influences of nutrients on SARS-CoV-2 infection, in terms of either susceptibility or clinical course. Here we review the intersections between iron metabolism and COVID-19, belonging to the wider domain of the so-called “nutritional immunity”. A better understanding of such connections has potential broad implications, either from a mechanistic standpoint, or for the development of more effective strategies for managing COVID-19 and possible future pandemics.
Collapse
Affiliation(s)
- Domenico Girelli
- Department of Medicine, Section of Internal Medicine, University of Verona, Euro Blood Net Referral Center, Azienda Ospedaliera Universitaria Integrata Verona, Italy.
| | - Giacomo Marchi
- Department of Medicine, Section of Internal Medicine, University of Verona, Euro Blood Net Referral Center, Azienda Ospedaliera Universitaria Integrata Verona, Italy
| | - Fabiana Busti
- Department of Medicine, Section of Internal Medicine, University of Verona, Euro Blood Net Referral Center, Azienda Ospedaliera Universitaria Integrata Verona, Italy
| | - Alice Vianello
- Department of Medicine, Section of Internal Medicine, University of Verona, Euro Blood Net Referral Center, Azienda Ospedaliera Universitaria Integrata Verona, Italy
| |
Collapse
|
155
|
Grigorieva KN, Bitsadze VO, Khizroeva JK, Tretyakova MV, Blinov DV, Tsibizova VI, Ponomarev DA, Shkoda AS, Orudzhova EA, Grandone E, Rizzo G, Makatsariya AD. Macrophage activation syndrome in COVID-19. OBSTETRICS, GYNECOLOGY AND REPRODUCTION 2021; 15:313-320. [DOI: 10.17749/2313-7347/ob.gyn.rep.2021.217] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
The novel coronavirus epidemic is characterized by high rates of morbidity and relatively high mortality. Laboratory test results in patients include leukopenia, an increase in liver function tests and ferritin levels reaching hundreds, and sometimes thousands of units. These data remind us about the macrophage activation syndrome (MAC). Secondary hemophagocytic lymphohistiocytosis syndrome, MAC, which pathogenesis is based on a defect in the mechanisms of T-cell cytotoxicity and decreased level of natural killer cells associated with the defect in the perforin-encoding gene as well as hyperproduction of a number of cytokines – interleukin (IL)-1â, tumor necrosis factor-á, etc. by T-lymphocytes and histiocytes, indirectly leading to the activation of macrophages and production of proinflammatory cytokines, in particular IL-6 hyperproduction. MAC is one of "hyperferritinemic syndromes". These disorders have similar clinical and laboratory manifestations, and they also respond to similar treatments, suggesting that hyperferritinemia may be involved in the overall pathogenesis and is characterized by elevated ferritin level and cytokine storm. Despite the fact that data on the immune and inflammatory status in patients with COVID-19 have only started to appear, it is already clear that hyperinflammation and coagulopathy affect the disease severity and increase the risk of death in patients infected with SARS-CoV-2. Hence, understanding the pathogenesis of the novel coronavirus infection can help in its early diagnostics and treatment.
Collapse
Affiliation(s)
| | | | | | | | - D. V. Blinov
- Institute for Preventive and Social Medicine; Lapino Clinic Hospital, MD Medical Group
| | - V. I. Tsibizova
- Almazov National Medical Research Centre, Health Ministry of Russian Federation
| | - D. A. Ponomarev
- Vorokhobov City Clinical Hospital № 67, Moscow Healthcare Department
| | - A. S. Shkoda
- Vorokhobov City Clinical Hospital № 67, Moscow Healthcare Department
| | - E. A. Orudzhova
- Vorokhobov City Clinical Hospital № 67, Moscow Healthcare Department
| | - E. Grandone
- Sechenov University; Tor Vergata University of Rome
| | - G. Rizzo
- Sechenov University; Tor Vergata University of Rome
| | | |
Collapse
|
156
|
Haschka D, Tymoszuk P, Petzer V, Hilbe R, Heeke S, Dichtl S, Skvortsov S, Demetz E, Berger S, Seifert M, Mitterstiller AM, Moser P, Bumann D, Nairz M, Theurl I, Weiss G. Ferritin H deficiency deteriorates cellular iron handling and worsens Salmonella typhimurium infection by triggering hyperinflammation. JCI Insight 2021; 6:e141760. [PMID: 34236052 PMCID: PMC8410025 DOI: 10.1172/jci.insight.141760] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2020] [Accepted: 05/26/2021] [Indexed: 12/30/2022] Open
Abstract
Iron is an essential nutrient for mammals as well as for pathogens. Inflammation-driven changes in systemic and cellular iron homeostasis are central for host-mediated antimicrobial strategies. Here, we studied the role of the iron storage protein ferritin H (FTH) for the control of infections with the intracellular pathogen Salmonella enterica serovar Typhimurium by macrophages. Mice lacking FTH in the myeloid lineage (LysM-Cre+/+Fthfl/fl mice) displayed impaired iron storage capacities in the tissue leukocyte compartment, increased levels of labile iron in macrophages, and an accelerated macrophage-mediated iron turnover. While under steady-state conditions, LysM-Cre+/+Fth+/+ and LysM-Cre+/+Fthfl/fl animals showed comparable susceptibility to Salmonella infection, i.v. iron supplementation drastically shortened survival of LysM-Cre+/+Fthfl/fl mice. Mechanistically, these animals displayed increased bacterial burden, which contributed to uncontrolled triggering of NF-κB and inflammasome signaling and development of cytokine storm and death. Importantly, pharmacologic inhibition of the inflammasome and IL-1β pathways reduced cytokine levels and mortality and partly restored infection control in iron-treated ferritin-deficient mice. These findings uncover incompletely characterized roles of ferritin and cellular iron turnover in myeloid cells in controlling bacterial spread and for modulating NF-κB and inflammasome-mediated cytokine activation, which may be of vital importance in iron-overloaded individuals suffering from severe infections and sepsis.
Collapse
Affiliation(s)
- David Haschka
- Department of Internal Medicine II, Medical University of Innsbruck, Innsbruck, Austria
| | - Piotr Tymoszuk
- Department of Internal Medicine II, Medical University of Innsbruck, Innsbruck, Austria
| | - Verena Petzer
- Department of Internal Medicine II, Medical University of Innsbruck, Innsbruck, Austria
| | - Richard Hilbe
- Department of Internal Medicine II, Medical University of Innsbruck, Innsbruck, Austria
| | - Simon Heeke
- Department of Internal Medicine II, Medical University of Innsbruck, Innsbruck, Austria
| | - Stefanie Dichtl
- Department of Internal Medicine II, Medical University of Innsbruck, Innsbruck, Austria
| | - Sergej Skvortsov
- Department of Therapeutic Radiology and Oncology, Laboratory for Experimental and Translational Research on Radiation Oncology, Tyrolean Cancer Research Institute, Medical University of Innsbruck, Innsbruck, Austria
| | - Egon Demetz
- Department of Internal Medicine II, Medical University of Innsbruck, Innsbruck, Austria
| | - Sylvia Berger
- Department of Internal Medicine II, Medical University of Innsbruck, Innsbruck, Austria
| | - Markus Seifert
- Department of Internal Medicine II, Medical University of Innsbruck, Innsbruck, Austria
| | | | | | - Dirk Bumann
- Biozentrum, University of Basel, Klingelbergstrasse, Basel, Switzerland
| | - Manfred Nairz
- Department of Internal Medicine II, Medical University of Innsbruck, Innsbruck, Austria
| | - Igor Theurl
- Department of Internal Medicine II, Medical University of Innsbruck, Innsbruck, Austria
| | - Guenter Weiss
- Department of Internal Medicine II, Medical University of Innsbruck, Innsbruck, Austria
| |
Collapse
|
157
|
Serum Ferritin as a Diagnostic Biomarker for Severity of Childhood Sepsis. Indian Pediatr 2021. [DOI: 10.1007/s13312-021-2396-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
|
158
|
Cavone L, McCann T, Drake LK, Aguzzi EA, Oprişoreanu AM, Pedersen E, Sandi S, Selvarajah J, Tsarouchas TM, Wehner D, Keatinge M, Mysiak KS, Henderson BEP, Dobie R, Henderson NC, Becker T, Becker CG. A unique macrophage subpopulation signals directly to progenitor cells to promote regenerative neurogenesis in the zebrafish spinal cord. Dev Cell 2021; 56:1617-1630.e6. [PMID: 34033756 DOI: 10.1016/j.devcel.2021.04.031] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2020] [Revised: 03/15/2021] [Accepted: 04/28/2021] [Indexed: 12/14/2022]
Abstract
Central nervous system injury re-initiates neurogenesis in anamniotes (amphibians and fishes), but not in mammals. Activation of the innate immune system promotes regenerative neurogenesis, but it is fundamentally unknown whether this is indirect through the activation of known developmental signaling pathways or whether immune cells directly signal to progenitor cells using mechanisms that are unique to regeneration. Using single-cell RNA-seq of progenitor cells and macrophages, as well as cell-type-specific manipulations, we provide evidence for a direct signaling axis from specific lesion-activated macrophages to spinal progenitor cells to promote regenerative neurogenesis in zebrafish. Mechanistically, TNFa from pro-regenerative macrophages induces Tnfrsf1a-mediated AP-1 activity in progenitors to increase regeneration-promoting expression of hdac1 and neurogenesis. This establishes the principle that macrophages directly communicate to spinal progenitor cells via non-developmental signals after injury, providing potential targets for future interventions in the regeneration-deficient spinal cord of mammals.
Collapse
Affiliation(s)
- Leonardo Cavone
- Centre for Discovery Brain Sciences, University of Edinburgh, The Chancellor's Building, 49 Little France Crescent, Edinburgh EH16 4SB, UK
| | - Tess McCann
- Centre for Discovery Brain Sciences, University of Edinburgh, The Chancellor's Building, 49 Little France Crescent, Edinburgh EH16 4SB, UK
| | - Louisa K Drake
- Centre for Discovery Brain Sciences, University of Edinburgh, The Chancellor's Building, 49 Little France Crescent, Edinburgh EH16 4SB, UK
| | - Erika A Aguzzi
- Centre for Discovery Brain Sciences, University of Edinburgh, The Chancellor's Building, 49 Little France Crescent, Edinburgh EH16 4SB, UK
| | - Ana-Maria Oprişoreanu
- Centre for Discovery Brain Sciences, University of Edinburgh, The Chancellor's Building, 49 Little France Crescent, Edinburgh EH16 4SB, UK
| | - Elisa Pedersen
- Centre for Discovery Brain Sciences, University of Edinburgh, The Chancellor's Building, 49 Little France Crescent, Edinburgh EH16 4SB, UK
| | - Soe Sandi
- Centre for Discovery Brain Sciences, University of Edinburgh, The Chancellor's Building, 49 Little France Crescent, Edinburgh EH16 4SB, UK
| | - Jathurshan Selvarajah
- Centre for Discovery Brain Sciences, University of Edinburgh, The Chancellor's Building, 49 Little France Crescent, Edinburgh EH16 4SB, UK
| | - Themistoklis M Tsarouchas
- Centre for Discovery Brain Sciences, University of Edinburgh, The Chancellor's Building, 49 Little France Crescent, Edinburgh EH16 4SB, UK
| | - Daniel Wehner
- Centre for Discovery Brain Sciences, University of Edinburgh, The Chancellor's Building, 49 Little France Crescent, Edinburgh EH16 4SB, UK; Max Planck Institute for the Science of Light, Staudtstraße 2, Erlangen 91058, Germany; Max-Planck-Zentrum für Physik und Medizin, Staudtstraße 2, Erlangen 91058, Germany
| | - Marcus Keatinge
- Centre for Discovery Brain Sciences, University of Edinburgh, The Chancellor's Building, 49 Little France Crescent, Edinburgh EH16 4SB, UK
| | - Karolina S Mysiak
- Centre for Discovery Brain Sciences, University of Edinburgh, The Chancellor's Building, 49 Little France Crescent, Edinburgh EH16 4SB, UK
| | - Beth E P Henderson
- Centre for Inflammation Research, The Queen's Medical Research Institute, University of Edinburgh, Edinburgh, UK
| | - Ross Dobie
- Centre for Inflammation Research, The Queen's Medical Research Institute, University of Edinburgh, Edinburgh, UK
| | - Neil C Henderson
- Centre for Inflammation Research, The Queen's Medical Research Institute, University of Edinburgh, Edinburgh, UK; MRC Human Genetics Unit, Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, UK
| | - Thomas Becker
- Centre for Discovery Brain Sciences, University of Edinburgh, The Chancellor's Building, 49 Little France Crescent, Edinburgh EH16 4SB, UK.
| | - Catherina G Becker
- Centre for Discovery Brain Sciences, University of Edinburgh, The Chancellor's Building, 49 Little France Crescent, Edinburgh EH16 4SB, UK; Euan MacDonald Centre for Motor Neurone Disease Research University of Edinburgh, Edinburgh, UK.
| |
Collapse
|
159
|
Annous Y, Manning S, Khoujah D. Ferritin, fever, and frequent visits: Hyperferritinemic syndromes in the emergency department. Am J Emerg Med 2021; 48:249-254. [PMID: 34000525 DOI: 10.1016/j.ajem.2021.04.088] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2020] [Revised: 04/28/2021] [Accepted: 04/29/2021] [Indexed: 12/15/2022] Open
Abstract
Fever of unknown origin (FUO) is defined as persistent fevers without an identifiable cause despite extensive medical workup. Emergency physicians caring for patients reporting a persistent, nonspecific, febrile illness should carefully consider potentially serious non-infectious causes of FUO. We present a case of a 35-year-old man who presented to the emergency department (ED) three times over a 10-day period for persistent febrile illness and was ultimately diagnosed with Adult-Onset Still's Disease (AOSD) after a serum ferritin level was found to be over 42,000 μg/L. AOSD, along with macrophage activation syndrome, catastrophic antiphospholipid syndrome, and septic shock comprise the four hyperferritinemic syndromes. These are potentially life-threatening febrile illnesses that characteristically present with elevated ferritin levels. In this article, we highlight the value of a serum ferritin level in the workup of a patient with prolonged febrile illness and its utility in facilitating early diagnosis and prompt treatment of hyperferritinemic syndromes in the ED.
Collapse
Affiliation(s)
- Youssef Annous
- Department of Emergency Medicine, University of Maryland School of Medicine, Baltimore, MD 21201, USA.
| | - Sara Manning
- Department of Emergency Medicine, University of Maryland School of Medicine, Baltimore, MD 21201, USA.
| | - Danya Khoujah
- Department of Emergency Medicine, University of Maryland School of Medicine, Baltimore, MD 21201, USA.
| |
Collapse
|
160
|
Tajes M, Díez-López C, Enjuanes C, Moliner P, Ferreiro JL, Garay A, Jiménez-Marrero S, Yun S, Sosa SG, Alcoberro L, González-Costello J, García-Romero E, Yañez-Bisbe L, Benito B, Comín-Colet J. Neurohormonal activation induces intracellular iron deficiency and mitochondrial dysfunction in cardiac cells. Cell Biosci 2021; 11:89. [PMID: 34001233 PMCID: PMC8130332 DOI: 10.1186/s13578-021-00605-5] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Accepted: 05/06/2021] [Indexed: 12/17/2022] Open
Abstract
Background Iron deficiency (ID) is common in patients with heart failure (HF) and is associated with poor outcomes, yet its role in the pathophysiology of HF is not well-defined. We sought to determine the consequences of HF neurohormonal activation in iron homeostasis and mitochondrial function in cardiac cells. Methods HF was induced in C57BL/6 mice by using isoproterenol osmotic pumps and embryonic rat heart-derived H9c2 cells were subsequently challenged with Angiotensin II and/or Norepinephrine. The expression of several genes and proteins related to intracellular iron metabolism were assessed by Real time-PCR and immunoblotting, respectively. The intracellular iron levels were also determined. Mitochondrial function was analyzed by studying the mitochondrial membrane potential, the accumulation of radical oxygen species (ROS) and the adenosine triphosphate (ATP) production. Results Hearts from isoproterenol-stimulated mice showed a decreased in both mRNA and protein levels of iron regulatory proteins, transferrin receptor 1, ferroportin 1 and hepcidin compared to control mice. Furthermore, mitoferrin 2 and mitochondrial ferritin were also downregulated in the hearts from HF mice. Similar data regarding these key iron regulatory molecules were found in the H9c2 cells challenged with neurohormonal stimuli. Accordingly, a depletion of intracellular iron levels was found in the stimulated cells compared to non-stimulated cells, as well as in the hearts from the isoproterenol-induced HF mice. Finally, neurohormonal activation impaired mitochondrial function as indicated by the accumulation of ROS, the impaired mitochondrial membrane potential and the decrease in the ATP levels in the cardiac cells. Conclusions HF characteristic neurohormonal activation induced changes in the regulation of key molecules involved in iron homeostasis, reduced intracellular iron levels and impaired mitochondrial function. The current results suggest that iron could be involved in the pathophysiology of HF.
Collapse
Affiliation(s)
- M Tajes
- Bio-Heart Cardiovascular Diseases Research Group, Bellvitge Biomedical Research Institute (IDIBELL), L'Hospitalet de Llobregat, Barcelona, Spain.,Cardiology Department, Bellvitge University Hospital, L'Hospitalet de Llobregat, Barcelona, Spain
| | - C Díez-López
- Bio-Heart Cardiovascular Diseases Research Group, Bellvitge Biomedical Research Institute (IDIBELL), L'Hospitalet de Llobregat, Barcelona, Spain.,Cardiology Department, Bellvitge University Hospital, L'Hospitalet de Llobregat, Barcelona, Spain.,Advanced Heart Failure and Heart Transplant Unit, Cardiology Department, Bellvitge University Hospital, L'Hospitalet de Llobregat, Barcelona, Spain.,Department of Clinical Sciences, School of Medicine, University of Barcelona, Barcelona, Spain
| | - C Enjuanes
- Bio-Heart Cardiovascular Diseases Research Group, Bellvitge Biomedical Research Institute (IDIBELL), L'Hospitalet de Llobregat, Barcelona, Spain.,Cardiology Department, Bellvitge University Hospital, L'Hospitalet de Llobregat, Barcelona, Spain.,Community Heart Failure Program, Cardiology Department, Bellvitge University Hospital, L'Hospitalet de Llobregat, Barcelona, Spain
| | - P Moliner
- Bio-Heart Cardiovascular Diseases Research Group, Bellvitge Biomedical Research Institute (IDIBELL), L'Hospitalet de Llobregat, Barcelona, Spain.,Cardiology Department, Bellvitge University Hospital, L'Hospitalet de Llobregat, Barcelona, Spain.,Community Heart Failure Program, Cardiology Department, Bellvitge University Hospital, L'Hospitalet de Llobregat, Barcelona, Spain
| | - J L Ferreiro
- Bio-Heart Cardiovascular Diseases Research Group, Bellvitge Biomedical Research Institute (IDIBELL), L'Hospitalet de Llobregat, Barcelona, Spain.,Cardiology Department, Bellvitge University Hospital, L'Hospitalet de Llobregat, Barcelona, Spain
| | - A Garay
- Bio-Heart Cardiovascular Diseases Research Group, Bellvitge Biomedical Research Institute (IDIBELL), L'Hospitalet de Llobregat, Barcelona, Spain.,Cardiology Department, Bellvitge University Hospital, L'Hospitalet de Llobregat, Barcelona, Spain.,Community Heart Failure Program, Cardiology Department, Bellvitge University Hospital, L'Hospitalet de Llobregat, Barcelona, Spain
| | - S Jiménez-Marrero
- Bio-Heart Cardiovascular Diseases Research Group, Bellvitge Biomedical Research Institute (IDIBELL), L'Hospitalet de Llobregat, Barcelona, Spain.,Cardiology Department, Bellvitge University Hospital, L'Hospitalet de Llobregat, Barcelona, Spain.,Community Heart Failure Program, Cardiology Department, Bellvitge University Hospital, L'Hospitalet de Llobregat, Barcelona, Spain
| | - S Yun
- Bio-Heart Cardiovascular Diseases Research Group, Bellvitge Biomedical Research Institute (IDIBELL), L'Hospitalet de Llobregat, Barcelona, Spain.,Community Heart Failure Program, Cardiology Department, Bellvitge University Hospital, L'Hospitalet de Llobregat, Barcelona, Spain.,Department of Internal Medicine, Bellvitge University Hospital, L'Hospitalet de Llobregat, Barcelona, Spain
| | - S G Sosa
- Bio-Heart Cardiovascular Diseases Research Group, Bellvitge Biomedical Research Institute (IDIBELL), L'Hospitalet de Llobregat, Barcelona, Spain.,Cardiology Department, Bellvitge University Hospital, L'Hospitalet de Llobregat, Barcelona, Spain
| | - L Alcoberro
- Bio-Heart Cardiovascular Diseases Research Group, Bellvitge Biomedical Research Institute (IDIBELL), L'Hospitalet de Llobregat, Barcelona, Spain.,Cardiology Department, Bellvitge University Hospital, L'Hospitalet de Llobregat, Barcelona, Spain.,Community Heart Failure Program, Cardiology Department, Bellvitge University Hospital, L'Hospitalet de Llobregat, Barcelona, Spain
| | - J González-Costello
- Bio-Heart Cardiovascular Diseases Research Group, Bellvitge Biomedical Research Institute (IDIBELL), L'Hospitalet de Llobregat, Barcelona, Spain.,Cardiology Department, Bellvitge University Hospital, L'Hospitalet de Llobregat, Barcelona, Spain.,Advanced Heart Failure and Heart Transplant Unit, Cardiology Department, Bellvitge University Hospital, L'Hospitalet de Llobregat, Barcelona, Spain.,Department of Clinical Sciences, School of Medicine, University of Barcelona, Barcelona, Spain
| | - E García-Romero
- Bio-Heart Cardiovascular Diseases Research Group, Bellvitge Biomedical Research Institute (IDIBELL), L'Hospitalet de Llobregat, Barcelona, Spain.,Cardiology Department, Bellvitge University Hospital, L'Hospitalet de Llobregat, Barcelona, Spain.,Advanced Heart Failure and Heart Transplant Unit, Cardiology Department, Bellvitge University Hospital, L'Hospitalet de Llobregat, Barcelona, Spain
| | - L Yañez-Bisbe
- Vascular Biology and Metabolism Program, Vall d'Hebron Research Institute (VHIR), Barcelona, Spain
| | - B Benito
- Vascular Biology and Metabolism Program, Vall d'Hebron Research Institute (VHIR), Barcelona, Spain.,Cardiology Department, Hospital Vall d'Hebron Hospital, Barcelona, Spain.,Department of Medicine, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - J Comín-Colet
- Bio-Heart Cardiovascular Diseases Research Group, Bellvitge Biomedical Research Institute (IDIBELL), L'Hospitalet de Llobregat, Barcelona, Spain. .,Cardiology Department, Bellvitge University Hospital, L'Hospitalet de Llobregat, Barcelona, Spain. .,Community Heart Failure Program, Cardiology Department, Bellvitge University Hospital, L'Hospitalet de Llobregat, Barcelona, Spain. .,Department of Clinical Sciences, School of Medicine, University of Barcelona, Barcelona, Spain.
| |
Collapse
|
161
|
Hyperferritinemia-A Clinical Overview. J Clin Med 2021; 10:jcm10092008. [PMID: 34067164 PMCID: PMC8125175 DOI: 10.3390/jcm10092008] [Citation(s) in RCA: 61] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Revised: 05/03/2021] [Accepted: 05/05/2021] [Indexed: 02/07/2023] Open
Abstract
Ferritin is one of the most frequently requested laboratory tests in primary and secondary care, and levels often deviate from reference ranges. Serving as an indirect marker for total body iron stores, low ferritin is highly specific for iron deficiency. Hyperferritinemia is, however, a non-specific finding, which is frequently overlooked in general practice. In routine medical practice, only 10% of cases are related to an iron overload, whilst the rest is seen as a result of acute phase reactions and reactive increases in ferritin due to underlying conditions. Differentiation of the presence or absence of an associated iron overload upon hyperferritinemia is essential, although often proves to be complex. In this review, we have performed a review of a selection of the literature based on the authors’ own experiences and assessments in accordance with international recommendations and guidelines. We address the biology, etiology, and epidemiology of hyperferritinemia. Finally, an algorithm for the diagnostic workup and management of hyperferritinemia is proposed, and general principles regarding the treatment of iron overload are discussed.
Collapse
|
162
|
Heme Oxygenase-1 Signaling and Redox Homeostasis in Physiopathological Conditions. Biomolecules 2021; 11:biom11040589. [PMID: 33923744 PMCID: PMC8072688 DOI: 10.3390/biom11040589] [Citation(s) in RCA: 150] [Impact Index Per Article: 37.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Revised: 04/07/2021] [Accepted: 04/14/2021] [Indexed: 02/06/2023] Open
Abstract
Heme-oxygenase is the enzyme responsible for degradation of endogenous iron protoporphyirin heme; it catalyzes the reaction’s rate-limiting step, resulting in the release of carbon monoxide (CO), ferrous ions, and biliverdin (BV), which is successively reduced in bilirubin (BR) by biliverdin reductase. Several studies have drawn attention to the controversial role of HO-1, the enzyme inducible isoform, pointing out its implications in cancer and other diseases development, but also underlining the importance of its antioxidant activity. The contribution of HO-1 in redox homeostasis leads to a relevant decrease in cells oxidative damage, which can be reconducted to its cytoprotective effects explicated alongside other endogenous mechanisms involving genes like TIGAR (TP53-induced glycolysis and apoptosis regulator), but also to the therapeutic functions of heme main transformation products, especially carbon monoxide (CO), which has been shown to be effective on GSH levels implementation sustaining body’s antioxidant response to oxidative stress. The aim of this review was to collect most of the knowledge on HO-1 from literature, analyzing different perspectives to try and put forward a hypothesis on revealing yet unknown HO-1-involved pathways that could be useful to promote development of new therapeutical strategies, and lay the foundation for further investigation to fully understand this important antioxidant system.
Collapse
|
163
|
Wlazlo E, Mehrad B, Morel L, Scindia Y. Iron Metabolism: An Under Investigated Driver of Renal Pathology in Lupus Nephritis. Front Med (Lausanne) 2021; 8:643686. [PMID: 33912577 PMCID: PMC8071941 DOI: 10.3389/fmed.2021.643686] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Accepted: 03/10/2021] [Indexed: 12/13/2022] Open
Abstract
Nephritis is a common manifestation of systemic lupus erythematosus, a condition associated with inflammation and iron imbalance. Renal tubules are the work horse of the nephron. They contain a large number of mitochondria that require iron for oxidative phosphorylation, and a tight control of intracellular iron prevents excessive generation of reactive oxygen species. Iron supply to the kidney is dependent on systemic iron availability, which is regulated by the hepcidin-ferroportin axis. Most of the filtered plasma iron is reabsorbed in proximal tubules, a process that is controlled in part by iron regulatory proteins. This review summarizes tubulointerstitial injury in lupus nephritis and current understanding of how renal tubular cells regulate intracellular iron levels, highlighting the role of iron imbalance in the proximal tubules as a driver of tubulointerstitial injury in lupus nephritis. We propose a model based on the dynamic ability of iron to catalyze reactive oxygen species, which can lead to an accumulation of lipid hydroperoxides in proximal tubular epithelial cells. These iron-catalyzed oxidative species can also accentuate protein and autoantibody-induced inflammatory transcription factors leading to matrix, cytokine/chemokine production and immune cell infiltration. This could potentially explain the interplay between increased glomerular permeability and the ensuing tubular injury, tubulointerstitial inflammation and progression to renal failure in LN, and open new avenues of research to develop novel therapies targeting iron metabolism.
Collapse
Affiliation(s)
- Ewa Wlazlo
- Department of Medicine, University of Virginia School of Medicine, Charlottesville, VA, United States
| | - Borna Mehrad
- Division of Pulmonary, Critical Care, and Sleep Medicine, University of Florida, Gainesville, FL, United States.,Department of Pathology, University of Florida, Gainesville, FL, United States
| | - Laurence Morel
- Department of Pathology, University of Florida, Gainesville, FL, United States
| | - Yogesh Scindia
- Division of Pulmonary, Critical Care, and Sleep Medicine, University of Florida, Gainesville, FL, United States.,Department of Pathology, University of Florida, Gainesville, FL, United States.,Division of Nephrology, University of Florida, Gainesville, FL, United States
| |
Collapse
|
164
|
Chimeric Protein IPath ® with Chelating Activity Improves Atlantic Salmon's Immunity against Infectious Diseases. Vaccines (Basel) 2021; 9:vaccines9040361. [PMID: 33918540 PMCID: PMC8068967 DOI: 10.3390/vaccines9040361] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Revised: 03/17/2021] [Accepted: 03/23/2021] [Indexed: 02/07/2023] Open
Abstract
Infection processes displayed by pathogens require the acquisition of essential inorganic nutrients and trace elements from the host to survive and proliferate. Without a doubt, iron is a crucial trace metal for all living organisms and also a pivotal component in the host–parasite interactions. In particular, the host reduces the iron available to face the infectious disease, increasing iron transport proteins’ expression and activating the heme synthesis and degradation pathways. Moreover, recent findings have suggested that iron metabolism modulation in fish promotes the immune response by reducing cellular iron toxicity. We hypothesized that recombinant proteins related to iron metabolism could modulate the fish’s immune system through iron metabolism and iron-responsive genes. Here a chimeric iron transport protein (IPath®) was bioinformatically designed and then expressed in a recombinant bacterial system. The IPath® protein showed a significant chelating activity under in vitro conditions and biological activity. Taking this evidence, a vaccine candidate based on IPath® was evaluated in Atlantic salmon challenged with three different fish pathogens. Experimental trials were conducted using two fish groups: one immunized with IPath® and another injected with adjutant as the control group. After 400 accumulated thermal units (ATUs), two different infection trials were performed. In the first one, fish were infected with the bacterium Aeromonas salmonicida, and in a second trial, fish were exposed to the ectoparasite Caligus rogercresseyi and subsequently infected with the intracellular bacterium Piscirickettsia salmonis. Fish immunized with IPath® showed a significant delay in the mortality curve in response to A. salmonicida and P. salmonis infections. However, no significant differences between infected and control fish groups were observed at the end of the experiment. Notably, sea lice burden reduction was observed in vaccinated Atlantic salmon. Transcriptional analysis evidenced a high modulation of iron-homeostasis-related genes in fish vaccinated with IPath® compared to the control group during the infection. Moreover, increasing expression of Atlantic salmon IgT was associated with IPath® immunization. This study provides evidence that the IPath® protein could be used as an antigen or booster in commercial fish vaccines, improving the immune response against relevant pathogens for salmon aquaculture.
Collapse
|
165
|
Stanford FA, Matthies N, Cseresnyés Z, Figge MT, Hassan MIA, Voigt K. Expression Patterns in Reductive Iron Assimilation and Functional Consequences during Phagocytosis of Lichtheimia corymbifera, an Emerging Cause of Mucormycosis. J Fungi (Basel) 2021; 7:jof7040272. [PMID: 33916756 PMCID: PMC8065604 DOI: 10.3390/jof7040272] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Revised: 03/17/2021] [Accepted: 03/29/2021] [Indexed: 12/30/2022] Open
Abstract
Iron is an essential micronutrient for most organisms and fungi are no exception. Iron uptake by fungi is facilitated by receptor-mediated internalization of siderophores, heme and reductive iron assimilation (RIA). The RIA employs three protein groups: (i) the ferric reductases (Fre5 proteins), (ii) the multicopper ferroxidases (Fet3) and (iii) the high-affinity iron permeases (Ftr1). Phenotyping under different iron concentrations revealed detrimental effects on spore swelling and hyphal formation under iron depletion, but yeast-like morphology under iron excess. Since access to iron is limited during pathogenesis, pathogens are placed under stress due to nutrient limitations. To combat this, gene duplication and differential gene expression of key iron uptake genes are utilized to acquire iron against the deleterious effects of iron depletion. In the genome of the human pathogenic fungus L. corymbifera, three, four and three copies were identified for FRE5, FTR1 and FET3 genes, respectively. As in other fungi, FET3 and FTR1 are syntenic and co-expressed in L. corymbifera. Expression of FRE5, FTR1 and FET3 genes is highly up-regulated during iron limitation (Fe-), but lower during iron excess (Fe+). Fe- dependent upregulation of gene expression takes place in LcFRE5 II and III, LcFTR1 I and II, as well as LcFET3 I and II suggesting a functional role in pathogenesis. The syntenic LcFTR1 I–LcFET3 I gene pair is co-expressed during germination, whereas LcFTR1 II- LcFET3 II is co-expressed during hyphal proliferation. LcFTR1 I, II and IV were overexpressed in Saccharomyces cerevisiae to represent high and moderate expression of intracellular transport of Fe3+, respectively. Challenge of macrophages with the yeast mutants revealed no obvious role for LcFTR1 I, but possible functions of LcFTR1 II and IVs in recognition by macrophages. RIA expression pattern was used for a new model of interaction between L. corymbifera and macrophages.
Collapse
Affiliation(s)
- Felicia Adelina Stanford
- Jena Microbial Resource Collection, Leibniz Institute for Natural Product Research, and Infection Biology—Hans Knöll Institute (HKI), 07745 Jena, Germany; (F.A.S.); (N.M.); (M.I.A.H.)
- Institute of Microbiology, Friedrich Schiller University Jena, 07743 Jena, Germany;
| | - Nina Matthies
- Jena Microbial Resource Collection, Leibniz Institute for Natural Product Research, and Infection Biology—Hans Knöll Institute (HKI), 07745 Jena, Germany; (F.A.S.); (N.M.); (M.I.A.H.)
- Institute of Microbiology, Friedrich Schiller University Jena, 07743 Jena, Germany;
| | - Zoltán Cseresnyés
- Applied Systems Biology, Leibniz Institute for Natural Product Research, and Infection Biology—Hans Knöll Institute, 12622 Jena, Germany;
| | - Marc Thilo Figge
- Institute of Microbiology, Friedrich Schiller University Jena, 07743 Jena, Germany;
- Applied Systems Biology, Leibniz Institute for Natural Product Research, and Infection Biology—Hans Knöll Institute, 12622 Jena, Germany;
| | - Mohamed I. Abdelwahab Hassan
- Jena Microbial Resource Collection, Leibniz Institute for Natural Product Research, and Infection Biology—Hans Knöll Institute (HKI), 07745 Jena, Germany; (F.A.S.); (N.M.); (M.I.A.H.)
- Institute of Microbiology, Friedrich Schiller University Jena, 07743 Jena, Germany;
- National Research Centre, Pests & Plant Protection Department, 33rd El Buhouth St., Dokki, Giza 12622, Egypt
| | - Kerstin Voigt
- Jena Microbial Resource Collection, Leibniz Institute for Natural Product Research, and Infection Biology—Hans Knöll Institute (HKI), 07745 Jena, Germany; (F.A.S.); (N.M.); (M.I.A.H.)
- Institute of Microbiology, Friedrich Schiller University Jena, 07743 Jena, Germany;
- Correspondence: or ; Tel.: +49-3641-532-1395
| |
Collapse
|
166
|
Hepatocyte growth factor protects PC12 cells against OGD/R-induced injury by reducing iron. Biosci Rep 2021; 40:222408. [PMID: 32186328 PMCID: PMC7109004 DOI: 10.1042/bsr20200287] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2020] [Revised: 03/11/2020] [Accepted: 03/16/2020] [Indexed: 01/20/2023] Open
Abstract
In the light of hepatocyte growth factor (HGF) the inhibiting role on the expression of hepcidin, we hypothesized that HGF might be able to reduce cell and tissue iron by increasing ferroportin 1 (Fpn1) content and Fpn1-mediated iron release from cells and tissues. The hypothesized ability of HGF to reduce iron might be one of the mechanisms associated with its neuroprotective action under the conditions of ischemia/reperfusion (I/R). Here, we investigated the effects of HGF on the expression of hepcidin as well as transferrin receptor 1 (TfR1), divalent metal transporter 1 (DMT1), Fpn1, ferritin and iron regulatory proteins (IRPs) in oxygen-glucose deprivation and reoxygenation (OGD/R)-treated PC12 cells by real-time PCR and Western blot analysis. We demonstrated that HGF could completely reverse the OGD/R-induced reduction in Fpn1 and IRP1 expression and increase in ferritin light chain protein and hepcidin mRNA levels in PC12 cells. It was concluded that HGF protects PC12 cells against OGD/R-induced injury mainly by reducing cell iron contents via the up-regulation of Fpn1 and increased Fpn1-mediated iron export from cells. Our findings suggested that HGF may also be able to ameliorate OGD/R or I/R-induced overloading of brain iron by promoting Fpn1 expression.
Collapse
|
167
|
Hernández-Fernández J, Pinzón-Velasco A, López EA, Rodríguez-Becerra P, Mariño-Ramírez L. Transcriptional Analyses of Acute Exposure to Methylmercury on Erythrocytes of Loggerhead Sea Turtle. TOXICS 2021; 9:70. [PMID: 33805397 PMCID: PMC8066450 DOI: 10.3390/toxics9040070] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/13/2021] [Revised: 03/11/2021] [Accepted: 03/17/2021] [Indexed: 01/09/2023]
Abstract
To understand changes in enzyme activity and gene expression as biomarkers of exposure to methylmercury, we exposed loggerhead turtle erythrocytes (RBCs) to concentrations of 0, 1, and 5 mg L-1 of MeHg and de novo transcriptome were assembled using RNA-seq. The analysis of differentially expressed genes (DEGs) indicated that 79 unique genes were dysregulated (39 upregulated and 44 downregulated genes). The results showed that MeHg altered gene expression patterns as a response to the cellular stress produced, reflected in cell cycle regulation, lysosomal activity, autophagy, calcium regulation, mitochondrial regulation, apoptosis, and regulation of transcription and translation. The analysis of DEGs showed a low response of the antioxidant machinery to MeHg, evidenced by the fact that genes of early response to oxidative stress were not dysregulated. The RBCs maintained a constitutive expression of proteins that represented a good part of the defense against reactive oxygen species (ROS) induced by MeHg.
Collapse
Affiliation(s)
- Javier Hernández-Fernández
- Department of Natural and Environmental Science, Marine Biology Program, Faculty of Science and Engineering, Genetics, Molecular Biology and Bioinformatic Research Group–GENBIMOL, Jorge Tadeo Lozano University, Cra. 4 No 22-61, Bogotá 110311, Colombia;
- Faculty of Sciences, Department of Biology, Pontificia Universidad Javeriana, Calle 45, Cra. 7, Bogotá 110231, Colombia
| | - Andrés Pinzón-Velasco
- Bioinformática y Biología de Sistemas, Universidad Nacional de Colombia, Calle 45, Cra. 30, Bogotá 111321, Colombia;
| | - Ellie Anne López
- IDEASA Research Group-Environment and Sustainability, Institute of Environmental Studies and Services, Sergio Arboleda University, Bogotá 111711, Colombia;
| | - Pilar Rodríguez-Becerra
- Department of Natural and Environmental Science, Marine Biology Program, Faculty of Science and Engineering, Genetics, Molecular Biology and Bioinformatic Research Group–GENBIMOL, Jorge Tadeo Lozano University, Cra. 4 No 22-61, Bogotá 110311, Colombia;
| | - Leonardo Mariño-Ramírez
- NCBI, NLM, NIH Computational Biology Branch, Building 38A, Room 6S614M 8600 Rockville Pike, MSC 6075, Bethesda, MD 20894-6075, USA;
| |
Collapse
|
168
|
Moreto F, Ferron AJT, Francisqueti-Ferron FV, D'Amato A, Garcia JL, Costa MR, Silva CCVA, Altomare A, Correa CR, Aldini G, Ferreira ALA. Differentially expressed proteins obtained by label-free quantitative proteomic analysis reveal affected biological processes and functions in Western diet-induced steatohepatitis. J Biochem Mol Toxicol 2021; 35:1-11. [PMID: 33729641 DOI: 10.1002/jbt.22751] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Revised: 10/26/2020] [Accepted: 02/24/2021] [Indexed: 12/12/2022]
Abstract
Nonalcoholic steatohepatitis (NASH) is a pathological manifestation with a progressive incidence in response to the epidemic of hepatic steatosis caused primarily by excessive energy intake. The present study unravels affected biological processes and functions by the presence of NASH in rats using a label-free quantitative proteomic strategy. NASH was induced by a Western high-sugar and high-fat diet for 20 weeks. The liver tissue was collected for histology and for a mass spectrometry-based proteomic protocol. The NASH group showed severe lipidosis, hepatocyte ballooning, and the presence of collagen deposition. Among upregulated proteins in NASH perilipin-2 (Plin-2; F6QBA3; difference [diff]: 2.29), ferritin heavy (Fth1; Q66HI5; diff: 2.19) and light (Ftl1; P02793; diff: 1.75) chains, macrophage migration inhibitory factor 1 (Mif; P30904; diff: 1.69), and fibronectin (Fn1; F1LST1; diff: 0.35) were observed, whereas among downregulated proteins, plectin (Q6S399; diff: -3.34), some Cyp2 family proteins of the cytochrome P450 complex, glutathione S-transferases, flavin-containing monooxygenase 1 (Fmo1; P36365; diff: -2.08), acetyl-CoA acetyltransferase 2 (Acat2; Q5XI22; diff: -2.25), acyl-CoA oxidase 2 (Acox2; F1LNW3; diff: -1.59), and acyl-CoA oxidase 3 (Acox3; F1M9A7; diff: -2.41) were observed. Also, biological processes and functions such as LPS/IL-1 inhibition of RXR, fatty acid metabolism, Nrf2-mediated oxidative stress response, xenobiotic metabolism, and PXR/RXR and CAR/RXR activations were predicted to be affected. In conclusion, the liver of rats with NASH induced by Western diet shows a decreased capacity of metabolizing lipids, fatty acids, and xenobiotic compounds that predispose fibrosis development.
Collapse
Affiliation(s)
- Fernando Moreto
- Medical School, Sao Paulo State University, Botucatu, Brazil
| | | | | | - Alfonsina D'Amato
- Department of Pharmaceutical Sciences, University of Milan, Milan, Italy
| | | | - Mariane R Costa
- Medical School, Sao Paulo State University, Botucatu, Brazil
| | | | | | | | - Giancarlo Aldini
- Department of Pharmaceutical Sciences, University of Milan, Milan, Italy
| | | |
Collapse
|
169
|
Serum ferritin at admission in hospitalized COVID-19 patients as a predictor of mortality. Braz J Infect Dis 2021; 25:101569. [PMID: 33736948 PMCID: PMC7959266 DOI: 10.1016/j.bjid.2021.101569] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Revised: 01/16/2021] [Accepted: 02/10/2021] [Indexed: 12/23/2022] Open
Abstract
Introduction Some COVID-19 patients have higher mortality and the responsible factors for this unfavorable outcome is still not well understood. Objective To study the association between ferritin levels at admission, representing an inflammatory state, and hospital mortality in COVID-19 patients. Methods From May through July 2020, SARS-CoV-2 positive patients with moderate to severe clinical symptoms were evaluated at admission, regarding clinical and laboratory data on renal and hepatic function, hematologic parameters, cytomegalovirus co-infection, and acute phase proteins. Results A total of 97 patients were included; mean age = 59.9 ± 16.3 years, 58.8% male, 57.7% non-white, in-hospital mortality = 45.4%. Age, ferritin, C-reactive protein, serum albumin and creatinine were significantly associated with mortality. Ferritin showed area under the curve (AUC) of 0.79 (p < 0.001) for the cut-off of 1873.0 ng/mL, sensitivity of 68.4% and specificity of 79.3% in predicting in-hospital mortality. Age ≥60 years had an odds ratio (OR) of 10.5 (95% CI = 1.8–59.5; p = 0.008) and ferritin ≥1873.0 ng/mL had an OR of 6.0 (95% CI = 1.4–26.2; p = 0.016), both independently associated with mortality based on logistic regression analysis. Conclusion The magnitude of inflammation present at admission of COVID-19 patients, represented by high ferritin levels, is independently predictive of in-hospital mortality.
Collapse
|
170
|
Iron control of erythroid microtubule cytoskeleton as a potential target in treatment of iron-restricted anemia. Nat Commun 2021; 12:1645. [PMID: 33712594 PMCID: PMC7955080 DOI: 10.1038/s41467-021-21938-2] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2020] [Accepted: 02/20/2021] [Indexed: 12/17/2022] Open
Abstract
Anemias of chronic disease and inflammation (ACDI) result from restricted iron delivery to erythroid progenitors. The current studies reveal an organellar response in erythroid iron restriction consisting of disassembly of the microtubule cytoskeleton and associated Golgi disruption. Isocitrate supplementation, known to abrogate the erythroid iron restriction response, induces reassembly of microtubules and Golgi in iron deprived progenitors. Ferritin, based on proteomic profiles, regulation by iron and isocitrate, and putative interaction with microtubules, is assessed as a candidate mediator. Knockdown of ferritin heavy chain (FTH1) in iron replete progenitors induces microtubule collapse and erythropoietic blockade; conversely, enforced ferritin expression rescues erythroid differentiation under conditions of iron restriction. Fumarate, a known ferritin inducer, synergizes with isocitrate in reversing molecular and cellular defects of iron restriction and in oral remediation of murine anemia. These findings identify a cytoskeletal component of erythroid iron restriction and demonstrate potential for its therapeutic targeting in ACDI. Debilitating anemias in chronic diseases can result from deficient iron delivery to red cell precursors. Here, the authors show how this deficiency damages the cytoskeletal framework of progenitor cells and identify a targeted strategy for cytoskeletal repair, leading to anemia correction.
Collapse
|
171
|
Transcriptomic and Histopathological Effects of Bifenthrin to the Brain of Juvenile Rainbow Trout ( Oncorhynchus mykiss). TOXICS 2021; 9:toxics9030048. [PMID: 33807887 PMCID: PMC8000926 DOI: 10.3390/toxics9030048] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Revised: 03/03/2021] [Accepted: 03/04/2021] [Indexed: 01/07/2023]
Abstract
The increased global use of pyrethroids raises concern for non-target aquatic species. Bifenthrin, among the most predominantly detected pyrethroids in the environment, is frequently measured in water samples above concentrations reported to induce neuroendocrine and neurotoxic effects to several threatened and endangered fish species, such as the Chinook salmon and steelhead trout. To better characterize the neurotoxic effect of bifenthrin to salmonids, rainbow trout were treated with environmentally relevant concentrations of bifenthrin (15 and 30 ng/L) for two weeks and assessed for changes in transcriptomic profiles and histopathological alterations. The top bioinformatic pathways predicted to be impaired in bifenthrin-exposed trout were involved in gonadotropin releasing hormone signaling, the dysregulation of iron homeostasis, reduced extracellular matrix stability and adhesion, and cell death. Subsequent histopathological analysis showed a significant increase in TUNEL positive cells in the cerebellum and optic tectum of bifenthrin-treated trout, relative to controls (p < 0.05). These findings suggest that low, ng/L concentrations of bifenthrin are capable of dysregulating proper neuroendocrine function, impair the structural integrity of the extracellular matrix and cell signaling pathways in the brain, and induce apoptosis in neurons of juvenile salmonids following bifenthrin treatment, which is consistent with metabolomic profiles demonstrating a common target and mechanism.
Collapse
|
172
|
Liu Q, Wu J, Zhang X, Wu X, Zhao Y, Ren J. Iron homeostasis and disorders revisited in the sepsis. Free Radic Biol Med 2021; 165:1-13. [PMID: 33486088 DOI: 10.1016/j.freeradbiomed.2021.01.025] [Citation(s) in RCA: 59] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/20/2020] [Revised: 12/31/2020] [Accepted: 01/11/2021] [Indexed: 12/26/2022]
Abstract
Sepsis is a life-threatening condition caused by a dysregulated host-response to inflammation, although it currently lacks a fully elucidated pathobiology. Iron is a crucial trace element that is essential for fundamental processes in both humans and bacteria. During sepsis, iron metabolism is altered, including increased iron transport and uptake into cells and decreased iron export. The intracellular sequestration of iron limits its availability to circulating pathogens, which serves as a conservative strategy against the pathogens. Although iron retention has been showed to have protective protect effects, an increase in labile iron may cause oxidative injury and cell death (e.g., pyroptosis, ferroptosis) as the condition progresses. Moreover, iron disorders are substantial and correlate with the severity of sepsis. This also suggests that iron may be useful as a diagnostic marker for evaluating the severity and predicting the outcome of the disease. Further knowledge about these disorders could help in evaluating how drugs targeting iron homeostasis can be optimally applied to improve the treatment of patients with sepsis. Here, we present a comprehensive review of recent advances in the understanding of iron metabolism, focusing on the regulatory mechanisms and iron-mediated injury in sepsis.
Collapse
Affiliation(s)
- Qinjie Liu
- Research Institute of General Surgery, Jinling Hospital, Medical School of Nanjing University, Nanjing, 210002, PR China.
| | - Jie Wu
- Department of General Surgery, BenQ Medical Center, The Affiliated BenQ Hospital of Nanjing Medical University, Nanjing, 210002, PR China.
| | - Xufei Zhang
- Research Institute of General Surgery, Jinling Hospital, Nanjing Medical University, Nanjing, 210002, PR China.
| | - Xiuwen Wu
- Research Institute of General Surgery, Jinling Hospital, Nanjing, 210002, PR China.
| | - Yun Zhao
- Department of General Surgery, BenQ Medical Center, The Affiliated BenQ Hospital of Nanjing Medical University, Nanjing, 210002, PR China.
| | - Jianan Ren
- Research Institute of General Surgery, Jinling Hospital, Medical School of Nanjing University, Nanjing, 210002, PR China; Department of General Surgery, BenQ Medical Center, The Affiliated BenQ Hospital of Nanjing Medical University, Nanjing, 210002, PR China; Research Institute of General Surgery, Jinling Hospital, Nanjing Medical University, Nanjing, 210002, PR China.
| |
Collapse
|
173
|
Human Parainfluenza Virus Type 2 V Protein Modulates Iron Homeostasis. J Virol 2021; 95:JVI.01861-20. [PMID: 33408172 DOI: 10.1128/jvi.01861-20] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2020] [Accepted: 12/15/2020] [Indexed: 12/19/2022] Open
Abstract
Intracellular iron concentration is tightly controlled for cell viability. It is known to affect the growth of several viruses, but the molecular mechanisms are not well understood. We found that iron chelators inhibit growth of human parainfluenza virus type 2 (hPIV-2). Furthermore, infection with hPIV-2 alters ferritin localization from granules to a homogenous distribution within cytoplasm of iron-stimulated cells. The V protein of hPIV-2 interacts with ferritin heavy chain 1 (FTH1), a ferritin subunit. It also binds to nuclear receptor coactivator 4 (NCOA4), which mediates autophagic degradation of ferritin, so-called ferritinophagy. V protein consequently interferes with interaction between FTH1 and NCOA4. hPIV-2 growth is inhibited in FTH1 knockdown cell line where severe hPIV-2-induced apoptosis is shown. In contrast, NCOA4 knockdown results in the promotion of hPIV-2 growth and limited apoptosis. Our data collectively suggest that hPIV-2 V protein inhibits FTH1-NCOA4 interaction and subsequent ferritinophagy. This iron homeostasis modulation allows infected cells to avoid apoptotic cell death, resulting in effective growth of hPIV-2.IMPORTANCE hPIV-2 V protein interferes with interaction between FTH1 and NCOA4 and inhibits NCOA4-mediated ferritin degradation, leading to the inhibition of iron release to the cytoplasm. This iron homeostasis modulation allows infected cells to avoid apoptotic cell death, resulting in effective growth of hPIV-2.
Collapse
|
174
|
Ryan BJ, Foug KL, Gioscia-Ryan RA, Varshney P, Ludzki AC, Ahn C, Schleh MW, Gillen JB, Chenevert TL, Horowitz JF. Exercise training decreases whole-body and tissue iron storage in adults with obesity. Exp Physiol 2021; 106:820-827. [PMID: 33559926 DOI: 10.1113/ep089272] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2020] [Accepted: 01/29/2021] [Indexed: 11/08/2022]
Abstract
NEW FINDINGS What is the central question of this study? Does exercise training modify tissue iron storage in adults with obesity? What is the main finding and its importance? Twelve weeks of moderate-intensity exercise or high-intensity interval training lowered whole-body iron stores, decreased the abundance of the key iron storage protein in skeletal muscle (ferritin) and tended to lower hepatic iron content. These findings show that exercise training can reduce tissue iron storage in adults with obesity and might have important implications for obese individuals with dysregulated iron homeostasis. ABSTRACT The regulation of iron storage is crucial to human health, because both excess and deficient iron storage have adverse consequences. Recent studies suggest altered iron storage in adults with obesity, with increased iron accumulation in their liver and skeletal muscle. Exercise training increases iron use for processes such as red blood cell production and can lower whole-body iron stores in humans. However, the effects of exercise training on liver and muscle iron stores in adults with obesity have not been assessed. The aim of this study was to determine the effects of 12 weeks of exercise training on whole-body iron stores, liver iron content and the abundance of ferritin (the key iron storage protein) in skeletal muscle in adults with obesity. Twenty-two inactive adults (11 women and 11 men; age, 31 ± 6 years; body mass index, 33 ± 3 kg/m2 ) completed 12 weeks (four sessions/week) of either moderate-intensity continuous training (MICT; 45 min at 70% of maximal heart rate; n = 11) or high-intensity interval training (HIIT; 10 × 1 min at 90% of maximal heart rate, interspersed with 1 min active recovery; n = 11). Whole-body iron stores were lower after training, as indicated by decreased plasma concentrations of ferritin (P = 3 × 10-5 ) and hepcidin (P = 0.02), without any change in C-reactive protein. Hepatic R2*, an index of liver iron content, was 6% lower after training (P = 0.06). Training reduced the skeletal muscle abundance of ferritin by 10% (P = 0.03), suggesting lower muscle iron storage. Interestingly, these adaptations were similar in MICT and HIIT groups. Our findings indicate that exercise training decreased iron storage in adults with obesity, which might have important implications for obese individuals with dysregulated iron homeostasis.
Collapse
Affiliation(s)
- Benjamin J Ryan
- Substrate Metabolism Laboratory, School of Kinesiology, University of Michigan, Ann Arbor, Michigan, USA
| | - Katherine L Foug
- Substrate Metabolism Laboratory, School of Kinesiology, University of Michigan, Ann Arbor, Michigan, USA
| | - Rachel A Gioscia-Ryan
- Substrate Metabolism Laboratory, School of Kinesiology, University of Michigan, Ann Arbor, Michigan, USA
| | - Pallavi Varshney
- Substrate Metabolism Laboratory, School of Kinesiology, University of Michigan, Ann Arbor, Michigan, USA
| | - Alison C Ludzki
- Substrate Metabolism Laboratory, School of Kinesiology, University of Michigan, Ann Arbor, Michigan, USA
| | - Cheehoon Ahn
- Substrate Metabolism Laboratory, School of Kinesiology, University of Michigan, Ann Arbor, Michigan, USA
| | - Michael W Schleh
- Substrate Metabolism Laboratory, School of Kinesiology, University of Michigan, Ann Arbor, Michigan, USA
| | - Jenna B Gillen
- Substrate Metabolism Laboratory, School of Kinesiology, University of Michigan, Ann Arbor, Michigan, USA.,Faculty of Kinesiology and Physical Education, University of Toronto, Toronto, Ontario, Canada
| | - Thomas L Chenevert
- Department of Radiology, University of Michigan, Ann Arbor, Michigan, USA
| | - Jeffrey F Horowitz
- Substrate Metabolism Laboratory, School of Kinesiology, University of Michigan, Ann Arbor, Michigan, USA
| |
Collapse
|
175
|
Han ZB, Wu J, Liu J, Li HM, Guo K, Sun T. Adult-onset Still's disease evolving with multiple organ failure and death: A case report and review of the literature. World J Clin Cases 2021; 9:886-897. [PMID: 33585636 PMCID: PMC7852636 DOI: 10.12998/wjcc.v9.i4.886] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/05/2020] [Revised: 11/28/2020] [Accepted: 12/10/2020] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Adult-onset Still’s disease (AOSD) is a rare systemic inflammatory disease, which is characterized by daily fever and arthritis, with an evanescent rash and neutrophilic leukocytosis. To date, there has been no definite laboratory or imaging test available for diagnosing AOSD; the diagnosis is one of exclusion, which can be very challenging. In particular, AOSD patients may experience different complications affecting their clinical picture, management, and prognosis. The treatment of AOSD remains largely empirical and involves therapeutic agents.
CASE SUMMARY We report the case of a 36-year-old woman who presented with fever, red rash, arthralgia, and sore throat. Her serum ferritin level and white blood cell count were markedly elevated, and the first diagnosis 22 years prior was "juvenile rheumatoid arthritis of systemic type". The patient was treated with prednisone, sulfasalazine, methotrexate, and leflunomide. After remission of her symptoms, the patient stopped taking the medications, and the disease recurred. Ultimately, the patient was diagnosed with adult-onset Still's disease. Relapse occurred several times due to self-medication withdrawal, and an interleukin-6 antagonist (tocilizumab/Actemra) was administered to control the disease. Recently, she was hospitalized because an incision did not heal, and the patient suddenly developed high fever and diarrhea during hospitalization. The patient's disease progressed violently and quickly developed into macrophage activation syndrome, disseminated intravascular coagulation, shock, and multiple organ failure. The patient had sudden cardiac arrest, and she died despite emergency rescue efforts.
CONCLUSION AOSD patients need regular follow-up in the long-term treatment process, and must press formulary standard medication, and do not voluntarily withdraw or reduce the dose. Otherwise it may cause disease back-and-forth or serious life-threatening complications. Meanwhile, strict management of trauma, infections, tumors, and other diseases may contribute to improved outcomes in patients with complications.
Collapse
Affiliation(s)
- Zhong-Bin Han
- Department of Surgery, Affiliated Zhongshan Hospital of Dalian University, Dalian 116001, Liaoning Province, China
| | - Ju Wu
- Department of Surgery, Affiliated Zhongshan Hospital of Dalian University, Dalian 116001, Liaoning Province, China
| | - Jing Liu
- Department of ICU, Affiliated Zhongshan Hospital of Dalian University, Dalian 116001, Liaoning Province, China
| | - He-Ming Li
- Department of Oncology, Affiliated Zhongshan Hospital of Dalian University, Dalian 116001, Liaoning Province, China
| | - Kai Guo
- Department of Surgery, Second People's Hospital of Jinzhong City, Jinzhong 030600, Shanxin Province, China
| | - Tong Sun
- Department of ICU, Affiliated Zhongshan Hospital of Dalian University, Dalian 116001, Liaoning Province, China
| |
Collapse
|
176
|
Tung ML, Tan B, Cherian R, Chandra B. Anti-phospholipid syndrome and COVID-19 thrombosis: connecting the dots. Rheumatol Adv Pract 2021; 5:rkaa081. [PMID: 33615129 PMCID: PMC7882149 DOI: 10.1093/rap/rkaa081] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2020] [Revised: 12/02/2020] [Indexed: 02/06/2023] Open
Abstract
As the coronavirus disease 2019 (COVID-19) pandemic, which is caused by the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), is spreading rapidly worldwide, it has emerged as a leading cause of mortality, resulting in >1 million deaths over the past 10 months. The pathophysiology of COVID-19 remains unclear, posing a great challenge to the medical management of patients. Recent studies have reported an unusually high prevalence of thromboembolic events in COVID-19 patients, although the mechanism remains elusive. Several studies have reported the presence of aPLs in COVID-19 patients. We have noticed similarities between COVID-19 and APS, which is an autoimmune prothrombotic disease that is often associated with an infective aetiology. Molecular mimicry and endothelial dysfunction could plausibly explain the mechanism of thrombogenesis in acquired APS. In this review, we discuss the clinicopathological similarities between COVID-19 and APS, and the potential role of therapeutic targets based on the anti-phospholipid model for COVID-19 disease.
Collapse
Affiliation(s)
- Moon Ley Tung
- Department of Hematology and Oncology, National University Cancer Institute
- Yong Loo Lin School of Medicine, National University of Singapore
| | - Bryce Tan
- Department of Medicine, National University Hospital
| | - Robin Cherian
- Yong Loo Lin School of Medicine, National University of Singapore
- Department of Cardiology, National University Heart Centre Singapore
| | - Bharatendu Chandra
- Yong Loo Lin School of Medicine, National University of Singapore
- Division of Neurology, Department of Medicine, National University Hospital, Singapore, Singapore
| |
Collapse
|
177
|
Zhang N, Yu X, Xie J, Xu H. New Insights into the Role of Ferritin in Iron Homeostasis and Neurodegenerative Diseases. Mol Neurobiol 2021; 58:2812-2823. [PMID: 33507490 DOI: 10.1007/s12035-020-02277-7] [Citation(s) in RCA: 112] [Impact Index Per Article: 28.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2020] [Accepted: 12/28/2020] [Indexed: 12/11/2022]
Abstract
Growing evidence has indicated that iron deposition is one of the key factors leading to neuronal death in the neurodegenerative diseases. Ferritin is a hollow iron storage protein composed of 24 subunits of two types, ferritin heavy chain (FTH) and ferritin light chain (FTL), which plays an important role in maintaining iron homeostasis. Recently, the discovery of extracellular ferritin and ferritin in exosomes indicates that ferritin might be not only an iron storage protein within the cell, but might also be an important factor in the regulation of tissue and body iron homeostasis. In this review, we first described the structural characteristics, regulation and the physiological functions of ferritin. Secondly, we reviewed the current evidence concerning the mechanisms underlying the secretion of ferritin and the possible role of secreted ferritin in the brain. Then, we summarized the relationship between ferritin and the neurodegenerative diseases including Parkinson's disease (PD), Alzheimer's disease (AD) and neuroferritinopathy (NF). Given the importance and relationship between iron and neurodegenerative diseases, understanding the role of ferritin in the brain can be expected to contribute to our knowledge of iron dysfunction and neurodegenerative diseases.
Collapse
Affiliation(s)
- Na Zhang
- Department of Physiology, Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders, School of Basic Medicine, Qingdao University, Qingdao, 266071, China.,Institute of Brain Science and Disease, Qingdao University, Qingdao, 266071, China
| | - Xiaoqi Yu
- Department of Physiology, Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders, School of Basic Medicine, Qingdao University, Qingdao, 266071, China.,Institute of Brain Science and Disease, Qingdao University, Qingdao, 266071, China
| | - Junxia Xie
- Department of Physiology, Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders, School of Basic Medicine, Qingdao University, Qingdao, 266071, China. .,Institute of Brain Science and Disease, Qingdao University, Qingdao, 266071, China.
| | - Huamin Xu
- Department of Physiology, Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders, School of Basic Medicine, Qingdao University, Qingdao, 266071, China. .,Institute of Brain Science and Disease, Qingdao University, Qingdao, 266071, China.
| |
Collapse
|
178
|
SARS-CoV-2 Mediated Hyperferritinemia and Cardiac Arrest: Preliminary Insights. Drug Discov Today 2021; 26:1265-1274. [PMID: 33493677 PMCID: PMC7826001 DOI: 10.1016/j.drudis.2021.01.014] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2020] [Revised: 12/14/2020] [Accepted: 01/08/2021] [Indexed: 02/06/2023]
|
179
|
Ma J, Qian C, Bao Y, Liu MY, Ma HM, Shen MQ, Li W, Wang JJ, Bao YX, Liu Y, Ke Y, Qian ZM. Apolipoprotein E deficiency induces a progressive increase in tissue iron contents with age in mice. Redox Biol 2021; 40:101865. [PMID: 33493903 PMCID: PMC7823209 DOI: 10.1016/j.redox.2021.101865] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2020] [Revised: 01/10/2021] [Accepted: 01/10/2021] [Indexed: 12/21/2022] Open
Abstract
Association of both iron/hepcidin and apolipoprotein E (ApoE) with development of Alzheimer disease (AD) and atherosclerosis led us to hypothesize that ApoE might be required for body iron homeostasis. Here, we demonstrated that ApoE knock-out (KO) induced a progressive accumulation of iron with age in the liver and spleen of mice. Subsequent investigations showed that the increased iron in the liver and spleen was due to phosphorylated extracellular regulated protein kinases (pERK) mediated up-regulation of transferrin receptor 1 (TfR1), and nuclear factor erythroid 2-related factor-2 (Nrf2)-dependent down-regulation of ferroportin 1. Furthermore, replenishment of ApoE could partially reverse the iron-related phenotype in ApoE KO mice. The findings imply that ApoE may be essential for body iron homeostasis and also suggest that clinical late-onset diseases with unexplained iron abnormality may partly be related to deficiency or reduced expression of ApoE. Apolipoprotein E deficiency induces a progressive increase in tissue iron contents with age in mice. ApoE−/− induced a progressive accumulation of iron with age in the liver and spleen of mice. The increased iron was due to upregulation of TfR1 and downregulation of Fpn1. Replenishment of ApoE could partially reverse the iron-related phenotype in ApoE KO mice. ApoE may be essential for body iron homeostasis.
Collapse
Affiliation(s)
- Juan Ma
- Institute of Translational and Precision Medicine, Nantong University, 19 Qi Xiu Road, Nantong, 226001, China; Laboratory of Neuropharmacology of Pharmacy School, and National Clinical Research Center for Aging and Medicine of Huashan Hospital, Fudan University, Shanghai, 201203, China
| | - Christopher Qian
- School of Biomedical Sciences and Gerald Choa Neuroscience Centre, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, NT, Hong Kong, China
| | - Yong Bao
- Institute of Translational and Precision Medicine, Nantong University, 19 Qi Xiu Road, Nantong, 226001, China
| | - Meng-Yue Liu
- Institute of Translational and Precision Medicine, Nantong University, 19 Qi Xiu Road, Nantong, 226001, China
| | - Hui-Min Ma
- Institute of Translational and Precision Medicine, Nantong University, 19 Qi Xiu Road, Nantong, 226001, China
| | - Meng-Qi Shen
- Institute of Translational and Precision Medicine, Nantong University, 19 Qi Xiu Road, Nantong, 226001, China
| | - Wei Li
- Institute of Translational and Precision Medicine, Nantong University, 19 Qi Xiu Road, Nantong, 226001, China
| | - Jiao-Jiao Wang
- Laboratory of Neuropharmacology of Pharmacy School, and National Clinical Research Center for Aging and Medicine of Huashan Hospital, Fudan University, Shanghai, 201203, China; Research Center for Medicine and Biology, Zunyi Medical University, Zunyi, 563000, Guizhou, China
| | - Yu-Xin Bao
- Research Center for Medicine and Biology, Zunyi Medical University, Zunyi, 563000, Guizhou, China
| | - Yong Liu
- Department of Pain and Rehabilitation, The Second Affiliated Hospital, The Army Medical University, Chongqing, China
| | - Ya Ke
- School of Biomedical Sciences and Gerald Choa Neuroscience Centre, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, NT, Hong Kong, China.
| | - Zhong-Ming Qian
- Institute of Translational and Precision Medicine, Nantong University, 19 Qi Xiu Road, Nantong, 226001, China; Laboratory of Neuropharmacology of Pharmacy School, and National Clinical Research Center for Aging and Medicine of Huashan Hospital, Fudan University, Shanghai, 201203, China.
| |
Collapse
|
180
|
Ishihara A, Yamauchi T, Ikeda K, Fukuyoshi Y, Yokoyama T, Yonemura Y, Uchiba M, Matsui H. Glycosylated ferritin as an improved marker for post-transfusion iron overload. Int J Hematol 2021; 113:537-546. [PMID: 33400141 DOI: 10.1007/s12185-020-03056-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2020] [Revised: 12/01/2020] [Accepted: 12/01/2020] [Indexed: 11/25/2022]
Abstract
Red blood cell (RBC) transfusion is an effective therapy for anemia, but repeated transfusions may cause iron overload-related damage to various organs. Iron chelation therapy, now widely available for patients who have received transfusions, is expected to reduce organ damage even in patients who received many transfusions. Therefore, determining when to start iron chelation therapy is important. In guidelines for iron chelation therapy, the serum ferritin level has been widely accepted as a practical marker for estimating iron overload. However, guidelines recommend multiple measurements of serum ferritin, because levels often fluctuate. Here, we investigated the usefulness of glycosylated ferritin as a marker of iron overload using a cohort consisted of 103 patients who had a total ferritin value over 1000 ng/mL. We found that the volume of RBCs transfused was clearly associated with the glycosylated ferritin level. We also found that acute inflammation, as represented by C-reactive protein values, was associated with increased non-glycosylated ferritin and that patients with hematopoietic diseases had higher glycosylated ferritin levels, possibly because of repeated RBC transfusions. We thus conclude that glycosylated ferritin may be an improved marker for predicting iron overload status.
Collapse
Affiliation(s)
- Ayako Ishihara
- Department of Clinical Laboratory Medicine, Kumamoto University Hospital, Kumamoto University, Kumamoto, Japan
| | - Tsuyuko Yamauchi
- Department of Clinical Laboratory Medicine, Kumamoto University Hospital, Kumamoto University, Kumamoto, Japan
| | - Katsuyoshi Ikeda
- Department of Medical Technology, Kumamoto Health Science University, Kumamoto, Japan
| | - Yoko Fukuyoshi
- Department of Clinical Laboratory Medicine, Kumamoto University Hospital, Kumamoto University, Kumamoto, Japan
| | - Toshiro Yokoyama
- Department of Clinical Laboratory Medicine, Kumamoto University Hospital, Kumamoto University, Kumamoto, Japan
| | - Yuji Yonemura
- Department of Transfusion Medicine and Cell Therapy, Kumamoto University Hospital, Kumamoto University, Kumamoto, Japan
- Department of Hematology, Rheumatology and Infectious Disease, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan
| | - Mitsuhiro Uchiba
- Department of Transfusion Medicine and Cell Therapy, Kumamoto University Hospital, Kumamoto University, Kumamoto, Japan
- Department of Hematology, Rheumatology and Infectious Disease, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan
| | - Hirotaka Matsui
- Department of Clinical Laboratory Medicine, Kumamoto University Hospital, Kumamoto University, Kumamoto, Japan.
- Department of Molecular Laboratory Medicine, Faculty of Life Sciences, Kumamoto University, 1-1-1 Honjo, Chuo-ku, Kumamoto, 860-8556, Japan.
| |
Collapse
|
181
|
Kim HY, Kim J, Noh E, Ahn KH, Cho GJ, Hong SC, Oh MJ, Kim HJ. Prepregnancy hemoglobin levels and gestational diabetes mellitus in pregnancy. Diabetes Res Clin Pract 2021; 171:108608. [PMID: 33310123 DOI: 10.1016/j.diabres.2020.108608] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/24/2020] [Revised: 11/15/2020] [Accepted: 12/01/2020] [Indexed: 12/16/2022]
Abstract
AIM To identify the influence of prepregnancy hemoglobin levels on gestational diabetes mellitus. MATERIALS AND METHODS Korean women who had given birth between January 1st, 2006 and December 31st, 2015 and who had undergone a biannual national health screening examination within 6 months prior to pregnancy were enrolled. Subjects were divided into three groups according to their hemoglobin levels. Multivariate logistic regression analysis was used to estimate the adjusted odds ratio and 95% confidence interval for GDM. RESULTS Of the 366,122 participants, GDM developed in 14,799 (4%) women. More specifically, GDM developed in 3.6% of women with prepregnancy anemia (hemoglobin < 11 g/dL), 3.57% with normal hemoglobin levels, and 4.47% with hemoglobin levels higher than 13 g/dL. We did not find any association between prepregnancy anemia and the risk of developing GDM (OR 1.002 [95% CI 0.90-1.11]). After adjusting for potential confounding factors (adjusted odds ratio 1.41; 95% CI 1.29-1.54), high hemoglobin levels were associated with insulin requiring GDM. CONCLUSIONS Our study identified an association between high prepregnancy hemoglobin levels and GDM risk.
Collapse
Affiliation(s)
- Ho Yeon Kim
- Department of Obstetrics and Gynecology, Korea University College of Medicine, Seoul, Republic of Korea
| | - Jinsil Kim
- Korea University Guro Hospital Smart Healthcare Center, Seoul, Republic of Korea
| | - Eunjin Noh
- Korea University Guro Hospital Smart Healthcare Center, Seoul, Republic of Korea
| | - Ki Hoon Ahn
- Department of Obstetrics and Gynecology, Korea University College of Medicine, Seoul, Republic of Korea
| | - Geum Joon Cho
- Department of Obstetrics and Gynecology, Korea University College of Medicine, Seoul, Republic of Korea.
| | - Soon-Cheol Hong
- Department of Obstetrics and Gynecology, Korea University College of Medicine, Seoul, Republic of Korea
| | - Min-Jeong Oh
- Department of Obstetrics and Gynecology, Korea University College of Medicine, Seoul, Republic of Korea
| | - Hai-Joong Kim
- Department of Obstetrics and Gynecology, Korea University College of Medicine, Seoul, Republic of Korea
| |
Collapse
|
182
|
Venkataramani V. Iron Homeostasis and Metabolism: Two Sides of a Coin. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1301:25-40. [PMID: 34370286 DOI: 10.1007/978-3-030-62026-4_3] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Iron is an ancient, essential and versatile transition metal found in almost all living organisms on Earth. This fundamental trace element is used in the synthesis of heme and iron-sulfur (Fe-S) containing proteins and other vital cofactors that are involved in respiration, redox reactions, catalysis, DNA synthesis and transcription. At the same time, the ability of iron to cycle between its oxidized, ferric (Fe3+) and its reduced, ferrous (Fe2+) state contributes to the production of free radicals that can damage biomolecules, including proteins, lipids and DNA. In particular, the regulated non-apoptotic cell death ferroptosis is driven by Fe2+-dependent lipid peroxidation that can be prevented by iron chelation or genetic inhibition of cellular iron uptake. Therefore, iron homeostasis must be tightly regulated to avoid iron toxicity. This review provides an overview of the origin and chemistry of iron that makes it suitable for a variety of biological functions and addresses how organisms evolved various strategies, including their scavenging and antioxidant machinery, to manage redox-associated drawbacks. Finally, key mechanisms of iron metabolism are highlighted in human diseases and model organisms, underlining the perils of dysfunctional iron handlings.
Collapse
Affiliation(s)
- Vivek Venkataramani
- Institute of Pathology, University Medical Center Göttingen (UMG), Göttingen, Germany.
| |
Collapse
|
183
|
Li S, Zheng L, Zhang J, Liu X, Wu Z. Inhibition of ferroptosis by up-regulating Nrf2 delayed the progression of diabetic nephropathy. Free Radic Biol Med 2021; 162:435-449. [PMID: 33152439 DOI: 10.1016/j.freeradbiomed.2020.10.323] [Citation(s) in RCA: 244] [Impact Index Per Article: 61.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Revised: 10/13/2020] [Accepted: 10/28/2020] [Indexed: 12/20/2022]
Abstract
Diabetic nephropathy (DN) is now considered the leading cause of end-stage renal disease. In diabetes, the accumulation of reactive oxygen species (ROS) and iron overload are important determinants that promote the occurrence of DN. However, the underlying mechanism of how they cause diabetic kidney damage remains unclear. Ferroptosis, characterized by iron-dependent lipid peroxidation, provided us with a new idea to explore the progression of DN. Iron overload, reduced antioxidant capability, massive ROS and lipid peroxidation were detected in the kidneys of streptozotocin-induced DBA/2J diabetic mice and high-glucose cultured human renal proximal tubular (HK-2) cells, which were the symbolic changes of ferroptosis. Furthermore, the characteristic mitochondrial morphological changes of ferroptosis were observed in high glucose cultured cells. Additional treatment of Ferrostatin-1 (Fer-1) in DN models significantly rescued these changes and alleviated the renal pathological injuries in diabetic mice. Besides, the decreased NFE2-related factor 2 (Nrf2) was observed in DN models. The specific knockdown of Nrf2 increased the sensitivity of cells to ferroptosis in the high glucose condition. In Nrf2 knockdown cells, up-regulating Nrf2 by treating with fenofibrate improved the situation of ferroptosis, which was verified in RSL-3 induced cells. Moreover, the ferroptosis-related changes were inhibited by increasing Nrf2 in fenofibrate treated diabetic mice, which delayed the progression of DN. Collectively, we demonstrated that ferroptosis was involved in the development of DN, and up-regulating Nrf2 by treating with fenofibrate inhibited diabetes-related ferroptosis, delaying the progression of DN. Our research revealed the development mechanism of DN from a new perspective, and provide a new approach delaying the progression of DN.
Collapse
Affiliation(s)
- Shuangwen Li
- NHC Key Laboratory of Hormones and Development, Tianjin Key Laboratory of Metabolic Diseases, Chu Hsien-I Memorial Hospital & Tianjin Institute of Endocrinology, Tianjin Medical University, Tianjin, 300134, China
| | - Lisi Zheng
- NHC Key Laboratory of Hormones and Development, Tianjin Key Laboratory of Metabolic Diseases, Chu Hsien-I Memorial Hospital & Tianjin Institute of Endocrinology, Tianjin Medical University, Tianjin, 300134, China
| | - Jun Zhang
- NHC Key Laboratory of Hormones and Development, Tianjin Key Laboratory of Metabolic Diseases, Chu Hsien-I Memorial Hospital & Tianjin Institute of Endocrinology, Tianjin Medical University, Tianjin, 300134, China
| | - Xuejun Liu
- Department of Neurology, Chu Hsien-I Memorial Hospital, Tianjin Medical University, Tianjin, 300134, China.
| | - Zhongming Wu
- NHC Key Laboratory of Hormones and Development, Tianjin Key Laboratory of Metabolic Diseases, Chu Hsien-I Memorial Hospital & Tianjin Institute of Endocrinology, Tianjin Medical University, Tianjin, 300134, China.
| |
Collapse
|
184
|
Outcome Predictive Value of Serum Ferritin in ICU Patients with Long ICU Stay. ACTA ACUST UNITED AC 2020; 57:medicina57010001. [PMID: 33375016 PMCID: PMC7822040 DOI: 10.3390/medicina57010001] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2020] [Revised: 12/15/2020] [Accepted: 12/21/2020] [Indexed: 12/24/2022]
Abstract
Background and Objectives: The simplified interpretation of serum ferritin levels, according to which low ferritin levels indicate iron deficiency and high levels indicate hemochromatosis is obsolete, as in the presence of inflammation serum ferritin levels, no longer correlate with iron stores. However, further data are needed to interpret serum ferritin levels correctly in patients with ongoing inflammation. Our study aimed to assess serum iron and ferritin dynamics in patients with long ICU stay and the possible correlations with organ dysfunction progression and outcome. Materials and Methods: We conducted a prospective study in a university hospital intensive care unit (ICU) over six months. All patients with an ICU length-of-stay of more than seven days were enrolled. Collected data included: demographics, Sequential Organ Failure Assessment (SOFA) score, admission, weekly serum iron and ferritin levels, ICU length-of-stay and outcome. Interactions between organ dysfunction progression and serum iron and ferritin levels changes were investigated. Outcome predictive value of serum ferritin was assessed. Results: Seventy-two patients with a mean ICU length-of-stay of 15 (4.4) days were enrolled in the study. The average age of patients was 62 (16.8) years. There were no significant differences between survivors (39 patients, 54%) and nonsurvivors (33 patients, 46%) regarding demographics, serum iron and ferritin levels and SOFA score on ICU admission. Over time, serum iron levels remained normal or low, while serum ferritin levels statedly increased in all patients. Serum ferritin increase was higher in nonsurvivors than survivors. There was a significant positive correlation between SOFA score and serum ferritin (r = 0.7, 95%CI for r = 0.64 to 0.76, p < 0.01). The predictive outcome accuracy of serum ferritin was similar to the SOFA score. Conclusions: In patients with prolonged ICU stay, serum ferritin dynamics reflects organ dysfunction progression and parallels SOFA score in terms of outcome predictive accuracy.
Collapse
|
185
|
Kasatkina MY, Zhanin IS, Gulyaeva NV. Ischemic Stroke and Depression Biomarkers: Are There Specific Markers for Post-Stroke Depression? NEUROCHEM J+ 2020. [DOI: 10.1134/s1819712420040030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
|
186
|
Fertrin KY. Diagnosis and management of iron deficiency in chronic inflammatory conditions (CIC): is too little iron making your patient sick? HEMATOLOGY. AMERICAN SOCIETY OF HEMATOLOGY. EDUCATION PROGRAM 2020; 2020:478-486. [PMID: 33275757 PMCID: PMC7727593 DOI: 10.1182/hematology.2020000132] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/20/2023]
Abstract
While iron deficiency remains the most common cause of anemia worldwide, low iron stores are associated with symptoms regardless of the presence of typical microcytic, hypochromic anemia and may be hard to recognize in patients with concurrent inflammation. Diagnosing and treating iron deficiency become more of a challenge because markers of iron status are influenced by low-grade inflammation present in common conditions, such as chronic kidney disease, cirrhosis, or heart failure. Here I present a pragmatic way of interpreting diagnostic lab tests to help clinicians recognize patients who are most likely to benefit from iron supplementation, choose between oral and parenteral administration, and make personalized decisions when patients do not fit usual guidelines.
Collapse
|
187
|
Puentes-Pardo JD, Moreno-SanJuan S, Carazo Á, León J. Heme Oxygenase-1 in Gastrointestinal Tract Health and Disease. Antioxidants (Basel) 2020; 9:antiox9121214. [PMID: 33276470 PMCID: PMC7760122 DOI: 10.3390/antiox9121214] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Revised: 11/25/2020] [Accepted: 11/29/2020] [Indexed: 12/14/2022] Open
Abstract
Heme oxygenase 1 (HO-1) is the rate-limiting enzyme of heme oxidative degradation, generating carbon monoxide (CO), free iron, and biliverdin. HO-1, a stress inducible enzyme, is considered as an anti-oxidative and cytoprotective agent. As many studies suggest, HO-1 is highly expressed in the gastrointestinal tract where it is involved in the response to inflammatory processes, which may lead to several diseases such as pancreatitis, diabetes, fatty liver disease, inflammatory bowel disease, and cancer. In this review, we highlight the pivotal role of HO-1 and its downstream effectors in the development of disorders and their beneficial effects on the maintenance of the gastrointestinal tract health. We also examine clinical trials involving the therapeutic targets derived from HO-1 system for the most common diseases of the digestive system.
Collapse
Affiliation(s)
- Jose D. Puentes-Pardo
- Research Unit, Instituto de Investigacion Biosanitaria de Granada, ibs.GRANADA, 18012 Granada, Spain
- Department of Pharmacology, Faculty of Pharmacy, University of Granada, 18011 Granada, Spain
- Correspondence: (J.D.P.-P.); (J.L.); Tel.: +34-958-023-706 (J.L.)
| | - Sara Moreno-SanJuan
- Cytometry and Microscopy Research Service, Instituto de Investigacion Biosanitaria de Granada, ibs.GRANADA, 18012 Granada, Spain;
| | - Ángel Carazo
- Genomic Research Service, Instituto de Investigacion Biosanitaria de Granada, ibs.GRANADA, 18012 Granada, Spain;
| | - Josefa León
- Research Unit, Instituto de Investigacion Biosanitaria de Granada, ibs.GRANADA, 18012 Granada, Spain
- Clinical Management Unit of Digestive Disease, San Cecilio University Hospital, 18016 Granada, Spain
- Correspondence: (J.D.P.-P.); (J.L.); Tel.: +34-958-023-706 (J.L.)
| |
Collapse
|
188
|
Serpunja S, Kim I. Supplementation of a low-energy diet with recombinant ferritin fromPerinereissp. can be beneficial to finishing pigs. CANADIAN JOURNAL OF ANIMAL SCIENCE 2020. [DOI: 10.1139/cjas-2018-0129] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
A total of 90 finishing pigs [(Yorkshire × Landrace) × Duroc] with an average body weight (BW) of 50.02 ± 1.78 kg were used in a 10 wk experiment. The pigs were distributed into three dietary treatments replicated six times with five pigs (two barrows and three gilts) per pen. The treatment diets were a positive control (PC; high-energy diet), a negative control (NC; low-energy diet), and an NC + 0.05% ferritin diet (TRT1). The supplementation of ferritin in a low-energy diet tended (P = 0.06) to increase the BW at week 5 compared with pigs fed low-energy diets without ferritin. At week 5 and overall period, the gain-to-feed ratio of pigs fed high-energy diets was higher (P < 0.05) compared with pigs fed low-energy diets. The pigs receiving a ferritin-supplemented diet had a comparable growth performance to pigs fed high-energy diets. At week 10, fecal Lactobacilli counts of pigs fed high-energy diets were higher (P < 0.05) compared with pigs fed low-energy diets. The supplementation of low-energy diets with ferritin resulted in comparable growth performance to pigs fed high-energy diets and had no adverse effect on digestibility and fecal gas emissions. Thus, it seems beneficial to include ferritin in low-energy diets of finishing pigs.
Collapse
Affiliation(s)
- S. Serpunja
- Department of Animal Resource and Science, Dankook University, 119, Dandae-ro, Dongnam-gu, Cheonan-si, Chungam 31116, South Korea
- Department of Animal Resource and Science, Dankook University, 119, Dandae-ro, Dongnam-gu, Cheonan-si, Chungam 31116, South Korea
| | - I.H. Kim
- Department of Animal Resource and Science, Dankook University, 119, Dandae-ro, Dongnam-gu, Cheonan-si, Chungam 31116, South Korea
- Department of Animal Resource and Science, Dankook University, 119, Dandae-ro, Dongnam-gu, Cheonan-si, Chungam 31116, South Korea
| |
Collapse
|
189
|
Simão M, Leite RB, Cancela ML. Expression of four new ferritins from grooved carpet shell clam Ruditapes decussatus challenged with Perkinsus olseni and metals (Cd, Cu and Zn). AQUATIC TOXICOLOGY (AMSTERDAM, NETHERLANDS) 2020; 229:105675. [PMID: 33197689 DOI: 10.1016/j.aquatox.2020.105675] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/27/2020] [Revised: 10/30/2020] [Accepted: 10/31/2020] [Indexed: 06/11/2023]
Abstract
Iron has a fundamental role in life and in its biochemical reactions but, when in excess, it can promote the formation of free radicals which can lead to cell death. Therefore, managing the levels of iron is essential to regulate the production of oxidative stress related to iron, and ferritins are one of the main protein families involved in this process. Ferritins are ≈480 kDa multimeric proteins composed by 24 subunits, each with 19-26 kDa, which can accumulate up to 4500 iron atoms. Besides their role in managing iron bioavailability, they have also developed a role in organism immunity and defence present throughout evolution. In this work, we identified and characterized, for the first time, four different ferritin subunits in the clam Ruditapes decussatus, a bivalve commercially and ecologically important along the south Atlantic coast and in the Mediterranean basin, which is a major target of the parasitic protozoa Perkinsus olseni, considered one of the main causes of high levels of clam mortality. Following phylogenetic annotation, the four ferritins subunits identified were subdivided into two cytosolic and two secreted forms. All four subunits maintain the canonical ferritin structure with four main helices α (A-D) and a small helix (E), but the secreted ferritins present an additional helix in their N-terminal region (F), located after the signal peptide and with possible antimicrobial properties. Additionally, we identified in ferritin 4 an extra helix α (G) located between helices B and C. These alpha helix domains revealed high degree of similarity with antimicrobial peptides associated with antibacterial and antifungal activities. Analysis of the expression of these subunits showed that ferritins 1 and 2 are ubiquitously expressed while ferritins 3 and 4 are present mainly in visceral mass. Ferritin 1 lacked a putative functional iron response element (IRE) and appeared to be under a tight regulation. Ferritins 2 and 3 showed a strong response to infection by parasite Perkinsus olseni in contrast to ferritin 4, whose main response was related to exposure to a combination of metals. The synergistic effect between metals and infection promoted a general upregulation of the four ferritins. In conclusion, our results suggest that ferritins, besides their function in iron and metals detoxification, may play a determinant role in clam immune response.
Collapse
Affiliation(s)
- Márcio Simão
- Centre of Marine Sciences (CCMAR), Universidade do Algarve, Faro, Portugal; Department of Biomedical Sciences and Medicine (DCBM), Universidade do Algarve, Faro, Portugal.
| | - Ricardo B Leite
- Centre of Marine Sciences (CCMAR), Universidade do Algarve, Faro, Portugal
| | - M Leonor Cancela
- Centre of Marine Sciences (CCMAR), Universidade do Algarve, Faro, Portugal; Department of Biomedical Sciences and Medicine (DCBM), Universidade do Algarve, Faro, Portugal; Algarve Biomedical Center (ABC) and Center for Biomedical Research (CBMR), Universidade do Algarve, Campus de Gambelas, 8005-139, Faro, Portugal
| |
Collapse
|
190
|
Iron Dysregulation in Human Cancer: Altered Metabolism, Biomarkers for Diagnosis, Prognosis, Monitoring and Rationale for Therapy. Cancers (Basel) 2020; 12:cancers12123524. [PMID: 33255972 PMCID: PMC7761132 DOI: 10.3390/cancers12123524] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2020] [Revised: 11/21/2020] [Accepted: 11/24/2020] [Indexed: 02/07/2023] Open
Abstract
Simple Summary Iron is the more abundant metal ion in humans. It is essential for life as it has a role in various cellular processes involved, for instance, in cell metabolism and DNA synthesis. These functions are crucial for cell proliferation, and it is therefore not surprising that iron is accumulated in tumors. In this review, we describe normal and altered iron homeostasis mechanisms. We also provide a vision of iron-related proteins with altered expression in cancers and discuss their potential as diagnostic and/or prognostic biomarkers. Finally, we give an overview of therapeutic strategies acting on iron metabolism to fight against cancers. Abstract Iron (Fe) is a trace element that plays essential roles in various biological processes such as DNA synthesis and repair, as well as cellular energy production and oxygen transport, and it is currently widely recognized that iron homeostasis is dysregulated in many cancers. Indeed, several iron homeostasis proteins may be responsible for malignant tumor initiation, proliferation, and for the metastatic spread of tumors. A large number of studies demonstrated the potential clinical value of utilizing these deregulated proteins as prognostic and/or predictive biomarkers of malignancy and/or response to anticancer treatments. Additionally, the iron present in cancer cells and the importance of iron in ferroptosis cell death signaling pathways prompted the development of therapeutic strategies against advanced stage or resistant cancers. In this review, we select relevant and promising studies in the field of iron metabolism in cancer research and clinical oncology. Besides this, we discuss some co-existing discrepant findings. We also present and discuss the latest lines of research related to targeting iron, or its regulatory pathways, as potential promising anticancer strategies for human therapy. Iron chelators, such as deferoxamine or iron-oxide-based nanoparticles, which are already tested in clinical trials, alone or in combination with chemotherapy, are also reported.
Collapse
|
191
|
Aher A, Udhwani T, Khandelwal R, Limaye A, Hussain T, Nayarisseri A, Singh SK. In silico Insights on IL-6: A Potential Target for Multicentric Castleman Disease. Curr Comput Aided Drug Des 2020; 16:641-653. [DOI: 10.2174/1573409915666190902142524] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2019] [Revised: 06/01/2019] [Accepted: 07/11/2019] [Indexed: 12/29/2022]
Abstract
Background:
Multicentric Castleman Disease (MCD) is a confrontational lymphoproliferative
disorder described by symptoms such as lymph node proliferation, unwarranted secretion of
inflammatory cytokines, hyperactive immune system, and in severe cases, multiple organ dysfunction.
Interleukin-6 (IL-6) is a pleiotropic cytokine which is involved in a large range of physiological
processes in our body such as pro-inflammation, anti-inflammation, differentiation of T-cells
and is reported to be a key pathological factor in MCD. In the case of MCD, it was observed that
IL-6 is overproduced from T-cells and macrophages which disturb Hepcidin, a vital regulator of
iron trafficking in macrophage. The present study endeavour to expound the inhibitor which binds
to IL-6 protein receptor with high affinity.
Methods:
MolegroVirtual Docker software was employed to find the best-established drug from
the list of selected inhibitors of IL-6. This compound was subjected to virtual screening against
PubChem database to get inhibitors with a very similar structure. These inhibitors were docked to
obtain a compound binding with high affinity to the target protein. The established compound and
the virtual screened compound were subjected to relative analysis of interactivity energy variables
and ADMET profile studies.
Results:
Among all the selected inhibitors, the virtual screened compound PubChem CID:
101119084 is seen to possess the highest affinity with the target protein. Comparative studies and
ADMET analysis further implicate this compound as a better inhibitor of the IL-6 protein.
Conclusion:
Hence, this compound recognized in the study possesses high potential as an IL-6 inhibitor
which might assist in the treatment of Multicentric Castleman Disease and should be examined
for its efficiency by in vivo studies.
Collapse
Affiliation(s)
- Abhishek Aher
- In Silico Research Laboratory, Eminent Biosciences, Mahalakshmi Nagar, Indore – 452010, Madhya Pradesh, India
| | - Trishang Udhwani
- In Silico Research Laboratory, Eminent Biosciences, Mahalakshmi Nagar, Indore – 452010, Madhya Pradesh, India
| | - Ravina Khandelwal
- In Silico Research Laboratory, Eminent Biosciences, Mahalakshmi Nagar, Indore – 452010, Madhya Pradesh, India
| | - Akanksha Limaye
- In Silico Research Laboratory, Eminent Biosciences, Mahalakshmi Nagar, Indore – 452010, Madhya Pradesh, India
| | - Tajamul Hussain
- Center of Excellence in Biotechnology Research, College of Science, King Saud University, Riyadh, Saudi Arabia
| | - Anuraj Nayarisseri
- In Silico Research Laboratory, Eminent Biosciences, Mahalakshmi Nagar, Indore – 452010, Madhya Pradesh, India
| | - Sanjeev Kumar Singh
- Computer Aided Drug Designing and Molecular Modeling Lab, Department of Bioinformatics, Alagappa University, Karaikudi-630 003, Tamil Nadu, India
| |
Collapse
|
192
|
Líšková S, Bališ P, Mičurová A, Kluknavský M, Okuliarová M, Puzserová A, Škrátek M, Sekaj I, Maňka J, Valovič P, Bernátová I. Effect of iron oxide nanoparticles on vascular function and nitric oxide production in acute stress-exposed rats. Physiol Res 2020; 69:1067-1083. [PMID: 33129250 DOI: 10.33549/physiolres.934567] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
We investigated whether polyethylene glycol-coated Fe3O4 nanoparticles (IONs), acute stress and their combination modifies vascular functions, nitric oxide synthase (NOS) activity, mean arterial pressure (MAP) as well as hepcidin and ferritin H gene expressions in Wistar-Kyoto rats. Rats were divided into control, ION-treated rats (1 mg Fe/kg i.v.), repeated acute air-jet stress-exposed rats and IONs-and-stress co-exposed rats. Maximal acetylcholine (ACh)-induced and sodium nitroprusside (SNP)-induced relaxations in the femoral arteries did not differ among the groups. IONs alone significantly elevated the N?-nitro-L-arginine methyl ester (L-NAME)-sensitive component of ACh-induced relaxation and reduced the sensitivity of vascular smooth muscle cells to SNP. IONs alone also elevated NOS activity in the brainstem and hypothalamus, reduced NOS activity in the kidneys and had no effect in the liver. Acute stress alone failed to affect vascular function and NOS activities in all the tissues investigated but it elevated ferritin H expression in the liver. In the ION-and-stress group, NOS activity was elevated in the kidneys and liver, but reduced in the brainstem and hypothalamus vs. IONs alone. IONs also accentuated air-jet stress-induced MAP responses vs. stress alone. Interestingly, stress reduced ION-originated iron content in blood and liver while it was elevated in the kidneys. In conclusion, the results showed that 1) acute administration of IONs altered vascular function, increased L-NAME-sensitive component of ACh-induced relaxation and had tissue-dependent effects on NOS activity, 2) ION effects were considerably reduced by co-exposure to repeated acute stress, likely related to decrease of ION-originated iron in blood due to elevated decomposition and/or excretion.
Collapse
Affiliation(s)
- S Líšková
- Institute of Pharmacology and Clinical Pharmacology, Faculty of Medicine, Comenius University, Bratislava, Slovakia, , and Institute of Normal and Pathological Physiology, Centre of Experimental Medicine, Slovak Academy of Sciences, Bratislava, Slovakia,
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
193
|
Khoshlahni N, Sagha M, Mirzapour T, Zarif MN, Mohammadzadeh-Vardin M. Iron depletion with deferoxamine protects bone marrow-derived mesenchymal stem cells against oxidative stress-induced apoptosis. Cell Stress Chaperones 2020; 25:1059-1069. [PMID: 32729002 PMCID: PMC7591652 DOI: 10.1007/s12192-020-01142-9] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2020] [Revised: 07/12/2020] [Accepted: 07/20/2020] [Indexed: 01/11/2023] Open
Abstract
Bone marrow mesenchymal stem cells (BM-MSCs) are multipotent cells with self-renewal properties, making them an ideal candidate for regenerative medicine. Recently, numerous studies show that about more than 99% of transplanted cells are destroyed because of the stressful microenvironment. Meanwhile, in the target organs, iron overload can produce oxidative stress introducing it as the most important stress factor. The present study was aimed at increasing BM-MSCs' viability against oxidative stress microenvironment using iron depletion by deferoxamine (DFO). Mesenchymal stem cells are isolated and characterized from rat bone marrow. Then, the sensitivity of BM-MSCs against H2O2-induced oxidative stress was evaluated through half of the inhibitory concentration (IC50) estimation by using MTT assay. The maximum non-inhibitory concentration of DFO on BM-MSCs was determined. The next step was the comparison between DFO pre-treated BM-MSCs and untreated cells against H2O2-induced apoptosis. BM-MSCs were identified with morphologic and flow cytometry analysis. IC50 of H2O2 was determined as 0.55 mM at 4 h. Also, the maximum non-inhibitory concentration of DFO was ascertained as 5 μM at 48 h. Our results demonstrated that pretreatment with DFO significantly potentiates BM-MSCs against H2O2-induced oxidative stress which was confirmed by MTT assay, AO/EB double staining, DAPI staining, and activated caspase 3 quantification as well as western blot test. Expression of cleaved caspase 3 and pAKT/AKT ratio obviously demonstrated DFO can resist the cells against cytotoxicity. These findings may help to develop better stem cell culture medium for MSC-based cell therapy. Moreover, regulation of cell stress can be used in practical subjects.
Collapse
Affiliation(s)
- Nasrin Khoshlahni
- Department of Biology, Faculty of Science, University of Mohaghegh Ardabili, Ardabil, Iran
- Research Laboratory for Embryology and Stem Cells, Department of Anatomical Sciences, School of Medicine, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Mohsen Sagha
- Research Laboratory for Embryology and Stem Cells, Department of Anatomical Sciences, School of Medicine, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Tooba Mirzapour
- Department of Biology, Faculty of Science, University of Guilan, Rasht, Iran
| | - Mahin Nikougoftar Zarif
- Blood Transfusion Research Center, High Institute for Research and Education in Transfusion Medicine, Tehran, Iran
| | - Mohammad Mohammadzadeh-Vardin
- Research Laboratory for Embryology and Stem Cells, Department of Anatomical Sciences, School of Medicine, Ardabil University of Medical Sciences, Ardabil, Iran.
| |
Collapse
|
194
|
Ulagesan S, Choi JW, Nam TJ, Choi YH. Characterization of recombinant protein ferritin from Pyropia yezoensis (rPyFer) and its biological activities. Food Sci Biotechnol 2020; 29:1501-1509. [PMID: 33088599 DOI: 10.1007/s10068-020-00821-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2020] [Revised: 08/09/2020] [Accepted: 08/31/2020] [Indexed: 11/29/2022] Open
Abstract
Ferritins are iron-binding proteins that are basically participated in iron storage, detoxification, and immune response. In the present study, ferritin gene from the marine red algae Pyropia yezoensis was cloned into a pET21d expression vector. High-efficiency transformation was performed in Escherichia coli BL21, the recombinant protein was expressed by induction with 0.1 mM isopropyl-β-D-thiogalactoside and purified via ammonium sulfate precipitation, anion exchange and size exclusion chromatography. The purified recombinant ferritin from P. yezoensis (rPyFer) was characterized and analyzed for its antimicrobial activity against both Gram-negative and Gram-positive bacterial cultures and exhibited significant antibacterial activity against Gram-positive cultures. The recombinant protein was also analyzed for its iron-uptake and radical-scavenging activities; rPyFer exhibited significant iron-uptake activity at low concentrations, and its radical-scavenging activity increased in a dose-dependent manner. This research will contribute to the development of new therapeutic proteins from marine algae.
Collapse
Affiliation(s)
- Selvakumari Ulagesan
- Institute of Fisheries Sciences, Pukyong National University, Busan, 46041 Republic of Korea
| | - Jeong-Wook Choi
- Institute of Fisheries Sciences, Pukyong National University, Busan, 46041 Republic of Korea
| | - Taek-Jeong Nam
- Institute of Fisheries Sciences, Pukyong National University, Busan, 46041 Republic of Korea
| | - Youn-Hee Choi
- Institute of Fisheries Sciences, Pukyong National University, Busan, 46041 Republic of Korea.,Department of Marine Bio-materials and Aquaculture, Pukyong National University, 45, Yongso-ro, Nam-Gu, Busan, 48513 Republic of Korea
| |
Collapse
|
195
|
Shibabaw T, Teferi B, Molla MD, Ayelign B. Inflammation Mediated Hepcidin-Ferroportin Pathway and Its Therapeutic Window in Breast Cancer. BREAST CANCER-TARGETS AND THERAPY 2020; 12:165-180. [PMID: 33116818 PMCID: PMC7585830 DOI: 10.2147/bctt.s276404] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/10/2020] [Accepted: 10/12/2020] [Indexed: 12/16/2022]
Abstract
Experimental and clinical data strongly support that iron is an essential element which plays a big role in cancer biology. Thus, hepcidin (Hp) and ferroportin (Fpn) are molecules that regulate and maintain the metabolism of iron. A peptide hormone hepcidin limits recycled and stored iron fluxes in macrophage and hepatic hepatocyte, respectively, to the blood stream by promoting degradation of the only iron exporter, Fpn, in the target cells. Moreover, the inflammatory microenvironment of breast cancer and altered hepcidin/ferroportin pathway is intimately linked. Breast cancer exhibits an iron seeking phenotype that is accomplished by tumor-associated macrophage (TAM). Because macrophages contribute to breast cancer growth and progression, this review will discuss TAM with an emphasis on describing how TAM (M2Ф phenotypic) interacts with their surrounding microenvironment and results in dysregulated Hp/Fpn and pathologic accumulation of iron as a hallmark of its malignant condition. Moreover, the underlying stroma or tumor microenvironment releases significant inflammatory cytokines like IL-6 and bone morphogenetic proteins like BMP-2 and 6 leading in aberrant Hp/Fpn pathways in breast cancer. Inflammation is primarily associated with the high intracellular iron levels, deregulated hepcidin/ferroportin pathway, and its upstream signaling in breast cancer. Subsequently, scholars have been reported that reducing iron level and manipulating the signaling molecules involved in iron metabolism can be used as a promising strategy of tumor chemotherapy. Here, we review the key molecular aspects of iron metabolism and its regulatory mechanisms of the hepcidin/ferroportin pathways and its current therapeutic strategies in breast cancer.
Collapse
Affiliation(s)
- Tewodros Shibabaw
- Department of Biochemistry, School of Medicine, College of Medicine and Health Sciences, University of Gondar, Gondar, Ethiopia
| | - Banchamlak Teferi
- Department of Clinical Pharmacy, School of Pharmacy, College of Medicine and Health Sciences, University of Gondar, Gondar, Ethiopia
| | - Meseret Derbew Molla
- Department of Biochemistry, School of Medicine, College of Medicine and Health Sciences, University of Gondar, Gondar, Ethiopia
| | - Birhanu Ayelign
- Department of Immunology and Molecular Biology, School of Biomedical and Laboratory Sciences, College of Medicine and Health Sciences, University of Gondar, Gondar, Ethiopia
| |
Collapse
|
196
|
Bednarz A, Lipiński P, Starzyński RR, Tomczyk M, Kraszewska I, Herman S, Kowalski K, Gruca E, Jończy A, Mazgaj R, Szudzik M, Rajfur Z, Baster Z, Józkowicz A, Lenartowicz M. Exacerbation of Neonatal Hemolysis and Impaired Renal Iron Handling in Heme Oxygenase 1-Deficient Mice. Int J Mol Sci 2020; 21:ijms21207754. [PMID: 33092142 PMCID: PMC7589678 DOI: 10.3390/ijms21207754] [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] [Subscribe] [Scholar Register] [Received: 09/22/2020] [Revised: 10/15/2020] [Accepted: 10/16/2020] [Indexed: 01/03/2023] Open
Abstract
In most mammals, neonatal intravascular hemolysis is a benign and moderate disorder that usually does not lead to anemia. During the neonatal period, kidneys play a key role in detoxification and recirculation of iron species released from red blood cells (RBC) and filtered out by glomeruli to the primary urine. Activity of heme oxygenase 1 (HO1), a heme-degrading enzyme localized in epithelial cells of proximal tubules, seems to be of critical importance for both processes. We show that, in HO1 knockout mouse newborns, hemolysis was prolonged despite a transient state and exacerbated, which led to temporal deterioration of RBC status. In neonates lacking HO1, functioning of renal molecular machinery responsible for iron reabsorption from the primary urine (megalin/cubilin complex) and its transfer to the blood (ferroportin) was either shifted in time or impaired, respectively. Those abnormalities resulted in iron loss from the body (excreted in urine) and in iron retention in the renal epithelium. We postulate that, as a consequence of these abnormalities, a tight systemic iron balance of HO1 knockout neonates may be temporarily affected.
Collapse
Affiliation(s)
- Aleksandra Bednarz
- Department of Genetics and Evolution, Institute of Zoology and Biomedical Research, Jagiellonian University, Gronostajowa 9, 30-387 Kraków, Poland; (A.B.); (S.H.); (K.K.); (E.G.)
| | - Paweł Lipiński
- Department of Molecular Biology, Institute of Genetics and Animal Biotechnology, Polish Academy of Sciences, Jastrzębiec, 05-552 Magdalenka, Poland; (P.L.); (R.R.S.); (A.J.); (R.M.); (M.S.)
| | - Rafał R. Starzyński
- Department of Molecular Biology, Institute of Genetics and Animal Biotechnology, Polish Academy of Sciences, Jastrzębiec, 05-552 Magdalenka, Poland; (P.L.); (R.R.S.); (A.J.); (R.M.); (M.S.)
| | - Mateusz Tomczyk
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Gronostajowa 7, 30-387 Kraków, Poland; (M.T.); (I.K.); (A.J.)
| | - Izabela Kraszewska
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Gronostajowa 7, 30-387 Kraków, Poland; (M.T.); (I.K.); (A.J.)
| | - Sylwia Herman
- Department of Genetics and Evolution, Institute of Zoology and Biomedical Research, Jagiellonian University, Gronostajowa 9, 30-387 Kraków, Poland; (A.B.); (S.H.); (K.K.); (E.G.)
| | - Kacper Kowalski
- Department of Genetics and Evolution, Institute of Zoology and Biomedical Research, Jagiellonian University, Gronostajowa 9, 30-387 Kraków, Poland; (A.B.); (S.H.); (K.K.); (E.G.)
| | - Ewelina Gruca
- Department of Genetics and Evolution, Institute of Zoology and Biomedical Research, Jagiellonian University, Gronostajowa 9, 30-387 Kraków, Poland; (A.B.); (S.H.); (K.K.); (E.G.)
| | - Aneta Jończy
- Department of Molecular Biology, Institute of Genetics and Animal Biotechnology, Polish Academy of Sciences, Jastrzębiec, 05-552 Magdalenka, Poland; (P.L.); (R.R.S.); (A.J.); (R.M.); (M.S.)
| | - Rafał Mazgaj
- Department of Molecular Biology, Institute of Genetics and Animal Biotechnology, Polish Academy of Sciences, Jastrzębiec, 05-552 Magdalenka, Poland; (P.L.); (R.R.S.); (A.J.); (R.M.); (M.S.)
| | - Mateusz Szudzik
- Department of Molecular Biology, Institute of Genetics and Animal Biotechnology, Polish Academy of Sciences, Jastrzębiec, 05-552 Magdalenka, Poland; (P.L.); (R.R.S.); (A.J.); (R.M.); (M.S.)
| | - Zenon Rajfur
- Department of Molecular and Interfacial Biophysics, Faculty of Physics, Astronomy and Applied Computer Science, Jagiellonian University, Łojasiewicza 11, 30-348 Kraków, Poland; (Z.R.); (Z.B.)
| | - Zbigniew Baster
- Department of Molecular and Interfacial Biophysics, Faculty of Physics, Astronomy and Applied Computer Science, Jagiellonian University, Łojasiewicza 11, 30-348 Kraków, Poland; (Z.R.); (Z.B.)
| | - Alicja Józkowicz
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Gronostajowa 7, 30-387 Kraków, Poland; (M.T.); (I.K.); (A.J.)
| | - Małgorzata Lenartowicz
- Department of Genetics and Evolution, Institute of Zoology and Biomedical Research, Jagiellonian University, Gronostajowa 9, 30-387 Kraków, Poland; (A.B.); (S.H.); (K.K.); (E.G.)
- Correspondence:
| |
Collapse
|
197
|
Kallianpur AR, Wen W, Erwin AL, Clifford DB, Hulgan T, Robbins GK. Higher iron stores and the HFE 187C>G variant delay onset of peripheral neuropathy during combination antiretroviral therapy. PLoS One 2020; 15:e0239758. [PMID: 33057367 PMCID: PMC7561201 DOI: 10.1371/journal.pone.0239758] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2020] [Accepted: 09/13/2020] [Indexed: 02/07/2023] Open
Abstract
OBJECTIVE People with HIV (PWH) continue to experience sensory neuropathy and neuropathic pain in the combination antiretroviral therapy (cART) era for unclear reasons. This study evaluated the role of iron in a previously reported association of iron-loading hemochromatosis (HFE) gene variants with reduced risk of neuropathy in PWH who received more neurotoxic cART, since an iron-related mechanism also might be relevant to neuropathic symptoms in PWH living in low-resource settings today. DESIGN This time-to-event analysis addressed the impact of systemic iron levels on the rapidity of neuropathy onset in PWH who initiated cART. METHODS Soluble transferrin receptor (sTFR), the sTFR-ferritin index of iron stores, and high-sensitivity C-reactive protein (hsCRP) levels were determined in stored baseline sera from participants of known HFE genotype from AIDS Clinical Trials Group (ACTG) Study 384, a multicenter randomized clinical trial that evaluated cART strategies. Associations with incident neuropathy were evaluated in proportional-hazards, time-to-event regression models, adjusting for potential confounders. RESULTS Of 151 eligible participants with stored serum who were included in the original genetic study, 43 had cART-associated neuropathy; 108 had sufficient serum for analysis, including 30 neuropathy cases. Carriers of HFE variants had higher systemic iron (lower sTFR and sTFR-ferritin index) and lower hsCRP levels than non-carriers (all p<0.05). Higher sTFR or iron stores, the HFE 187C>G variant, and lower baseline hsCRP were associated with significantly delayed neuropathy in self-reported whites (n = 28; all p-values<0.05), independent of age, CD4+ T-cell count, plasma HIV RNA, and cART regimen. CONCLUSIONS Higher iron stores, the HFE 187C>G variant, and lower hsCRP predicted delayed onset of neuropathy among self-reported white individuals initating cART. These findings require confirmation but may have implications for cART in HIV+ populations in areas with high endemic iron deficiency, especially those PWH in whom older, more neurotoxic antiretroviral drugs are occasionally still used.
Collapse
Affiliation(s)
- Asha R. Kallianpur
- Genomic Medicine Institute, Cleveland Clinic/Lerner Research Institute, Cleveland, Ohio, United States of America
- Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland, Ohio, United States of America
- Department of Population and Quantitative Health Sciences, Case Western Reserve University School of Medicine, Cleveland, Ohio, United States of America
- * E-mail:
| | - Wanqing Wen
- Division of Epidemiology, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, United States of America
| | - Angelika L. Erwin
- Genomic Medicine Institute, Cleveland Clinic/Lerner Research Institute, Cleveland, Ohio, United States of America
| | - David B. Clifford
- Division of Infectious Diseases, Departments of Medicine and Neurology, Washington University School of Medicine, Saint Louis, Missouri, United States of America
| | - Todd Hulgan
- Division of Infectious Diseases, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, United States of America
| | - Gregory K. Robbins
- Division of Infectious Diseases, Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts, United States of America
| |
Collapse
|
198
|
Tian Y, Lu J, Hao X, Li H, Zhang G, Liu X, Li X, Zhao C, Kuang W, Chen D, Zhu M. FTH1 Inhibits Ferroptosis Through Ferritinophagy in the 6-OHDA Model of Parkinson's Disease. Neurotherapeutics 2020; 17:1796-1812. [PMID: 32959272 PMCID: PMC7851296 DOI: 10.1007/s13311-020-00929-z] [Citation(s) in RCA: 264] [Impact Index Per Article: 52.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/04/2020] [Indexed: 02/08/2023] Open
Abstract
Parkinson's disease (PD) is a neurodegenerative disorder characterized by degeneration of dopaminergic neurons associated with dysregulation of iron homeostasis in the brain. Ferroptosis is an iron-dependent cell death process that serves as a significant regulatory mechanism in PD. However, its underlying mechanisms are not yet fully understood. By performing RNA sequencing analysis, we found that the main iron storage protein ferritin heavy chain 1 (FTH1) is differentially expressed in the rat 6-hydroyxdopamine (6-OHDA) model of PD compared with control rats. Our present work demonstrates that FTH1 is involved in iron accumulation and the ferroptosis pathway in this model. Knockdown of FTH1 in PC-12 cells significantly inhibited cell viability and caused mitochondrial dysfunction. Moreover, FTH1 was found to be involved in ferritinophagy, a selective form of autophagy involving the degradation of ferritin by ferroptosis. Overexpression of FTH1 in PC-12 cells impaired ferritinophagy and downregulated microtubule-associated protein light chain 3 and nuclear receptor coactivator 4 expression, ultimately suppressing cell death induced by ferroptosis. Consistent with these findings, the ferritinophagy inhibitors chloroquine and bafilomycin A1 inhibited ferritin degradation and ferroptosis in 6-OHDA-treated PC-12 cells. This entire process was mediated by the cyclic regulation of FTH1 and ferritinophagy. Taken together, these results suggest that FTH1 links ferritinophagy and ferroptosis in the 6-OHDA model of PD, and provide a new perspective and potential for a pharmacological target in this disease.
Collapse
Affiliation(s)
- Ye Tian
- Shenzhen Bao'an Traditional Chinese Medicine Hospital (Group), Guangzhou University of Chinese Medicine, Shenzhen, 518133, China
| | - Juan Lu
- Shenzhen Hospital of Southern Medical University, Shenzhen, 518000, China
| | - Xiaoqian Hao
- Shenzhen Hospital of Integrated Traditional Chinese and Western Medicine, Guangzhou University of Chinese Medicine, Shenzhen, 518104, China
| | - Hang Li
- Shenzhen Bao'an Traditional Chinese Medicine Hospital (Group), Guangzhou University of Chinese Medicine, Shenzhen, 518133, China
| | - Guiyu Zhang
- Shenzhen Bao'an Traditional Chinese Medicine Hospital (Group), Guangzhou University of Chinese Medicine, Shenzhen, 518133, China
| | - Xuelei Liu
- Shenzhen Bao'an Traditional Chinese Medicine Hospital (Group), Guangzhou University of Chinese Medicine, Shenzhen, 518133, China
| | - Xinrong Li
- Shenzhen Bao'an Traditional Chinese Medicine Hospital (Group), Guangzhou University of Chinese Medicine, Shenzhen, 518133, China
| | - Caiping Zhao
- Shenzhen Bao'an Traditional Chinese Medicine Hospital (Group), Guangzhou University of Chinese Medicine, Shenzhen, 518133, China
| | - Weihong Kuang
- The Second Clinical Medical College, Guangdong Medical University, Dongguan, 524023, China
| | - Dongfeng Chen
- The Research Center of Basic Integrative Medicine, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China.
| | - Meiling Zhu
- Shenzhen Hospital of Integrated Traditional Chinese and Western Medicine, Guangzhou University of Chinese Medicine, Shenzhen, 518104, China.
| |
Collapse
|
199
|
Nairz M, Weiss G. Iron in infection and immunity. Mol Aspects Med 2020; 75:100864. [PMID: 32461004 DOI: 10.1016/j.mam.2020.100864] [Citation(s) in RCA: 199] [Impact Index Per Article: 39.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2020] [Revised: 04/25/2020] [Accepted: 05/05/2020] [Indexed: 12/12/2022]
Abstract
Iron is an essential micronutrient for virtually all living cells. In infectious diseases, both invading pathogens and mammalian cells including those of the immune system require iron to sustain their function, metabolism and proliferation. On the one hand, microbial iron uptake is linked to the virulence of most human pathogens. On the other hand, the sequestration of iron from bacteria and other microorganisms is an efficient strategy of host defense in line with the principles of 'nutritional immunity'. In an acute infection, host-driven iron withdrawal inhibits the growth of pathogens. Chronic immune activation due to persistent infection, autoimmune disease or malignancy however, sequesters iron not only from infectious agents, autoreactive lymphocytes and neoplastic cells but also from erythroid progenitors. This is one of the key mechanisms which collectively result in the anemia of chronic inflammation. In this review, we highlight the most important interconnections between iron metabolism and immunity, focusing on host defense against relevant infections and on the clinical consequences of anemia of inflammation.
Collapse
Affiliation(s)
- Manfred Nairz
- Department of Internal Medicine II, Infectious Diseases, Immunology, Rheumatology, Pneumology, Medical University of Innsbruck, Austria
| | - Günter Weiss
- Department of Internal Medicine II, Infectious Diseases, Immunology, Rheumatology, Pneumology, Medical University of Innsbruck, Austria; Christian Doppler Laboratory for Iron Metabolism and Anemia Research, Medical University of Innsbruck, Austria.
| |
Collapse
|
200
|
Cheng L, Li H, Li L, Liu C, Yan S, Chen H, Li Y. Ferritin in the coronavirus disease 2019 (COVID-19): A systematic review and meta-analysis. J Clin Lab Anal 2020; 34:e23618. [PMID: 33078400 PMCID: PMC7595919 DOI: 10.1002/jcla.23618] [Citation(s) in RCA: 202] [Impact Index Per Article: 40.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2020] [Revised: 09/21/2020] [Accepted: 09/24/2020] [Indexed: 12/15/2022] Open
Abstract
OBJECTIVE The coronavirus disease 2019 (COVID-19) has rapidly developed into a pandemic. Increased levels of ferritin due to cytokine storm and secondary hemophagocytic lymphohistiocytosis were found in severe COVID-19 patients. Therefore, the aim of this study was to determine the role of ferritin in COVID-19. METHODS Studies investigating ferritin in COVID-19 were collected from PubMed, EMBASE, CNKI, SinoMed, and WANFANG. A meta-analysis was performed to compare the ferritin level between different patient groups: non-survivors versus survivors; more severe versus less severe; with comorbidity versus without comorbidity; ICU versus non-ICU; with mechanical ventilation versus without mechanical ventilation. RESULTS A total of 52 records involving 10 614 COVID-19-confirmed patients between December 25, 2019, and June 1, 2020, were included in this meta-analysis, and 18 studies were included in the qualitative synthesis. The ferritin level was significantly increased in severe patients compared with the level in non-severe patients [WMD 397.77 (95% CI 306.51-489.02), P < .001]. Non-survivors had a significantly higher ferritin level compared with the one in survivors [WMD 677.17 (95% CI 391.01-963.33), P < .001]. Patients with one or more comorbidities including diabetes, thrombotic complication, and cancer had significantly higher levels of ferritin than those without (P < .01). Severe acute liver injury was significantly associated with high levels of ferritin, and its level was associated with intensive supportive care, including ICU transfer and mechanical ventilation. CONCLUSIONS Ferritin was associated with poor prognosis and could predict the worsening of COVID-19 patients.
Collapse
Affiliation(s)
- Linlin Cheng
- Department of Clinical LaboratoryPeking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical SciencesBeijingChina
| | - Haolong Li
- Department of Clinical LaboratoryPeking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical SciencesBeijingChina
| | - Liubing Li
- Department of Clinical LaboratoryPeking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical SciencesBeijingChina
| | - Chenxi Liu
- Department of Clinical LaboratoryPeking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical SciencesBeijingChina
| | - Songxin Yan
- Department of Clinical LaboratoryPeking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical SciencesBeijingChina
| | - Haizhen Chen
- Department of Clinical LaboratoryPeking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical SciencesBeijingChina
- Department of Clinical LaboratoryThe First Hospital of Jilin UniversityJilinChina
| | - Yongzhe Li
- Department of Clinical LaboratoryPeking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical SciencesBeijingChina
| |
Collapse
|