1
|
Sangokoya C. The FIRE biosensor illuminates iron regulatory protein activity and cellular iron homeostasis. CELL REPORTS METHODS 2025; 5:100960. [PMID: 39824193 DOI: 10.1016/j.crmeth.2024.100960] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/21/2024] [Revised: 11/18/2024] [Accepted: 12/20/2024] [Indexed: 01/20/2025]
Abstract
On Earth, iron is abundant, bioavailable, and crucial for initiating the first catalytic reactions of life from prokaryotes to plants to mammals. Iron-complexed proteins are critical to biological pathways and essential cellular functions. While it is well known that the regulation of iron is necessary for mammalian development, little is known about the timeline of how specific transcripts network and interact in response to cellular iron regulation to shape cell fate, function, and plasticity in the developing embryo and beyond. Here, we present a ratiometric genetically encoded dual biosensor called FIRE (Fe-IRE [iron-responsive element]) to evaluate iron regulatory protein (IRP)-binding activity and cellular iron status in live cells, allowing for the study and dissection of dynamic changes in cellular iron and IRP activity over developmental time. FIRE reveals a previously unrecognized foundational timeline of IRP activity and cellular iron homeostasis during stem cell pluripotency transition and early differentiation.
Collapse
Affiliation(s)
- Carolyn Sangokoya
- Department of Pathology, University of California, San Francisco, San Francisco, CA, USA; The Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, CA, USA.
| |
Collapse
|
2
|
Dev S, Asthana S, Singh P, Seth P, Banerjee C, Mukhopadhyay CK. Dopamine degrades ferritin by chaperone-mediated autophagy to elevate mitochondrial iron level in astroglial cells. Free Radic Biol Med 2025; 229:39-57. [PMID: 39818240 DOI: 10.1016/j.freeradbiomed.2025.01.021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/25/2024] [Revised: 01/05/2025] [Accepted: 01/13/2025] [Indexed: 01/18/2025]
Abstract
Iron accumulation and mitochondrial dysfunction in astroglia are reported in Parkinson's disease (PD). Astroglia control iron availability in neurons in which dopamine (DA) synthesis is affected in PD. Despite their intimate relationship the role of DA in astroglial iron homeostasis is limited. Here we show that DA degrades iron storage protein ferritin in astroglial cells involving lysosomal proteolysis. Lysosomal ferritinophagy is mainly associated with macroautophagy; however, we revealed the involvement of chaperone-mediated autophagy (CMA) in DA-induced ferritin degradation. In CMA, cytosolic proteins containing a specific pentapeptide motif bind with HSC70 to be transported to lysosome mediated by LAMP2A. We identified the conserved pentapeptide motif in ferritin-H (Ft-H), mutations of which resulted loss of its interaction with HSC70. Pharmacological inhibitors of HSC70 or LAMP2/2A knockdown blocks DA-induced Ft-H degradation. DA also induces cytosolic cargo NCOA4 for ferritinophagy. We further reveal that DA promotes cathepsin B to lysis ferritin within the lysosome. Inhibitor of cathepsin B, knocking down of LAMP2, or HSC70 inhibitor attenuate DA-induced elevated mitochondrial iron level. Our results establish a direct role of DA on astroglial iron homeostasis and novel involvement of CMA in ferritin degradation in response to a biological stimulus. These results also may help in better understanding iron dyshomeostasis and mitochondrial dysfunction reported in PD.
Collapse
Affiliation(s)
- Som Dev
- Special Centre for Molecular Medicine, Jawaharlal Nehru University, New Delhi, 110067, India; Department of Biochemistry, All India Institute of Medical Sciences, Kalyani, West Bengal, India, 741245
| | - Somya Asthana
- Special Centre for Molecular Medicine, Jawaharlal Nehru University, New Delhi, 110067, India
| | - Pratibha Singh
- Special Centre for Molecular Medicine, Jawaharlal Nehru University, New Delhi, 110067, India
| | - Pankaj Seth
- National Brain Research Centre, Manesar, Haryana, 122052, India
| | - Chayanika Banerjee
- Special Centre for Molecular Medicine, Jawaharlal Nehru University, New Delhi, 110067, India
| | - Chinmay K Mukhopadhyay
- Special Centre for Molecular Medicine, Jawaharlal Nehru University, New Delhi, 110067, India.
| |
Collapse
|
3
|
Isasi E, Olivera-Bravo S. Neurovascular unit impairment in iron deficiency anemia. Neuroscience 2024; 567:56-66. [PMID: 39733822 DOI: 10.1016/j.neuroscience.2024.12.050] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2024] [Revised: 12/16/2024] [Accepted: 12/26/2024] [Indexed: 12/31/2024]
Abstract
Iron is one of the crucial elements for CNS development and function and its deficiency (ID) is the most common worldwide nutrient deficit in the world. Iron deficiency anemia (IDA) in pregnant women and infants is a worldwide health problem due to its high prevalence and its irreversible long-lasting effects on brain development. Even with iron supplementation, IDA during pregnancy and/or breastfeeding can result in irreversible cognitive, motor, and behavioral impairments. The neurovascular unit (NVU) plays an important role in iron transport within the CNS as well as in the blood brain-barrier (BBB) formation and maturation, vasculogenesis/angiogenesis, neurovascular coupling and metabolic waste clearance. In animal models of IDA, significant changes have been observed at the capillary level, including alterations in iron transport, vasculogenesis, astrocyte endfeet, and pericytes. Despite these findings, the role of the NVU in IDA remains poorly understood. This review summarizes the potential effects of ID/IDA on brain development, myelination and neuronal function and discusses the role of NVU cells in iron metabolism, BBB, vasculogenesis/angiogenesis, neurovascular coupling and metabolic waste clearance. Furthermore, it emphasizes the need to view the NVU as a whole and as a potential target for ID/IDA. However, it remains unclear to what extent NVU alterations contribute to neuronal dysfunction, myelination abnormalities, and synaptic disturbances described in IDA.
Collapse
Affiliation(s)
- Eugenia Isasi
- Unidad Académica de Histología y Embriología, Facultad de Medicina, UdelaR, Montevideo, Uruguay; Departamento de Neurobiología y Neuropatología, IIBCE, MEC, Montevideo, Uruguay
| | - Silvia Olivera-Bravo
- Departamento de Neurobiología y Neuropatología, IIBCE, MEC, Montevideo, Uruguay.
| |
Collapse
|
4
|
Zhang T, Liu Q, Chen Q, Wu H. Iron regulatory protein two facilitates ferritinophagy and DNA damage/repair through guiding ATG9A trafficking. J Biol Chem 2024; 300:107767. [PMID: 39276939 PMCID: PMC11490887 DOI: 10.1016/j.jbc.2024.107767] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2024] [Revised: 07/17/2024] [Accepted: 08/25/2024] [Indexed: 09/17/2024] Open
Abstract
Trace elemental iron is an essential nutrient that participates in diverse metabolic processes. Dysregulation of cellular iron homeostasis, both iron deficiency and iron overload, is detrimental and tightly associated with disease pathogenesis. IRPs-IREs system is located at the center for iron homeostasis regulation. Additionally, ferritinophagy, the autophagy-dependent ferritin catabolism for iron recycling, is emerging as a novel mechanism for iron homeostasis regulation. It is still unclear whether IRPs-IREs system and ferritinophagy are synergistic or redundant in determining iron homeostasis. Here we report that IRP2, but not IRP1, is indispensable for ferritinophagy in response to iron depletion. Mechanistically, IRP2 ablation results in compromised AMPK activation and defective ATG9A endosomal trafficking, leading to the decreased engulfment of NCOA4-ferritin complex by endosomes and the subsequent dysregulated endosomal microferritinophagy. Moreover, this defective endosomal microferritinophagy exacerbates DNA damage and reduces colony formation in IRP2-depleted cells. Collectively, this study expands the physiological function of IRP2 in endosomal microferritinophagy and highlights potential crosstalk between IRPs-IREs and ferritinophagy in manipulating iron homeostasis.
Collapse
Affiliation(s)
- Ting Zhang
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China; Hubei Hongshan Laboratory, Wuhan, Hubei, China
| | - Qian Liu
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China; Hubei Hongshan Laboratory, Wuhan, Hubei, China
| | - Quan Chen
- State Key Laboratory of Medicinal Chemical Biology, College of Life Sciences, Nankai University, Tianjin, China
| | - Hao Wu
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China; Hubei Hongshan Laboratory, Wuhan, Hubei, China.
| |
Collapse
|
5
|
Bitar S, Baumann T, Weber C, Abusaada M, Rojas-Charry L, Ziegler P, Schettgen T, Randerath IE, Venkataramani V, Michalke B, Hanschmann EM, Arena G, Krueger R, Zhang L, Methner A. Iron-sulfur cluster loss in mitochondrial CISD1 mediates PINK1 loss-of-function phenotypes. eLife 2024; 13:e97027. [PMID: 39159312 PMCID: PMC11383524 DOI: 10.7554/elife.97027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2024] [Accepted: 07/10/2024] [Indexed: 08/21/2024] Open
Abstract
Parkinson's disease (PD) is characterized by the progressive loss of dopaminergic neurons in the substantia nigra of the midbrain. Familial cases of PD are often caused by mutations of PTEN-induced kinase 1 (PINK1) and the ubiquitin ligase Parkin, both pivotal in maintaining mitochondrial quality control. CISD1, a homodimeric mitochondrial iron-sulfur-binding protein, is a major target of Parkin-mediated ubiquitination. We here discovered a heightened propensity of CISD1 to form dimers in Pink1 mutant flies and in dopaminergic neurons from PINK1 mutation patients. The dimer consists of two monomers that are covalently linked by a disulfide bridge. In this conformation CISD1 cannot coordinate the iron-sulfur cofactor. Overexpressing Cisd, the Drosophila ortholog of CISD1, and a mutant Cisd incapable of binding the iron-sulfur cluster in Drosophila reduced climbing ability and lifespan. This was more pronounced with mutant Cisd and aggravated in Pink1 mutant flies. Complete loss of Cisd, in contrast, rescued all detrimental effects of Pink1 mutation on climbing ability, wing posture, dopamine levels, lifespan, and mitochondrial ultrastructure. Our results suggest that Cisd, probably iron-depleted Cisd, operates downstream of Pink1 shedding light on PD pathophysiology and implicating CISD1 as a potential therapeutic target.
Collapse
Affiliation(s)
- Sara Bitar
- University Medical Center of the Johannes Gutenberg-University Mainz, Institute for Molecular Medicine, Mainz, Germany
| | - Timo Baumann
- University Medical Center of the Johannes Gutenberg-University Mainz, Institute for Molecular Medicine, Mainz, Germany
| | - Christopher Weber
- University Medical Center of the Johannes Gutenberg-University Mainz, Institute for Molecular Medicine, Mainz, Germany
| | - Majd Abusaada
- University Medical Center of the Johannes Gutenberg-University Mainz, Institute for Molecular Medicine, Mainz, Germany
| | - Liliana Rojas-Charry
- University Medical Center of the Johannes Gutenberg-University Mainz, Institute for Molecular Medicine, Mainz, Germany
| | - Patrick Ziegler
- Institute for Occupational, Social and Environmental Medicine, RWTH Aachen University, Aachen, Germany
| | - Thomas Schettgen
- Institute for Occupational, Social and Environmental Medicine, RWTH Aachen University, Aachen, Germany
| | - Isabella Eva Randerath
- Institute for Occupational, Social and Environmental Medicine, RWTH Aachen University, Aachen, Germany
| | - Vivek Venkataramani
- Comprehensive Cancer Center Mainfranken, University Hospital Würzburg, Würzburg, Germany
| | - Bernhard Michalke
- Research Unit Analytical BioGeoChemistry, Helmholtz Zentrum München-German, Research Center for Environmental Health GmbH, Neuherberg, Germany
| | - Eva-Maria Hanschmann
- Experimental and Translational Research, Department of Otorhinolaryngology, University Hospital Essen, Essen, Germany
| | - Giuseppe Arena
- University of Luxembourg, Luxembourg Centre for Systems Biomedicine, Esch-sur-Alzette, Luxembourg
| | - Rejko Krueger
- University of Luxembourg, Luxembourg Centre for Systems Biomedicine, Esch-sur-Alzette, Luxembourg
- Luxembourg Institute of Health (LIH), Strassen, Luxembourg
- Centre Hospitalier de Luxembourg (CHL), Luxembourg, Luxembourg
| | - Li Zhang
- University Medical Center of the Johannes Gutenberg-University Mainz, Institute for Molecular Medicine, Mainz, Germany
| | - Axel Methner
- University Medical Center of the Johannes Gutenberg-University Mainz, Institute for Molecular Medicine, Mainz, Germany
| |
Collapse
|
6
|
Nadimpalli HP, Katsioudi G, Arpa ES, Chikhaoui L, Arpat AB, Liechti A, Palais G, Tessmer C, Hofmann I, Galy B, Gatfield D. Diurnal control of iron responsive element containing mRNAs through iron regulatory proteins IRP1 and IRP2 is mediated by feeding rhythms. Genome Biol 2024; 25:128. [PMID: 38773499 PMCID: PMC11106963 DOI: 10.1186/s13059-024-03270-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2024] [Accepted: 05/09/2024] [Indexed: 05/23/2024] Open
Abstract
BACKGROUND Cellular iron homeostasis is regulated by iron regulatory proteins (IRP1 and IRP2) that sense iron levels (and other metabolic cues) and modulate mRNA translation or stability via interaction with iron regulatory elements (IREs). IRP2 is viewed as the primary regulator in the liver, yet our previous datasets showing diurnal rhythms for certain IRE-containing mRNAs suggest a nuanced temporal control mechanism. The purpose of this study is to gain insights into the daily regulatory dynamics across IRE-bearing mRNAs, specific IRP involvement, and underlying systemic and cellular rhythmicity cues in mouse liver. RESULTS We uncover high-amplitude diurnal oscillations in the regulation of key IRE-containing transcripts in the liver, compatible with maximal IRP activity at the onset of the dark phase. Although IRP2 protein levels also exhibit some diurnal variations and peak at the light-dark transition, ribosome profiling in IRP2-deficient mice reveals that maximal repression of target mRNAs at this timepoint still occurs. We further find that diurnal regulation of IRE-containing mRNAs can continue in the absence of a functional circadian clock as long as feeding is rhythmic. CONCLUSIONS Our findings suggest temporally controlled redundancy in IRP activities, with IRP2 mediating regulation of IRE-containing transcripts in the light phase and redundancy, conceivably with IRP1, at dark onset. Moreover, we highlight the significance of feeding-associated signals in driving rhythmicity. Our work highlights the dynamic nature and regulatory complexity in a metabolic pathway that had previously been considered well-understood.
Collapse
Affiliation(s)
| | - Georgia Katsioudi
- Center for Integrative Genomics, University of Lausanne, Lausanne, 1015, Switzerland
| | - Enes Salih Arpa
- Center for Integrative Genomics, University of Lausanne, Lausanne, 1015, Switzerland
| | - Lies Chikhaoui
- Center for Integrative Genomics, University of Lausanne, Lausanne, 1015, Switzerland
| | - Alaaddin Bulak Arpat
- Center for Integrative Genomics, University of Lausanne, Lausanne, 1015, Switzerland
| | - Angelica Liechti
- Center for Integrative Genomics, University of Lausanne, Lausanne, 1015, Switzerland
| | - Gaël Palais
- German Cancer Research Center (DKFZ), Division of Virus-Associated Carcinogenesis, Im Neuenheimer Feld 280, Heidelberg, 69120, Germany
| | - Claudia Tessmer
- German Cancer Research Center (DKFZ), Core Facility Antibodies, Im Neuenheimer Feld 280, Heidelberg, 69120, Germany
| | - Ilse Hofmann
- German Cancer Research Center (DKFZ), Core Facility Antibodies, Im Neuenheimer Feld 280, Heidelberg, 69120, Germany
| | - Bruno Galy
- German Cancer Research Center (DKFZ), Division of Virus-Associated Carcinogenesis, Im Neuenheimer Feld 280, Heidelberg, 69120, Germany
| | - David Gatfield
- Center for Integrative Genomics, University of Lausanne, Lausanne, 1015, Switzerland.
| |
Collapse
|
7
|
Galy B, Conrad M, Muckenthaler M. Mechanisms controlling cellular and systemic iron homeostasis. Nat Rev Mol Cell Biol 2024; 25:133-155. [PMID: 37783783 DOI: 10.1038/s41580-023-00648-1] [Citation(s) in RCA: 165] [Impact Index Per Article: 165.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/31/2023] [Indexed: 10/04/2023]
Abstract
In mammals, hundreds of proteins use iron in a multitude of cellular functions, including vital processes such as mitochondrial respiration, gene regulation and DNA synthesis or repair. Highly orchestrated regulatory systems control cellular and systemic iron fluxes ensuring sufficient iron delivery to target proteins is maintained, while limiting its potentially deleterious effects in iron-mediated oxidative cell damage and ferroptosis. In this Review, we discuss how cells acquire, traffick and export iron and how stored iron is mobilized for iron-sulfur cluster and haem biogenesis. Furthermore, we describe how these cellular processes are fine-tuned by the combination of various sensory and regulatory systems, such as the iron-regulatory protein (IRP)-iron-responsive element (IRE) network, the nuclear receptor co-activator 4 (NCOA4)-mediated ferritinophagy pathway, the prolyl hydroxylase domain (PHD)-hypoxia-inducible factor (HIF) axis or the nuclear factor erythroid 2-related factor 2 (NRF2) regulatory hub. We further describe how these pathways interact with systemic iron homeostasis control through the hepcidin-ferroportin axis to ensure appropriate iron fluxes. This knowledge is key for the identification of novel therapeutic opportunities to prevent diseases of cellular and/or systemic iron mismanagement.
Collapse
Affiliation(s)
- Bruno Galy
- German Cancer Research Center (DKFZ), Division of Virus-associated Carcinogenesis (F170), Heidelberg, Germany
| | - Marcus Conrad
- Helmholtz Zentrum München, Institute of Metabolism and Cell Death, Neuherberg, Germany
| | - Martina Muckenthaler
- Department of Paediatric Hematology, Oncology and Immunology, University of Heidelberg, Heidelberg, Germany.
- Molecular Medicine Partnership Unit, University of Heidelberg, Heidelberg, Germany.
- German Centre for Cardiovascular Research (DZHK), Partner site Heidelberg/Mannheim, Heidelberg, Germany.
- Translational Lung Research Center Heidelberg (TLRC), German Center for Lung Research (DZL), University of Heidelberg, Heidelberg, Germany.
| |
Collapse
|
8
|
Chang S, Wang P, Han Y, Ma Q, Liu Z, Zhong S, Lu Y, Chen R, Sun L, Wu Q, Gao G, Wang X, Chang YZ. Ferrodifferentiation regulates neurodevelopment via ROS generation. SCIENCE CHINA. LIFE SCIENCES 2023; 66:1841-1857. [PMID: 36929272 DOI: 10.1007/s11427-022-2297-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Accepted: 02/16/2023] [Indexed: 03/18/2023]
Abstract
Iron is important for life, and iron deficiency impairs development, but whether the iron level regulates neural differentiation remains elusive. In this study, with iron-regulatory proteins (IRPs) knockout embryonic stem cells (ESCs) that showed severe iron deficiency, we found that the Pax6- and Sox2-positive neuronal precursor cells and Tuj1 fibers in IRP1-/-IRP2-/- ESCs were significantly decreased after inducing neural differentiation. Consistently, in vivo study showed that the knockdown of IRP1 in IRP2-/- fetal mice remarkably affected the differentiation of neuronal precursors and the migration of neurons. These findings suggest that low intracellular iron status significantly inhibits neurodifferentiation. When supplementing IRP1-/-IRP2-/- ESCs with iron, these ESCs could differentiate normally. Further investigations revealed that the underlying mechanism was associated with an increase in reactive oxygen species (ROS) production caused by the substantially low level of iron and the down-regulation of iron-sulfur cluster protein ISCU, which, in turn, affected the proliferation and differentiation of stem cells. Thus, the appropriate amount of iron is crucial for maintaining normal neural differentiation that is termed ferrodifferentiation.
Collapse
Affiliation(s)
- Shiyang Chang
- Laboratory of Molecular Iron Metabolism, Ministry of Education Key Laboratory of Molecular and Cellular Biology, Hebei Key Laboratory of Animal Physiology, Biochemistry, and Molecular Biology, College of Life Sciences, Hebei Normal University, Shijiazhuang, 050024, China
- College of Basic Medicine, Hebei Medical University, Shijiazhuang, 050017, China
- State Key Laboratory of Brain and Cognitive Science, CAS Center for Excellence in Brain Science and Intelligence Technology (Shanghai), Institute of Biophysics, Chinese Academy of Sciences (CAS), BNU IDG/McGovern Institute for Brain Research, Beijing, 100101, China
| | - Peina Wang
- Laboratory of Molecular Iron Metabolism, Ministry of Education Key Laboratory of Molecular and Cellular Biology, Hebei Key Laboratory of Animal Physiology, Biochemistry, and Molecular Biology, College of Life Sciences, Hebei Normal University, Shijiazhuang, 050024, China
- College of Basic Medicine, Hebei Medical University, Shijiazhuang, 050017, China
| | - Yingying Han
- Laboratory of Molecular Iron Metabolism, Ministry of Education Key Laboratory of Molecular and Cellular Biology, Hebei Key Laboratory of Animal Physiology, Biochemistry, and Molecular Biology, College of Life Sciences, Hebei Normal University, Shijiazhuang, 050024, China
| | - Qiang Ma
- State Key Laboratory of Brain and Cognitive Science, CAS Center for Excellence in Brain Science and Intelligence Technology (Shanghai), Institute of Biophysics, Chinese Academy of Sciences (CAS), BNU IDG/McGovern Institute for Brain Research, Beijing, 100101, China
| | - Zeyuan Liu
- State Key Laboratory of Brain and Cognitive Science, CAS Center for Excellence in Brain Science and Intelligence Technology (Shanghai), Institute of Biophysics, Chinese Academy of Sciences (CAS), BNU IDG/McGovern Institute for Brain Research, Beijing, 100101, China
| | - Suijuan Zhong
- State Key Laboratory of Cognitive Neuroscience and Learning, Beijing Normal University, Beijing, 100875, China
| | - Yufeng Lu
- State Key Laboratory of Brain and Cognitive Science, CAS Center for Excellence in Brain Science and Intelligence Technology (Shanghai), Institute of Biophysics, Chinese Academy of Sciences (CAS), BNU IDG/McGovern Institute for Brain Research, Beijing, 100101, China
| | - Ruiguo Chen
- State Key Laboratory of Brain and Cognitive Science, CAS Center for Excellence in Brain Science and Intelligence Technology (Shanghai), Institute of Biophysics, Chinese Academy of Sciences (CAS), BNU IDG/McGovern Institute for Brain Research, Beijing, 100101, China
| | - Le Sun
- Beijing Institute of Brain Disorders, Laboratory of Brain Disorders, Ministry of Science and Technology, Collaborative Innovation Center for Brain Disorders, Capital Medical University, Beijing, 100069, China
| | - Qian Wu
- State Key Laboratory of Cognitive Neuroscience and Learning, Beijing Normal University, Beijing, 100875, China
| | - Guofen Gao
- Laboratory of Molecular Iron Metabolism, Ministry of Education Key Laboratory of Molecular and Cellular Biology, Hebei Key Laboratory of Animal Physiology, Biochemistry, and Molecular Biology, College of Life Sciences, Hebei Normal University, Shijiazhuang, 050024, China.
| | - Xiaoqun Wang
- State Key Laboratory of Brain and Cognitive Science, CAS Center for Excellence in Brain Science and Intelligence Technology (Shanghai), Institute of Biophysics, Chinese Academy of Sciences (CAS), BNU IDG/McGovern Institute for Brain Research, Beijing, 100101, China.
| | - Yan-Zhong Chang
- Laboratory of Molecular Iron Metabolism, Ministry of Education Key Laboratory of Molecular and Cellular Biology, Hebei Key Laboratory of Animal Physiology, Biochemistry, and Molecular Biology, College of Life Sciences, Hebei Normal University, Shijiazhuang, 050024, China.
| |
Collapse
|
9
|
Beavers CJ, Ambrosy AP, Butler J, Davidson BT, Gale SE, Piña IL, Mastoris I, Reza N, Mentz RJ, Lewis GD. Iron Deficiency in Heart Failure: A Scientific Statement from the Heart Failure Society of America. J Card Fail 2023; 29:1059-1077. [PMID: 37137386 DOI: 10.1016/j.cardfail.2023.03.025] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Revised: 03/10/2023] [Accepted: 03/23/2023] [Indexed: 05/05/2023]
Abstract
Iron deficiency is present in approximately 50% of patients with symptomatic heart failure and is independently associated with worse functional capacity, lower quality of, life and increased mortality. The purpose of this document is to summarize current knowledge of how iron deficiency is defined in heart failure and its epidemiology and pathophysiology, as well as pharmacological considerations for repletion strategies. This document also summarizes the rapidly expanding array of clinical trial evidence informing when, how, and in whom to consider iron repletion.
Collapse
Affiliation(s)
- Craig J Beavers
- University of Kentucky College of Pharmacy, Lexington, Kentucky.
| | - Andrew P Ambrosy
- Kaiser Permanente Northern California - Division of Research (DOR), Oakland, CA
| | - Javed Butler
- Baylor Scott and White Research Institute, Dallas, Texas; University of Mississippi, Jackson, Mississippi
| | - Beth T Davidson
- Centennial Heart Cardiovascular Consultants, Nashville, Tennessee
| | - Stormi E Gale
- Novant Health Matthews Medical Center, Matthews, North Carolina
| | - Ileana L Piña
- Thomas Jefferson University, Philadelphia, Pennsylvania
| | | | - Nosheen Reza
- Division of Cardiovascular Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania
| | - Robert J Mentz
- Duke University School of Medicine, Durham, North Carolina
| | | |
Collapse
|
10
|
Duan G, Li J, Duan Y, Zheng C, Guo Q, Li F, Zheng J, Yu J, Zhang P, Wan M, Long C. Mitochondrial Iron Metabolism: The Crucial Actors in Diseases. Molecules 2022; 28:29. [PMID: 36615225 PMCID: PMC9822237 DOI: 10.3390/molecules28010029] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Revised: 12/12/2022] [Accepted: 12/13/2022] [Indexed: 12/24/2022] Open
Abstract
Iron is a trace element necessary for cell growth, development, and cellular homeostasis, but insufficient or excessive level of iron is toxic. Intracellularly, sufficient amounts of iron are required for mitochondria (the center of iron utilization) to maintain their normal physiologic function. Iron deficiency impairs mitochondrial metabolism and respiratory activity, while mitochondrial iron overload promotes ROS production during mitochondrial electron transport, thus promoting potential disease development. This review provides an overview of iron homeostasis, mitochondrial iron metabolism, and how mitochondrial iron imbalances-induced mitochondrial dysfunction contribute to diseases.
Collapse
Affiliation(s)
- Geyan Duan
- CAS Key Laboratory of Agro-Ecological Processes in Subtropical Region, Hunan Provincial Key Laboratory of Animal Nutritional Physiology and Metabolic Process, National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha 410125, China
- College of Advanced Agricultural Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Jianjun Li
- CAS Key Laboratory of Agro-Ecological Processes in Subtropical Region, Hunan Provincial Key Laboratory of Animal Nutritional Physiology and Metabolic Process, National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha 410125, China
- College of Advanced Agricultural Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yehui Duan
- CAS Key Laboratory of Agro-Ecological Processes in Subtropical Region, Hunan Provincial Key Laboratory of Animal Nutritional Physiology and Metabolic Process, National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha 410125, China
- College of Advanced Agricultural Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Changbing Zheng
- College of Animal Science and Technology, Hunan Agricultural University, Changsha 410128, China
| | - Qiuping Guo
- CAS Key Laboratory of Agro-Ecological Processes in Subtropical Region, Hunan Provincial Key Laboratory of Animal Nutritional Physiology and Metabolic Process, National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha 410125, China
- College of Advanced Agricultural Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Fengna Li
- CAS Key Laboratory of Agro-Ecological Processes in Subtropical Region, Hunan Provincial Key Laboratory of Animal Nutritional Physiology and Metabolic Process, National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha 410125, China
- College of Advanced Agricultural Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Jie Zheng
- CAS Key Laboratory of Agro-Ecological Processes in Subtropical Region, Hunan Provincial Key Laboratory of Animal Nutritional Physiology and Metabolic Process, National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha 410125, China
- College of Advanced Agricultural Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Jiayi Yu
- CAS Key Laboratory of Agro-Ecological Processes in Subtropical Region, Hunan Provincial Key Laboratory of Animal Nutritional Physiology and Metabolic Process, National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha 410125, China
- College of Advanced Agricultural Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Peiwen Zhang
- College of Animal Science and Technology, Hunan Agricultural University, Changsha 410128, China
| | - Mengliao Wan
- College of Animal Science and Technology, Hunan Agricultural University, Changsha 410128, China
| | - Cimin Long
- CAS Key Laboratory of Agro-Ecological Processes in Subtropical Region, Hunan Provincial Key Laboratory of Animal Nutritional Physiology and Metabolic Process, National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha 410125, China
- College of Advanced Agricultural Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| |
Collapse
|
11
|
Tkaczyszyn M, Górniak KM, Lis WH, Ponikowski P, Jankowska EA. Iron Deficiency and Deranged Myocardial Energetics in Heart Failure. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2022; 19:17000. [PMID: 36554881 PMCID: PMC9778731 DOI: 10.3390/ijerph192417000] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/05/2022] [Revised: 12/10/2022] [Accepted: 12/12/2022] [Indexed: 06/17/2023]
Abstract
Among different pathomechanisms involved in the development of heart failure, adverse metabolic myocardial remodeling closely related to ineffective energy production, constitutes the fundamental feature of the disease and translates into further progression of both cardiac dysfunction and maladaptations occurring within other organs. Being the component of key enzymatic machineries, iron plays a vital role in energy generation and utilization, hence the interest in whether, by correcting systemic and/or cellular deficiency of this micronutrient, we can influence the energetic efficiency of tissues, including the heart. In this review we summarize current knowledge on disturbed energy metabolism in failing hearts as well as we analyze experimental evidence linking iron deficiency with deranged myocardial energetics.
Collapse
Affiliation(s)
- Michał Tkaczyszyn
- Institute of Heart Diseases, Wroclaw Medical University, 50-556 Wroclaw, Poland
- Institute of Heart Diseases, University Hospital, 50-566 Wroclaw, Poland
| | | | - Weronika Hanna Lis
- Institute of Heart Diseases, University Hospital, 50-566 Wroclaw, Poland
| | - Piotr Ponikowski
- Institute of Heart Diseases, Wroclaw Medical University, 50-556 Wroclaw, Poland
- Institute of Heart Diseases, University Hospital, 50-566 Wroclaw, Poland
| | - Ewa Anita Jankowska
- Institute of Heart Diseases, Wroclaw Medical University, 50-556 Wroclaw, Poland
- Institute of Heart Diseases, University Hospital, 50-566 Wroclaw, Poland
| |
Collapse
|
12
|
Liu Z, Xu S, Zhang Z, Wang H, Jing Q, Zhang S, Liu M, Han J, Kou Y, Wei Y, Wang L, Wang Y. FAM96A is essential for maintaining organismal energy balance and adipose tissue homeostasis in mice. Free Radic Biol Med 2022; 192:115-129. [PMID: 36150559 DOI: 10.1016/j.freeradbiomed.2022.09.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/07/2022] [Revised: 09/13/2022] [Accepted: 09/14/2022] [Indexed: 10/31/2022]
Abstract
The iron (Fe) metabolism plays important role in regulating systemic metabolism and obesity development. The Fe inside cells can form iron-sulfur (Fe-S) clusters, which are usually assembled into target proteins with the help of a conserved cluster assembly machinery. Family with sequence similarity 96A (FAM96A; also designated CIAO2A) is a cytosolic Fe-S assembly protein involved in the regulation of cellular Fe homeostasis. However, the biological function of FAM96A in vivo is still incompletely defined. Here, we tested the role of FAM96A in regulating organismal Fe metabolism, which is relevant to obesity and adipose tissue homeostasis. We found that in mice genetically lacking FAM96A globally, intracellular Fe homeostasis was interrupted in both white and brown adipocytes, but the systemic Fe level was normal. FAM96A deficiency led to adipocyte hypertrophy and organismal energy expenditure reduction even under nonobesogenic normal chow diet-fed conditions. Mechanistically, FAM96A deficiency promoted mechanistic target of rapamycin (mTOR) signaling in adipocytes, leading to an elevation of de novo lipogenesis and, therefore, fat mass accumulation. Furthermore, it also caused mitochondrial defects, including defects in mitochondrial number, ultrastructure, redox activity, and metabolic function in brown adipocytes, which are known to be critical for the control of energy balance. Moreover, adipocyte-selective FAM96A knockout partially phenocopied global FAM96A deficiency with adipocyte hypertrophy and organismal energy expenditure defects but the mice were resistant to high-fat diet-induced weight gain. Thus, FAM96A in adipocytes may autonomously act as a critical gatekeeper of organismal energy balance by coupling Fe metabolism to adipose tissue homeostasis.
Collapse
Affiliation(s)
- Zhuanzhuan Liu
- Department of Pathogen Biology and Immunology, Jiangsu Key Laboratory of Immunity and Metabolism, Xuzhou Medical University, Xuzhou, Jiangsu Province, China.
| | - Shihong Xu
- Department of Pathogen Biology and Immunology, Jiangsu Key Laboratory of Immunity and Metabolism, Xuzhou Medical University, Xuzhou, Jiangsu Province, China.
| | - Zhiwei Zhang
- Department of Pathogen Biology and Immunology, Jiangsu Key Laboratory of Immunity and Metabolism, Xuzhou Medical University, Xuzhou, Jiangsu Province, China.
| | - Hanying Wang
- Department of Pathogen Biology and Immunology, Jiangsu Key Laboratory of Immunity and Metabolism, Xuzhou Medical University, Xuzhou, Jiangsu Province, China.
| | - Qiyue Jing
- Department of Pathogen Biology and Immunology, Jiangsu Key Laboratory of Immunity and Metabolism, Xuzhou Medical University, Xuzhou, Jiangsu Province, China.
| | - Shenghan Zhang
- Department of Pathogen Biology and Immunology, Jiangsu Key Laboratory of Immunity and Metabolism, Xuzhou Medical University, Xuzhou, Jiangsu Province, China.
| | - Mengnan Liu
- Department of Pathogen Biology and Immunology, Jiangsu Key Laboratory of Immunity and Metabolism, Xuzhou Medical University, Xuzhou, Jiangsu Province, China.
| | - Jinzhi Han
- Department of Pathogen Biology and Immunology, Jiangsu Key Laboratory of Immunity and Metabolism, Xuzhou Medical University, Xuzhou, Jiangsu Province, China.
| | - Yanbo Kou
- Department of Pathogen Biology and Immunology, Jiangsu Key Laboratory of Immunity and Metabolism, Xuzhou Medical University, Xuzhou, Jiangsu Province, China.
| | - Yanxia Wei
- Department of Pathogen Biology and Immunology, Jiangsu Key Laboratory of Immunity and Metabolism, Xuzhou Medical University, Xuzhou, Jiangsu Province, China.
| | - Lu Wang
- Peking University Center for Human Disease Genomics, Beijing, China; Department of Immunology, School of Basic Medical Sciences, Peking University, Beijing, China; NHC Key Laboratory of Medical Immunology, Peking University, Beijing, China.
| | - Yugang Wang
- Department of Pathogen Biology and Immunology, Jiangsu Key Laboratory of Immunity and Metabolism, Xuzhou Medical University, Xuzhou, Jiangsu Province, China.
| |
Collapse
|
13
|
Bi YH, Wang J, Guo ZJ, Jia KN. Characterization of Ferroptosis-Related Molecular Subtypes with Immune Infiltrations in Neuropathic Pain. J Pain Res 2022; 15:3327-3348. [PMID: 36311291 PMCID: PMC9601606 DOI: 10.2147/jpr.s385228] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Accepted: 10/18/2022] [Indexed: 11/23/2022] Open
Abstract
Background Neuropathic pain (NP) caused by a lesion or disease of the somatosensory nervous system is a common chronic pain condition that has a major impact on quality of life. However, NP pathogenesis remains unclear. The purpose of this study was to identify differentially expressed genes (DEGs) and specific and meaningful gene targets for the diagnosis and treatment of NP. Methods Data from rat spinal nerve ligations and the sham group were downloaded from the Gene Expression Omnibus (GEO) database. Based on the single-sample gene set enrichment analysis (ssGSEA) method, 29 immune gene sets were identified in each sample, and these samples were correlated with the immune infiltration phenotype. LASSO regression modeling was used to screen key genes to identify diagnostic gene markers. According to GSEA and GSVA, NP is concentrated in a large number of immune-related pathways and genes. Additionally, we used the DGIdb database and correlation test to construct gene-drug and transcription factor interaction networks for differentially expressed genes relevant to NP-related ferroptosis. We used WGCNA to identify gene co-expression modules of NP, and explored the relationship between gene networks and phenotypes. Finally, we crossed core genes with diagnostic markers and analyzed gene correlation with molecular subtypes and immune cells. Results We identified 224 DEGs, including 191 upregulated genes and 33 downregulated genes. APC co-stimulation, CCR, cytolytic activity, humid-promoting, neutrophils, NK cells, and RGS4, CXCL2, DRD4 and other 7 genes related to ferroptosis were involved in NP development. Key genes of RGS4 and HIF-1 signaling pathway were screened. Conclusion This study contributes to our understanding of the neuroimmune mechanism of neuropathic pain, provides a reference for NP biomarkers and drug targets. Ferroptosis may be the next research direction to explore NP mechanism.
Collapse
Affiliation(s)
- Yan-Hua Bi
- Neurosurgery Department, Huabei Petroleum Administration Bureau General Hospital, Renqiu, People’s Republic of China
| | - Jia Wang
- Neurosurgery Department, Huabei Petroleum Administration Bureau General Hospital, Renqiu, People’s Republic of China
| | - Zhi-Jun Guo
- Medical Imaging Department, Huabei Petroleum Administration Bureau General Hospital, Renqiu, People’s Republic of China
| | - Kai-Ning Jia
- Clinical Trials Center, Huabei Petroleum Administration Bureau General Hospital, Renqiu, People’s Republic of China,Correspondence: Kai-Ning Jia, Clinical Trials Center, Huabei Petroleum Administration Bureau General Hospital, Renqiu, 062550, People’s Republic of China, Email
| |
Collapse
|
14
|
Bonadonna M, Altamura S, Tybl E, Palais G, Qatato M, Polycarpou-Schwarz M, Schneider M, Kalk C, Rüdiger W, Ertl A, Anstee N, Bogeska R, Helm D, Milsom MD, Galy B. Iron regulatory protein (IRP)-mediated iron homeostasis is critical for neutrophil development and differentiation in the bone marrow. SCIENCE ADVANCES 2022; 8:eabq4469. [PMID: 36197975 PMCID: PMC9534496 DOI: 10.1126/sciadv.abq4469] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Accepted: 08/18/2022] [Indexed: 06/01/2023]
Abstract
Iron is mostly devoted to the hemoglobinization of erythrocytes for oxygen transport. However, emerging evidence points to a broader role for the metal in hematopoiesis, including the formation of the immune system. Iron availability in mammalian cells is controlled by iron-regulatory protein 1 (IRP1) and IRP2. We report that global disruption of both IRP1 and IRP2 in adult mice impairs neutrophil development and differentiation in the bone marrow, yielding immature neutrophils with abnormally high glycolytic and autophagic activity, resulting in neutropenia. IRPs promote neutrophil differentiation in a cell intrinsic manner by securing cellular iron supply together with transcriptional control of neutropoiesis to facilitate differentiation to fully mature neutrophils. Unlike neutrophils, monocyte count was not affected by IRP and iron deficiency, suggesting a lineage-specific effect of iron on myeloid output. This study unveils the previously unrecognized importance of IRPs and iron metabolism in the formation of a major branch of the innate immune system.
Collapse
Affiliation(s)
- Michael Bonadonna
- German Cancer Research Center, “Division of Virus-Associated Carcinogenesis”, Im Neuenheimer Feld 280, 69120 Heidelberg, Germany
- Biosciences Faculty, University of Heidelberg, 69120 Heidelberg, Germany
| | - Sandro Altamura
- University of Heidelberg, Department of Pediatric Hematology, Oncology and Immunology, Im Neuenheimer Feld 350, 69120 Heidelberg, Germany
| | - Elisabeth Tybl
- German Cancer Research Center, “Division of Virus-Associated Carcinogenesis”, Im Neuenheimer Feld 280, 69120 Heidelberg, Germany
- IB-Cancer Research Foundation, Science Park 2, 66123 Saarbrücken, Germany
| | - Gael Palais
- German Cancer Research Center, “Division of Virus-Associated Carcinogenesis”, Im Neuenheimer Feld 280, 69120 Heidelberg, Germany
| | - Maria Qatato
- German Cancer Research Center, “Division of Virus-Associated Carcinogenesis”, Im Neuenheimer Feld 280, 69120 Heidelberg, Germany
| | - Maria Polycarpou-Schwarz
- German Cancer Research Center, “Division of Virus-Associated Carcinogenesis”, Im Neuenheimer Feld 280, 69120 Heidelberg, Germany
| | - Martin Schneider
- German Cancer Research Center, Mass Spectrometry based Protein Analysis Unit, Im Neuenheimer Feld 280, 69120 Heidelberg, Germany
| | - Christina Kalk
- German Cancer Research Center, “Division of Virus-Associated Carcinogenesis”, Im Neuenheimer Feld 280, 69120 Heidelberg, Germany
| | - Wibke Rüdiger
- German Cancer Research Center, “Division of Virus-Associated Carcinogenesis”, Im Neuenheimer Feld 280, 69120 Heidelberg, Germany
| | - Alina Ertl
- German Cancer Research Center, “Division of Virus-Associated Carcinogenesis”, Im Neuenheimer Feld 280, 69120 Heidelberg, Germany
| | - Natasha Anstee
- Heidelberg Institute for Stem Cell Technology and Experimental Medicine, Im Neuenheimer Feld 280, 69120 Heidelberg, Germany
- German Cancer Research Center, “Division of Experimental Hematology”, Im Neuenheimer Feld 280, 69120 Heidelberg, Germany
| | - Ruzhica Bogeska
- Heidelberg Institute for Stem Cell Technology and Experimental Medicine, Im Neuenheimer Feld 280, 69120 Heidelberg, Germany
- German Cancer Research Center, “Division of Experimental Hematology”, Im Neuenheimer Feld 280, 69120 Heidelberg, Germany
| | - Dominic Helm
- German Cancer Research Center, Mass Spectrometry based Protein Analysis Unit, Im Neuenheimer Feld 280, 69120 Heidelberg, Germany
| | - Michael D. Milsom
- Heidelberg Institute for Stem Cell Technology and Experimental Medicine, Im Neuenheimer Feld 280, 69120 Heidelberg, Germany
- German Cancer Research Center, “Division of Experimental Hematology”, Im Neuenheimer Feld 280, 69120 Heidelberg, Germany
| | - Bruno Galy
- German Cancer Research Center, “Division of Virus-Associated Carcinogenesis”, Im Neuenheimer Feld 280, 69120 Heidelberg, Germany
| |
Collapse
|
15
|
New Players in Neuronal Iron Homeostasis: Insights from CRISPRi Studies. Antioxidants (Basel) 2022; 11:antiox11091807. [PMID: 36139881 PMCID: PMC9495848 DOI: 10.3390/antiox11091807] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Revised: 09/02/2022] [Accepted: 09/07/2022] [Indexed: 11/16/2022] Open
Abstract
Selective regional iron accumulation is a hallmark of several neurodegenerative diseases, including Alzheimer’s disease and Parkinson’s disease. The underlying mechanisms of neuronal iron dyshomeostasis have been studied, mainly in a gene-by-gene approach. However, recent high-content phenotypic screens using CRISPR/Cas9-based gene perturbations allow for the identification of new pathways that contribute to iron accumulation in neuronal cells. Herein, we perform a bioinformatic analysis of a CRISPR-based screening of lysosomal iron accumulation and the functional genomics of human neurons derived from induced pluripotent stem cells (iPSCs). Consistent with previous studies, we identified mitochondrial electron transport chain dysfunction as one of the main mechanisms triggering iron accumulation, although we substantially expanded the gene set causing this phenomenon, encompassing mitochondrial complexes I to IV, several associated assembly factors, and coenzyme Q biosynthetic enzymes. Similarly, the loss of numerous genes participating through the complete macroautophagic process elicit iron accumulation. As a novelty, we found that the impaired synthesis of glycophosphatidylinositol (GPI) and GPI-anchored protein trafficking also trigger iron accumulation in a cell-autonomous manner. Finally, the loss of critical components of the iron transporters trafficking machinery, including MON2 and PD-associated gene VPS35, also contribute to increased neuronal levels. Our analysis suggests that neuronal iron accumulation can arise from the dysfunction of an expanded, previously uncharacterized array of molecular pathways.
Collapse
|
16
|
Greene CJ, Attwood K, Sharma NJ, Balderman B, Deng R, Muhitch JB, Smith GJ, Gross KW, Xu B, Kauffman EC. Iron accumulation typifies renal cell carcinoma tumorigenesis but abates with pathological progression, sarcomatoid dedifferentiation, and metastasis. Front Oncol 2022; 12:923043. [PMID: 35992801 PMCID: PMC9389085 DOI: 10.3389/fonc.2022.923043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2022] [Accepted: 06/29/2022] [Indexed: 11/13/2022] Open
Abstract
Iron is a potent catalyst of oxidative stress and cellular proliferation implicated in renal cell carcinoma (RCC) tumorigenesis, yet it also drives ferroptosis that suppresses cancer progression and represents a novel therapeutic target for advanced RCC. The von Hippel Lindau (VHL)/hypoxia-inducible factor-α (HIF-α) axis is a major regulator of cellular iron, and its inactivation underlying most clear cell (cc) RCC tumors introduces both iron dependency and ferroptosis susceptibility. Despite the central role for iron in VHL/HIF-α signaling and ferroptosis, RCC iron levels and their dynamics during RCC initiation/progression are poorly defined. Here, we conducted a large-scale investigation into the incidence and prognostic significance of total tissue iron in ccRCC and non-ccRCC patient primary tumor cancer cells, tumor microenvironment (TME), metastases and non-neoplastic kidneys. Prussian Blue staining was performed to detect non-heme iron accumulation in over 1600 needle-core sections across multiple tissue microarrays. We found that RCC had significantly higher iron staining scores compared with other solid cancers and, on average, >40 times higher than adjacent renal epithelium. RCC cell iron levels correlated positively with TME iron levels and inversely with RCC levels of the main iron uptake protein, transferrin receptor 1 (TfR1/TFRC/CD71). Intriguingly, RCC iron levels, including in the TME, decreased significantly with pathologic (size/stage/grade) progression, sarcomatoid dedifferentiation, and metastasis, particularly among patients with ccRCC, despite increasing TfR1 levels, consistent with an increasingly iron-deficient tumor state. Opposite to tumor iron changes, adjacent renal epithelial iron increased significantly with RCC/ccRCC progression, sarcomatoid dedifferentiation, and metastasis. Lower tumor iron and higher renal epithelial iron each predicted significantly shorter ccRCC patient metastasis-free survival. In conclusion, iron accumulation typifies RCC tumors but declines toward a relative iron-deficient tumor state during progression to metastasis, despite precisely opposite dynamics in adjacent renal epithelium. These findings raise questions regarding the historically presumed selective advantage for high iron during all phases of cancer evolution, suggesting instead distinct tissue-specific roles during RCC carcinogenesis and early tumorigenesis versus later progression. Future study is warranted to determine how the relative iron deficiency of advanced RCC contributes to ferroptosis resistance and/or introduces a heightened susceptibility to iron deprivation that might be therapeutically exploitable.
Collapse
Affiliation(s)
- Christopher J. Greene
- Department of Urology, Roswell Park Comprehensive Cancer Center, Buffalo, NY, United States
- Department of Biological Sciences, University at Buffalo, Buffalo, NY, United States
| | - Kristopher Attwood
- Department of Biostatistics and Bioinformatics, Roswell Park Comprehensive Cancer Center, Buffalo, NY, United States
| | - Nitika J. Sharma
- Department of Urology, Roswell Park Comprehensive Cancer Center, Buffalo, NY, United States
| | - Benjamin Balderman
- Department of Urology, Roswell Park Comprehensive Cancer Center, Buffalo, NY, United States
| | - Rongia Deng
- Department of Urology, Roswell Park Comprehensive Cancer Center, Buffalo, NY, United States
| | - Jason B. Muhitch
- Department of Immunology, Roswell Park Comprehensive Cancer Center, Buffalo, NY, United States
| | - Gary J. Smith
- Department of Urology, Roswell Park Comprehensive Cancer Center, Buffalo, NY, United States
| | - Kenneth W. Gross
- Department of Molecular and Cellular Biology, Roswell Park Comprehensive Cancer Center, Buffalo, NY, United States
| | - Bo Xu
- Department of Pathology, Roswell Park Comprehensive Cancer Center, Buffalo, NY, United States
| | - Eric C. Kauffman
- Department of Urology, Roswell Park Comprehensive Cancer Center, Buffalo, NY, United States
- Department of Cancer Genetics, Roswell Park Comprehensive Cancer Center, Buffalo, NY, United States
- *Correspondence: Eric C. Kauffman,
| |
Collapse
|
17
|
Influences of Vitamin D and Iron Status on Skeletal Muscle Health: A Narrative Review. Nutrients 2022; 14:nu14132717. [PMID: 35807896 PMCID: PMC9268405 DOI: 10.3390/nu14132717] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Revised: 06/24/2022] [Accepted: 06/27/2022] [Indexed: 12/02/2022] Open
Abstract
There is conflicting evidence of the roles vitamin D and iron have in isolation and combined in relation to muscle health. The purpose of this narrative review was to examine the current literature on the roles that vitamin D and iron have on skeletal muscle mass, strength, and function and how these nutrients are associated with skeletal muscle health in specific populations. Secondary purposes include exploring if low vitamin D and iron status are interrelated with skeletal muscle health and chronic inflammation and reviewing the influence of animal-source foods rich in these nutrients on health and performance. PubMed, Scopus, SPORT Discus, EMBAE, MEDLINE, and Google Scholar databases were searched to determine eligible studies. There was a positive effect of vitamin D on muscle mass, particularly in older adults. There was a positive effect of iron on aerobic and anaerobic performance. Studies reported mixed results for both vitamin D and iron on muscle strength and function. While vitamin D and iron deficiency commonly occur in combination, few studies examined effects on skeletal muscle health and inflammation. Isolated nutrients such as iron and vitamin D may have positive outcomes; however, nutrients within food sources may be most effective in improving skeletal muscle health.
Collapse
|
18
|
Zhang H, Jamieson KL, Grenier J, Nikhanj A, Tang Z, Wang F, Wang S, Seidman JG, Seidman CE, Thompson R, Seubert JM, Oudit GY. Myocardial Iron Deficiency and Mitochondrial Dysfunction in Advanced Heart Failure in Humans. J Am Heart Assoc 2022; 11:e022853. [PMID: 35656974 PMCID: PMC9238720 DOI: 10.1161/jaha.121.022853] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Background Myocardial iron deficiency (MID) in heart failure (HF) remains largely unexplored. We aim to establish defining criterion for MID, evaluate its pathophysiological role, and evaluate the applicability of monitoring it non‐invasively in human explanted hearts. Methods and Results Biventricular tissue iron levels were measured in both failing (n=138) and non‐failing control (NFC, n=46) explanted human hearts. Clinical phenotyping was complemented with comprehensive assessment of myocardial remodeling and mitochondrial functional profiles, including metabolic and oxidative stress. Myocardial iron status was further investigated by cardiac magnetic resonance imaging. Myocardial iron content in the left ventricle was lower in HF versus NFC (121.4 [88.1–150.3] versus 137.4 [109.2–165.9] μg/g dry weight), which was absent in the right ventricle. With a priori cutoff of 86.1 μg/g d.w. in left ventricle, we identified 23% of HF patients with MID (HF‐MID) associated with higher NYHA class and worsened left ventricle function. Respiratory chain and Krebs cycle enzymatic activities were suppressed and strongly correlated with depleted iron stores in HF‐MID hearts. Defenses against oxidative stress were severely impaired in association with worsened adverse remodeling in iron‐deficient hearts. Mechanistically, iron uptake pathways were impeded in HF‐MID including decreased translocation to the sarcolemma, while transmembrane fraction of ferroportin positively correlated with MID. Cardiac magnetic resonance with T2* effectively captured myocardial iron levels in failing hearts. Conclusions MID is highly prevalent in advanced human HF and exacerbates pathological remodeling in HF driven primarily by dysfunctional mitochondria and increased oxidative stress in the left ventricle. Cardiac magnetic resonance demonstrates clinical potential to non‐invasively monitor MID.
Collapse
Affiliation(s)
- Hao Zhang
- Division of Cardiology Department of Medicine Faculty of Medicine and DentistryEdmonton Alberta Canada.,Mazankowski Alberta Heart Institute Edmonton Alberta Canada
| | - K Lockhart Jamieson
- Department of Pharmacology Faculty of Medicine and DentistryEdmonton Alberta Canada
| | - Justin Grenier
- Mazankowski Alberta Heart Institute Edmonton Alberta Canada.,Department of Biomedical Engineering Faculty of Medicine and DentistryEdmonton Alberta Canada
| | - Anish Nikhanj
- Division of Cardiology Department of Medicine Faculty of Medicine and DentistryEdmonton Alberta Canada.,Mazankowski Alberta Heart Institute Edmonton Alberta Canada
| | - Zeyu Tang
- Division of Cardiology Department of Medicine Faculty of Medicine and DentistryEdmonton Alberta Canada.,Mazankowski Alberta Heart Institute Edmonton Alberta Canada
| | - Faqi Wang
- Division of Cardiology Department of Medicine Faculty of Medicine and DentistryEdmonton Alberta Canada.,Mazankowski Alberta Heart Institute Edmonton Alberta Canada
| | - Shaohua Wang
- Mazankowski Alberta Heart Institute Edmonton Alberta Canada.,Division of Cardiac Surgery Department of Surgery Faculty of Medicine and Dentistry University of Alberta Edmonton Alberta Canada
| | | | - Christine E Seidman
- Department of Genetics Harvard Medical School Boston MA.,Cardiovascular Division Brigham and Women's Hospital Boston MA
| | - Richard Thompson
- Mazankowski Alberta Heart Institute Edmonton Alberta Canada.,Department of Biomedical Engineering Faculty of Medicine and DentistryEdmonton Alberta Canada
| | - John M Seubert
- Mazankowski Alberta Heart Institute Edmonton Alberta Canada.,Department of Pharmacology Faculty of Medicine and DentistryEdmonton Alberta Canada
| | - Gavin Y Oudit
- Division of Cardiology Department of Medicine Faculty of Medicine and DentistryEdmonton Alberta Canada.,Mazankowski Alberta Heart Institute Edmonton Alberta Canada
| |
Collapse
|
19
|
Wyart E, Hsu MY, Sartori R, Mina E, Rausch V, Pierobon ES, Mezzanotte M, Pezzini C, Bindels LB, Lauria A, Penna F, Hirsch E, Martini M, Mazzone M, Roetto A, Geninatti Crich S, Prenen H, Sandri M, Menga A, Porporato PE. Iron supplementation is sufficient to rescue skeletal muscle mass and function in cancer cachexia. EMBO Rep 2022; 23:e53746. [PMID: 35199910 PMCID: PMC8982578 DOI: 10.15252/embr.202153746] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Revised: 01/20/2022] [Accepted: 01/25/2022] [Indexed: 12/25/2022] Open
Abstract
Cachexia is a wasting syndrome characterized by devastating skeletal muscle atrophy that dramatically increases mortality in various diseases, most notably in cancer patients with a penetrance of up to 80%. Knowledge regarding the mechanism of cancer-induced cachexia remains very scarce, making cachexia an unmet medical need. In this study, we discovered strong alterations of iron metabolism in the skeletal muscle of both cancer patients and tumor-bearing mice, characterized by decreased iron availability in mitochondria. We found that modulation of iron levels directly influences myotube size in vitro and muscle mass in otherwise healthy mice. Furthermore, iron supplementation was sufficient to preserve both muscle function and mass, prolong survival in tumor-bearing mice, and even rescues strength in human subjects within an unexpectedly short time frame. Importantly, iron supplementation refuels mitochondrial oxidative metabolism and energy production. Overall, our findings provide new mechanistic insights in cancer-induced skeletal muscle wasting, and support targeting iron metabolism as a potential therapeutic option for muscle wasting diseases.
Collapse
Affiliation(s)
- Elisabeth Wyart
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Torino, Turin, Italy
| | - Myriam Y Hsu
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Torino, Turin, Italy
| | - Roberta Sartori
- Department of Biomedical Sciences, University of Padova, Padova, Italy
| | - Erica Mina
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Torino, Turin, Italy
| | - Valentina Rausch
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Torino, Turin, Italy
| | - Elisa S Pierobon
- Department of Surgical, Oncological and Gastroenterological Sciences, Padova University Hospital, Padova, Italy
| | - Mariarosa Mezzanotte
- Department of Clinical and Biological Sciences, University of Torino, Turin, Italy
| | - Camilla Pezzini
- Department of Biomedical Sciences, University of Padova, Padova, Italy
| | - Laure B Bindels
- Metabolism and Nutrition Research Group, Louvain Drug Research Institute, Université catholique de Louvain (UCLouvain), Brussels, Belgium
| | - Andrea Lauria
- Department of Life Sciences and System Biology, University of Torino, Turin, Italy
| | - Fabio Penna
- Department of Clinical and Biological Sciences, University of Torino, Turin, Italy
| | - Emilio Hirsch
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Torino, Turin, Italy
| | - Miriam Martini
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Torino, Turin, Italy
| | - Massimiliano Mazzone
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Torino, Turin, Italy.,Laboratory of Tumor Inflammation and Angiogenesis, Center for Cancer Biology (CCB), Vlaams Instituut voor Biotechnologie (VIB), Leuven, Belgium.,Laboratory of Tumor Inflammation and Angiogenesis, Department of Oncology, Katholieke Universiteit Leuven (KUL), Leuven, Belgium
| | - Antonella Roetto
- Department of Clinical and Biological Sciences, University of Torino, Turin, Italy
| | - Simonetta Geninatti Crich
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Torino, Turin, Italy
| | - Hans Prenen
- Department of Medical Oncology, University Hospital Antwerp, Edegem, Belgium.,Center for Oncological Research (CORE), Integrated Personalized and Precision Oncology Network (IPPON), University of Antwerp, Antwerp, Belgium
| | - Marco Sandri
- Department of Biomedical Sciences, University of Padova, Padova, Italy
| | - Alessio Menga
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Torino, Turin, Italy
| | - Paolo E Porporato
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Torino, Turin, Italy
| |
Collapse
|
20
|
Jing JL, Ning TCY, Natali F, Eisenhaber F, Alfatah M. Iron Supplementation Delays Aging and Extends Cellular Lifespan through Potentiation of Mitochondrial Function. Cells 2022; 11:cells11050862. [PMID: 35269484 PMCID: PMC8909192 DOI: 10.3390/cells11050862] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2022] [Revised: 02/28/2022] [Accepted: 02/28/2022] [Indexed: 02/07/2023] Open
Abstract
Aging is the greatest challenge to humankind worldwide. Aging is associated with a progressive loss of physiological integrity due to a decline in cellular metabolism and functions. Such metabolic changes lead to age-related diseases, thereby compromising human health for the remaining life. Thus, there is an urgent need to identify geroprotectors that regulate metabolic functions to target the aging biological processes. Nutrients are the major regulator of metabolic activities to coordinate cell growth and development. Iron is an important nutrient involved in several biological functions, including metabolism. In this study using yeast as an aging model organism, we show that iron supplementation delays aging and increases the cellular lifespan. To determine how iron supplementation increases lifespan, we performed a gene expression analysis of mitochondria, the main cellular hub of iron utilization. Quantitative analysis of gene expression data reveals that iron supplementation upregulates the expression of the mitochondrial tricarboxylic acid (TCA) cycle and electron transport chain (ETC) genes. Furthermore, in agreement with the expression profiles of mitochondrial genes, ATP level is elevated by iron supplementation, which is required for increasing the cellular lifespan. To confirm, we tested the role of iron supplementation in the AMPK knockout mutant. AMPK is a highly conserved controller of mitochondrial metabolism and energy homeostasis. Remarkably, iron supplementation rescued the short lifespan of the AMPK knockout mutant and confirmed its anti-aging role through the enhancement of mitochondrial functions. Thus, our results suggest a potential therapeutic use of iron supplementation to delay aging and prolong healthspan.
Collapse
Affiliation(s)
- Jovian Lin Jing
- Bioinformatics Institute (BII), A*STAR, Singapore 138671, Singapore; (J.L.J.); (T.C.Y.N.)
| | - Trishia Cheng Yi Ning
- Bioinformatics Institute (BII), A*STAR, Singapore 138671, Singapore; (J.L.J.); (T.C.Y.N.)
| | - Federica Natali
- Singapore Institute of Food and Biotechnology Innovation (SIFBI), A*STAR, Singapore 138669, Singapore;
- School of Biological Sciences (SBS), Nanyang Technological University (NTU), Singapore 637551, Singapore
| | - Frank Eisenhaber
- Bioinformatics Institute (BII), A*STAR, Singapore 138671, Singapore; (J.L.J.); (T.C.Y.N.)
- School of Biological Sciences (SBS), Nanyang Technological University (NTU), Singapore 637551, Singapore
- Genome Institute of Singapore (GIS), A*STAR, Singapore 138672, Singapore
- Correspondence: (F.E.); (M.A.)
| | - Mohammad Alfatah
- Bioinformatics Institute (BII), A*STAR, Singapore 138671, Singapore; (J.L.J.); (T.C.Y.N.)
- Correspondence: (F.E.); (M.A.)
| |
Collapse
|
21
|
ENO1 suppresses cancer cell ferroptosis by degrading the mRNA of iron regulatory protein 1. NATURE CANCER 2022; 3:75-89. [PMID: 35121990 DOI: 10.1038/s43018-021-00299-1] [Citation(s) in RCA: 87] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/17/2020] [Accepted: 11/02/2021] [Indexed: 12/11/2022]
Abstract
α-Enolase 1 (ENO1) is a critical glycolytic enzyme whose aberrant expression drives the pathogenesis of various cancers. ENO1 has been indicated as having additional roles beyond its conventional metabolic activity, but the underlying mechanisms and biological consequences remain elusive. Here, we show that ENO1 suppresses iron regulatory protein 1 (IRP1) expression to regulate iron homeostasis and survival of hepatocellular carcinoma (HCC) cells. Mechanistically, we demonstrate that ENO1, as an RNA-binding protein, recruits CNOT6 to accelerate the messenger RNA decay of IRP1 in cancer cells, leading to inhibition of mitoferrin-1 (Mfrn1) expression and subsequent repression of mitochondrial iron-induced ferroptosis. Moreover, through in vitro and in vivo experiments and clinical sample analysis, we identified IRP1 and Mfrn1 as tumor suppressors by inducing ferroptosis in HCC cells. Taken together, this study establishes an important role for the ENO1-IRP1-Mfrn1 pathway in the pathogenesis of HCC and reveals a previously unknown connection between this pathway and ferroptosis, suggesting a potential innovative cancer therapy.
Collapse
|
22
|
Rethinking IRPs/IRE system in neurodegenerative disorders: Looking beyond iron metabolism. Ageing Res Rev 2022; 73:101511. [PMID: 34767973 DOI: 10.1016/j.arr.2021.101511] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Revised: 10/21/2021] [Accepted: 11/04/2021] [Indexed: 12/11/2022]
Abstract
Iron regulatory proteins (IRPs) and iron regulatory element (IRE) systems are well known in the progression of neurodegenerative disorders by regulating iron related proteins. IRPs are also regulated by iron homeostasis. However, an increasing number of studies have suggested a close relationship between the IRPs/IRE system and non-iron-related neurodegenerative disorders. In this paper, we reviewed that the IRPs/IRE system is not only controlled by iron ions, but also regulated by such factors as post-translational modification, oxygen, nitric oxide (NO), heme, interleukin-1 (IL-1), and metal ions. In addition, by regulating the transcription of non-iron related proteins, the IRPs/IRE system functioned in oxidative metabolism, cell cycle regulation, abnormal proteins aggregation, and neuroinflammation. Finally, by emphasizing the multiple regulations of IRPs/IRE system and its potential relationship with non-iron metabolic neurodegenerative disorders, we provided new strategies for disease treatment targeting IRPs/IRE system.
Collapse
|
23
|
Tirinato L, Marafioti MG, Pagliari F, Jansen J, Aversa I, Hanley R, Nisticò C, Garcia-Calderón D, Genard G, Guerreiro JF, Costanzo FS, Seco J. Lipid droplets and ferritin heavy chain: a devilish liaison in human cancer cell radioresistance. eLife 2021; 10:72943. [PMID: 34499029 PMCID: PMC8497056 DOI: 10.7554/elife.72943] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Accepted: 08/25/2021] [Indexed: 12/12/2022] Open
Abstract
Although much progress has been made in cancer treatment, the molecular mechanisms underlying cancer radioresistance (RR) as well as the biological signatures of radioresistant cancer cells still need to be clarified. In this regard, we discovered that breast, bladder, lung, neuroglioma, and prostate 6 Gy X-ray resistant cancer cells were characterized by an increase of lipid droplet (LD) number and that the cells containing highest LDs showed the highest clonogenic potential after irradiation. Moreover, we observed that LD content was tightly connected with the iron metabolism and in particular with the presence of the ferritin heavy chain (FTH1). In fact, breast and lung cancer cells silenced for the FTH1 gene showed a reduction in the LD numbers and, by consequence, became radiosensitive. FTH1 overexpression as well as iron-chelating treatment by Deferoxamine were able to restore the LD amount and RR. Overall, these results provide evidence of a novel mechanism behind RR in which LDs and FTH1 are tightly connected to each other, a synergistic effect that might be worth deeply investigating in order to make cancer cells more radiosensitive and improve the efficacy of radiation treatments.
Collapse
Affiliation(s)
- Luca Tirinato
- Biomedical Physics in Radiation Oncology, German Cancer Research Center (DKFZ), Im Neuenheimer Feld, Heidelberg, Germany.,Experimental and Clinical Medicine Department, University Magna Graecia of Catanzaro, Catanzaro, Italy
| | - Maria Grazia Marafioti
- Biomedical Physics in Radiation Oncology, German Cancer Research Center (DKFZ), Im Neuenheimer Feld, Heidelberg, Germany.,Experimental and Clinical Medicine Department, University Magna Graecia of Catanzaro, Catanzaro, Italy
| | - Francesca Pagliari
- Biomedical Physics in Radiation Oncology, German Cancer Research Center (DKFZ), Im Neuenheimer Feld, Heidelberg, Germany
| | - Jeannette Jansen
- Biomedical Physics in Radiation Oncology, German Cancer Research Center (DKFZ), Im Neuenheimer Feld, Heidelberg, Germany.,Department of Physics and Astronomy, Heidelberg University, Im Neuenheimer Feld, Heidelberg, Germany
| | - Ilenia Aversa
- Biomedical Physics in Radiation Oncology, German Cancer Research Center (DKFZ), Im Neuenheimer Feld, Heidelberg, Germany.,Experimental and Clinical Medicine Department, University Magna Graecia of Catanzaro, Catanzaro, Italy
| | - Rachel Hanley
- Biomedical Physics in Radiation Oncology, German Cancer Research Center (DKFZ), Im Neuenheimer Feld, Heidelberg, Germany
| | - Clelia Nisticò
- Biomedical Physics in Radiation Oncology, German Cancer Research Center (DKFZ), Im Neuenheimer Feld, Heidelberg, Germany.,Experimental and Clinical Medicine Department, University Magna Graecia of Catanzaro, Catanzaro, Italy
| | - Daniel Garcia-Calderón
- Biomedical Physics in Radiation Oncology, German Cancer Research Center (DKFZ), Im Neuenheimer Feld, Heidelberg, Germany.,Department of Physics and Astronomy, Heidelberg University, Im Neuenheimer Feld, Heidelberg, Germany
| | - Geraldine Genard
- Biomedical Physics in Radiation Oncology, German Cancer Research Center (DKFZ), Im Neuenheimer Feld, Heidelberg, Germany
| | - Joana Filipa Guerreiro
- Centro de Ciências e Tecnologias Nucleares, Instituto Superior Técnico, Universidade de Lisboa, Lisboa, Portugal
| | | | - Joao Seco
- Biomedical Physics in Radiation Oncology, German Cancer Research Center (DKFZ), Im Neuenheimer Feld, Heidelberg, Germany.,Department of Physics and Astronomy, Heidelberg University, Im Neuenheimer Feld, Heidelberg, Germany
| |
Collapse
|
24
|
López-Lluch G. Coenzyme Q homeostasis in aging: Response to non-genetic interventions. Free Radic Biol Med 2021; 164:285-302. [PMID: 33454314 DOI: 10.1016/j.freeradbiomed.2021.01.024] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/03/2020] [Revised: 12/30/2020] [Accepted: 01/11/2021] [Indexed: 12/28/2022]
Abstract
Coenzyme Q (CoQ) is a key component for many essential metabolic and antioxidant activities in cells in mitochondria and cell membranes. Mitochondrial dysfunction is one of the hallmarks of aging and age-related diseases. Deprivation of CoQ during aging can be the cause or the consequence of this mitochondrial dysfunction. In any case, it seems clear that aging-associated CoQ deprivation accelerates mitochondrial dysfunction in these diseases. Non-genetic prolongevity interventions, including CoQ dietary supplementation, can increase CoQ levels in mitochondria and cell membranes improving mitochondrial activity and delaying cell and tissue deterioration by oxidative damage. In this review, we discuss the importance of CoQ deprivation in aging and age-related diseases and the effect of prolongevity interventions on CoQ levels and synthesis and CoQ-dependent antioxidant activities.
Collapse
Affiliation(s)
- Guillermo López-Lluch
- Universidad Pablo de Olavide, Centro Andaluz de Biología Del Desarrollo, CABD-CSIC, CIBERER, Instituto de Salud Carlos III, Carretera de Utrera Km. 1, 41013, Sevilla, Spain.
| |
Collapse
|
25
|
Urrutia PJ, Bórquez DA, Núñez MT. Inflaming the Brain with Iron. Antioxidants (Basel) 2021; 10:antiox10010061. [PMID: 33419006 PMCID: PMC7825317 DOI: 10.3390/antiox10010061] [Citation(s) in RCA: 60] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Revised: 12/31/2020] [Accepted: 12/31/2020] [Indexed: 02/06/2023] Open
Abstract
Iron accumulation and neuroinflammation are pathological conditions found in several neurodegenerative diseases, including Alzheimer's disease (AD) and Parkinson's disease (PD). Iron and inflammation are intertwined in a bidirectional relationship, where iron modifies the inflammatory phenotype of microglia and infiltrating macrophages, and in turn, these cells secrete diffusible mediators that reshape neuronal iron homeostasis and regulate iron entry into the brain. Secreted inflammatory mediators include cytokines and reactive oxygen/nitrogen species (ROS/RNS), notably hepcidin and nitric oxide (·NO). Hepcidin is a small cationic peptide with a central role in regulating systemic iron homeostasis. Also present in the cerebrospinal fluid (CSF), hepcidin can reduce iron export from neurons and decreases iron entry through the blood-brain barrier (BBB) by binding to the iron exporter ferroportin 1 (Fpn1). Likewise, ·NO selectively converts cytosolic aconitase (c-aconitase) into the iron regulatory protein 1 (IRP1), which regulates cellular iron homeostasis through its binding to iron response elements (IRE) located in the mRNAs of iron-related proteins. Nitric oxide-activated IRP1 can impair cellular iron homeostasis during neuroinflammation, triggering iron accumulation, especially in the mitochondria, leading to neuronal death. In this review, we will summarize findings that connect neuroinflammation and iron accumulation, which support their causal association in the neurodegenerative processes observed in AD and PD.
Collapse
Affiliation(s)
- Pamela J. Urrutia
- Department of Biology, Faculty of Sciences, Universidad de Chile, 7800024 Santiago, Chile;
| | - Daniel A. Bórquez
- Center for Biomedical Research, Faculty of Medicine, Universidad Diego Portales, 8370007 Santiago, Chile;
| | - Marco Tulio Núñez
- Department of Biology, Faculty of Sciences, Universidad de Chile, 7800024 Santiago, Chile;
- Correspondence: ; Tel.: +56-2-29787360
| |
Collapse
|
26
|
Garza KR, Clarke SL, Ho YH, Bruss MD, Vasanthakumar A, Anderson SA, Eisenstein RS. Differential translational control of 5' IRE-containing mRNA in response to dietary iron deficiency and acute iron overload. Metallomics 2020; 12:2186-2198. [PMID: 33325950 DOI: 10.1039/d0mt00192a] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Iron regulatory proteins (IRPs) are iron-responsive RNA binding proteins that dictate changes in cellular iron metabolism in animal cells by controlling the fate of mRNAs containing iron responsive elements (IREs). IRPs have broader physiological roles as some targeted mRNAs encode proteins with functions beyond iron metabolism suggesting hierarchical regulation of IRP-targeted mRNAs. We observe that the translational regulation of IRP-targeted mRNAs encoding iron storage (L- and H-ferritins) and export (ferroportin) proteins have different set-points of iron responsiveness compared to that for the TCA cycle enzyme mitochondrial aconitase. The ferritins and ferroportin mRNA were largely translationally repressed in the liver of rats fed a normal diet whereas mitochondrial aconitase mRNA is primarily polysome bound. Consequently, acute iron overload increases polysome association of H- and L-ferritin and ferroportin mRNAs while mitochondrial aconitase mRNA showed little stimulation. Conversely, mitochondrial aconitase mRNA is most responsive in iron deficiency. These differences in regulation were associated with a faster off-rate of IRP1 for the IRE of mitochondrial aconitase in comparison to that of L-ferritin. Thus, hierarchical control of mRNA translation by IRPs involves selective control of cellular functions acting at different states of cellular iron status and that are critical for adaptations to iron deficiency or prevention of iron toxicity.
Collapse
Affiliation(s)
- Kerry R Garza
- University of Wisconsin-Madison, Department of Nutritional Sciences, 1415 Linden Drive, Madison, WI 53706, USA.
| | | | | | | | | | | | | |
Collapse
|
27
|
Schrage B, Rübsamen N, Schulz A, Münzel T, Pfeiffer N, Wild PS, Beutel M, Schmidtmann I, Lott R, Blankenberg S, Zeller T, Lackner KJ, Karakas M. Iron deficiency is a common disorder in general population and independently predicts all-cause mortality: results from the Gutenberg Health Study. Clin Res Cardiol 2020; 109:1352-1357. [PMID: 32215702 DOI: 10.1007/s00392-020-01631-y/tables/3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/24/2019] [Accepted: 03/11/2020] [Indexed: 05/20/2023]
Abstract
BACKGROUND Iron deficiency is now accepted as an independent entity beyond anemia. Recently, a new functional definition of iron deficiency was proposed and proved strong efficacy in randomized cardiovascular clinical trials of intravenous iron supplementation. Here, we characterize the impact of iron deficiency on all-cause mortality in the non-anemic general population based on two distinct definitions. METHODS The Gutenberg Health Study is a population-based, prospective, single-center cohort study. The 5000 individuals between 35 and 74 years underwent baseline and a planned follow-up visit at year 5. Tested definitions of iron deficiency were (1) functional iron deficiency-ferritin levels below 100 µg/l, or ferritin levels between 100 and 299 µg/l and transferrin saturation below 20%, and (2) absolute iron deficiency-ferritin below 30 µg/l. RESULTS At baseline, a total of 54.5% of participants showed functional iron deficiency at a mean hemoglobin of 14.3 g/dl; while, the rate of absolute iron deficiency was 11.8%, at a mean hemoglobin level of 13.4 g/dl. At year 5, proportion of newly diagnosed subjects was 18.5% and 4.8%, respectively. Rate of all-cause mortality was 7.2% (n = 361); while, median follow-up was 10.1 years. After adjustment for hemoglobin and major cardiovascular risk factors, the hazard ratio with 95% confidence interval of the association of iron deficiency with mortality was 1.3 (1.0-1.6; p = 0.023) for the functional definition, and 1.9 (1.3-2.8; p = 0.002) for absolute iron deficiency. CONCLUSIONS Iron deficiency is very common in the apparently healthy general population and independently associated with all-cause mortality in the mid to long term.
Collapse
Affiliation(s)
- Benedikt Schrage
- Department of General and Interventional Cardiology, University Heart and Vascular Center Hamburg, Hamburg, Germany
- DZHK (German Center for Cardiovascular Research), Partner site, Hamburg/Kiel/Lübeck, Germany
| | - Nicole Rübsamen
- Department of General and Interventional Cardiology, University Heart and Vascular Center Hamburg, Hamburg, Germany
| | - Andreas Schulz
- Centre of Medicine II (Statistics), University Medical Center, Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Thomas Münzel
- DZHK (German Center for Cardiovascular Research), Partner Site Rhine-Main, Mainz, Germany
- Center for Cardiology, University Medical Center, Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Norbert Pfeiffer
- Department for Opthalmology, University Medical Center, Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Philipp S Wild
- DZHK (German Center for Cardiovascular Research), Partner Site Rhine-Main, Mainz, Germany
- Preventive Cardiology and Preventive Medicine, Center for Cardiology, University Medical Center, Johannes Gutenberg-University Mainz, Mainz, Germany
- Center for Thrombosis and Hemostasis, University Medical Center, Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Manfred Beutel
- Department of Psychosomatic Medicine and Psychotherapy, University Medical Center, Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Irene Schmidtmann
- Institute for Medical Biometry, Epidemiology and Informatics, University Medical Center Mainz, Mainz, Germany
| | - Rosemarie Lott
- Institute for Clinical Chemistry and Laboratory Medicine, University Medical Center Mainz, Mainz, Germany
| | - Stefan Blankenberg
- Department of General and Interventional Cardiology, University Heart and Vascular Center Hamburg, Hamburg, Germany
- DZHK (German Center for Cardiovascular Research), Partner site, Hamburg/Kiel/Lübeck, Germany
| | - Tanja Zeller
- Department of General and Interventional Cardiology, University Heart and Vascular Center Hamburg, Hamburg, Germany
- DZHK (German Center for Cardiovascular Research), Partner site, Hamburg/Kiel/Lübeck, Germany
| | - Karl J Lackner
- DZHK (German Center for Cardiovascular Research), Partner Site Rhine-Main, Mainz, Germany
- Institute for Clinical Chemistry and Laboratory Medicine, University Medical Center Mainz, Mainz, Germany
| | - Mahir Karakas
- Department of General and Interventional Cardiology, University Heart and Vascular Center Hamburg, Hamburg, Germany.
- DZHK (German Center for Cardiovascular Research), Partner site, Hamburg/Kiel/Lübeck, Germany.
| |
Collapse
|
28
|
Systematic Surveys of Iron Homeostasis Mechanisms Reveal Ferritin Superfamily and Nucleotide Surveillance Regulation to be Modified by PINK1 Absence. Cells 2020; 9:cells9102229. [PMID: 33023155 PMCID: PMC7650593 DOI: 10.3390/cells9102229] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Revised: 09/21/2020] [Accepted: 09/29/2020] [Indexed: 12/13/2022] Open
Abstract
Iron deprivation activates mitophagy and extends lifespan in nematodes. In patients suffering from Parkinson’s disease (PD), PINK1-PRKN mutations via deficient mitophagy trigger iron accumulation and reduce lifespan. To evaluate molecular effects of iron chelator drugs as a potential PD therapy, we assessed fibroblasts by global proteome profiles and targeted transcript analyses. In mouse cells, iron shortage decreased protein abundance for iron-binding nucleotide metabolism enzymes (prominently XDH and ferritin homolog RRM2). It also decreased the expression of factors with a role for nucleotide surveillance, which associate with iron-sulfur-clusters (ISC), and are important for growth and survival. This widespread effect included prominently Nthl1-Ppat-Bdh2, but also mitochondrial Glrx5-Nfu1-Bola1, cytosolic Aco1-Abce1-Tyw5, and nuclear Dna2-Elp3-Pold1-Prim2. Incidentally, upregulated Pink1-Prkn levels explained mitophagy induction, the downregulated expression of Slc25a28 suggested it to function in iron export. The impact of PINK1 mutations in mouse and patient cells was pronounced only after iron overload, causing hyperreactive expression of ribosomal surveillance factor Abce1 and of ferritin, despite ferritin translation being repressed by IRP1. This misregulation might be explained by the deficiency of the ISC-biogenesis factor GLRX5. Our systematic survey suggests mitochondrial ISC-biogenesis and post-transcriptional iron regulation to be important in the decision, whether organisms undergo PD pathogenesis or healthy aging.
Collapse
|
29
|
Altamura S, Marques O, Colucci S, Mertens C, Alikhanyan K, Muckenthaler MU. Regulation of iron homeostasis: Lessons from mouse models. Mol Aspects Med 2020; 75:100872. [DOI: 10.1016/j.mam.2020.100872] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2020] [Revised: 06/28/2020] [Accepted: 07/02/2020] [Indexed: 12/13/2022]
|
30
|
Iron-responsive-like elements and neurodegenerative ferroptosis. ACTA ACUST UNITED AC 2020; 27:395-413. [PMID: 32817306 PMCID: PMC7433652 DOI: 10.1101/lm.052282.120] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Accepted: 07/02/2020] [Indexed: 12/26/2022]
Abstract
A set of common-acting iron-responsive 5′untranslated region (5′UTR) motifs can fold into RNA stem loops that appear significant to the biology of cognitive declines of Parkinson's disease dementia (PDD), Lewy body dementia (LDD), and Alzheimer's disease (AD). Neurodegenerative diseases exhibit perturbations of iron homeostasis in defined brain subregions over characteristic time intervals of progression. While misfolding of Aβ from the amyloid-precursor-protein (APP), alpha-synuclein, prion protein (PrP) each cause neuropathic protein inclusions in the brain subregions, iron-responsive-like element (IRE-like) RNA stem–loops reside in their transcripts. APP and αsyn have a role in iron transport while gene duplications elevate the expression of their products to cause rare familial cases of AD and PDD. Of note, IRE-like sequences are responsive to excesses of brain iron in a potential feedback loop to accelerate neuronal ferroptosis and cognitive declines as well as amyloidosis. This pathogenic feedback is consistent with the translational control of the iron storage protein ferritin. We discuss how the IRE-like RNA motifs in the 5′UTRs of APP, alpha-synuclein and PrP mRNAs represent uniquely folded drug targets for therapies to prevent perturbed iron homeostasis that accelerates AD, PD, PD dementia (PDD) and Lewy body dementia, thus preventing cognitive deficits. Inhibition of alpha-synuclein translation is an option to block manganese toxicity associated with early childhood cognitive problems and manganism while Pb toxicity is epigenetically associated with attention deficit and later-stage AD. Pathologies of heavy metal toxicity centered on an embargo of iron export may be treated with activators of APP and ferritin and inhibitors of alpha-synuclein translation.
Collapse
|
31
|
Abstract
Iron deficiency or overload poses an increasingly complex issue in cardiovascular disease, especially heart failure. The potential benefits and side effects of iron supplementation are still a matter of concern, even though current guidelines suggest therapeutic management of iron deficiency. In this review, we sought to examine the iron metabolism and to identify the rationale behind iron supplementation and iron chelation. Cardiovascular disease is increasingly linked with iron dysmetabolism, with an increased proportion of heart failure patients being affected by decreased plasma iron levels and in turn, by the decreased quality of life. Multiple studies have concluded on a benefit of iron administration, even if just for symptomatic relief. However, new studies field evidence for negative effects of dysregulated non-bound iron and its reactive oxygen species production, with concern to heart diseases. The molecular targets of iron usage, such as the mitochondria, are prone to deleterious effects of the polyvalent metal, added by the scarcely described processes of iron elimination. Iron supplementation and iron chelation show promise of therapeutic benefit in heart failure, with the extent and mechanisms of both prospects not being entirely understood. It may be that a state of decreased systemic and increased mitochondrial iron levels proves to be a useful frame for future advancements in understanding the interconnection of heart failure and iron metabolism.
Collapse
|
32
|
Weidmann H, Bannasch JH, Waldeyer C, Shrivastava A, Appelbaum S, Ojeda-Echevarria FM, Schnabel R, Lackner KJ, Blankenberg S, Zeller T, Karakas M. Iron Metabolism Contributes to Prognosis in Coronary Artery Disease: Prognostic Value of the Soluble Transferrin Receptor Within the AtheroGene Study. J Am Heart Assoc 2020; 9:e015480. [PMID: 32321351 PMCID: PMC7428563 DOI: 10.1161/jaha.119.015480] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Background Coronary heart disease is a leading cause of mortality worldwide. Iron deficiency, a frequent comorbidity of coronary heart disease, causes an increased expression of transferrin receptor and soluble transferrin receptor levels (sTfR) levels, while iron repletion returns sTfR levels to the normal physiological range. Recently, sTfR levels were proposed as a potential new marker of iron metabolism in cardiovascular diseases. Therefore, we aimed to evaluate the prognostic value of circulating sTfR levels in a large cohort of patients with coronary heart disease. Methods and Results The disease cohort comprised 3423 subjects who had angiographically documented coronary heart disease and who participated in the AtheroGene study. Serum levels of sTfR were determined at baseline using an automated immunoassay (Roche Cobas Integra 400). Two main outcomes were considered: a combined end point of myocardial infarction and cardiovascular death and cardiovascular death alone. During a median follow‐up of 4.0 years, 10.3% of the patients experienced an end point. In Cox regression analyses for sTfR levels, the hazard ratio (HR) for future cardiovascular death and/or myocardial infarction was 1.27 (95% CI, 1.11–1.44, P<0.001) after adjustment for sex and age. This association remained significant (HR, 1.23; 95% CI, 1.03–1.46, P=0.02) after additional adjustment for body mass index, smoking status, hypertension, diabetes mellitus, dyslipidemia, C‐reactive protein, and surrogates of cardiac function, size of myocardial necrosis (hs‐Tnl), and hemoglobin levels. Conclusions In this large cohort study, sTfR levels were strongly associated with future myocardial infarction and cardiovascular death. This implicates a role for sTfR in secondary cardiovascular risk prediction.
Collapse
Affiliation(s)
- Henri Weidmann
- Department of General and Interventional Cardiology University Heart Center Hamburg Hamburg Germany.,German Center for Cardiovascular Research (DZHK), Partner Site Hamburg, Lübeck, Kiel Hamburg Germany
| | - Johannes H Bannasch
- Department of General and Interventional Cardiology University Heart Center Hamburg Hamburg Germany
| | - Christoph Waldeyer
- Department of General and Interventional Cardiology University Heart Center Hamburg Hamburg Germany.,German Center for Cardiovascular Research (DZHK), Partner Site Hamburg, Lübeck, Kiel Hamburg Germany
| | - Apurva Shrivastava
- Department of General and Interventional Cardiology University Heart Center Hamburg Hamburg Germany.,German Center for Cardiovascular Research (DZHK), Partner Site Hamburg, Lübeck, Kiel Hamburg Germany
| | - Sebastian Appelbaum
- Department of General and Interventional Cardiology University Heart Center Hamburg Hamburg Germany
| | | | - Renate Schnabel
- Department of General and Interventional Cardiology University Heart Center Hamburg Hamburg Germany.,German Center for Cardiovascular Research (DZHK), Partner Site Hamburg, Lübeck, Kiel Hamburg Germany
| | - Karl J Lackner
- Department of Laboratory Medicine University Medical Center Johannes Gutenberg University Mainz Mainz Germany.,German Center for Cardiovascular Research (DZHK), Partner Site Rhein-Main Mainz Germany
| | - Stefan Blankenberg
- Department of General and Interventional Cardiology University Heart Center Hamburg Hamburg Germany.,German Center for Cardiovascular Research (DZHK), Partner Site Hamburg, Lübeck, Kiel Hamburg Germany
| | - Tanja Zeller
- Department of General and Interventional Cardiology University Heart Center Hamburg Hamburg Germany.,German Center for Cardiovascular Research (DZHK), Partner Site Hamburg, Lübeck, Kiel Hamburg Germany
| | - Mahir Karakas
- Department of General and Interventional Cardiology University Heart Center Hamburg Hamburg Germany.,German Center for Cardiovascular Research (DZHK), Partner Site Hamburg, Lübeck, Kiel Hamburg Germany
| |
Collapse
|
33
|
Schrage B, Rübsamen N, Schulz A, Münzel T, Pfeiffer N, Wild PS, Beutel M, Schmidtmann I, Lott R, Blankenberg S, Zeller T, Lackner KJ, Karakas M. Iron deficiency is a common disorder in general population and independently predicts all-cause mortality: results from the Gutenberg Health Study. Clin Res Cardiol 2020; 109:1352-1357. [PMID: 32215702 PMCID: PMC7588396 DOI: 10.1007/s00392-020-01631-y] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/24/2019] [Accepted: 03/11/2020] [Indexed: 01/01/2023]
Abstract
Background Iron deficiency is now accepted as an independent entity beyond anemia. Recently, a new functional definition of iron deficiency was proposed and proved strong efficacy in randomized cardiovascular clinical trials of intravenous iron supplementation. Here, we characterize the impact of iron deficiency on all-cause mortality in the non-anemic general population based on two distinct definitions. Methods The Gutenberg Health Study is a population-based, prospective, single-center cohort study. The 5000 individuals between 35 and 74 years underwent baseline and a planned follow-up visit at year 5. Tested definitions of iron deficiency were (1) functional iron deficiency—ferritin levels below 100 µg/l, or ferritin levels between 100 and 299 µg/l and transferrin saturation below 20%, and (2) absolute iron deficiency—ferritin below 30 µg/l. Results At baseline, a total of 54.5% of participants showed functional iron deficiency at a mean hemoglobin of 14.3 g/dl; while, the rate of absolute iron deficiency was 11.8%, at a mean hemoglobin level of 13.4 g/dl. At year 5, proportion of newly diagnosed subjects was 18.5% and 4.8%, respectively. Rate of all-cause mortality was 7.2% (n = 361); while, median follow-up was 10.1 years. After adjustment for hemoglobin and major cardiovascular risk factors, the hazard ratio with 95% confidence interval of the association of iron deficiency with mortality was 1.3 (1.0–1.6; p = 0.023) for the functional definition, and 1.9 (1.3–2.8; p = 0.002) for absolute iron deficiency. Conclusions Iron deficiency is very common in the apparently healthy general population and independently associated with all-cause mortality in the mid to long term. Graphic abstract ![]()
Collapse
Affiliation(s)
- Benedikt Schrage
- Department of General and Interventional Cardiology, University Heart and Vascular Center Hamburg, Hamburg, Germany.,DZHK (German Center for Cardiovascular Research), Partner site, Hamburg/Kiel/Lübeck, Germany
| | - Nicole Rübsamen
- Department of General and Interventional Cardiology, University Heart and Vascular Center Hamburg, Hamburg, Germany
| | - Andreas Schulz
- Centre of Medicine II (Statistics), University Medical Center, Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Thomas Münzel
- DZHK (German Center for Cardiovascular Research), Partner Site Rhine-Main, Mainz, Germany.,Center for Cardiology, University Medical Center, Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Norbert Pfeiffer
- Department for Opthalmology, University Medical Center, Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Philipp S Wild
- DZHK (German Center for Cardiovascular Research), Partner Site Rhine-Main, Mainz, Germany.,Preventive Cardiology and Preventive Medicine, Center for Cardiology, University Medical Center, Johannes Gutenberg-University Mainz, Mainz, Germany.,Center for Thrombosis and Hemostasis, University Medical Center, Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Manfred Beutel
- Department of Psychosomatic Medicine and Psychotherapy, University Medical Center, Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Irene Schmidtmann
- Institute for Medical Biometry, Epidemiology and Informatics, University Medical Center Mainz, Mainz, Germany
| | - Rosemarie Lott
- Institute for Clinical Chemistry and Laboratory Medicine, University Medical Center Mainz, Mainz, Germany
| | - Stefan Blankenberg
- Department of General and Interventional Cardiology, University Heart and Vascular Center Hamburg, Hamburg, Germany.,DZHK (German Center for Cardiovascular Research), Partner site, Hamburg/Kiel/Lübeck, Germany
| | - Tanja Zeller
- Department of General and Interventional Cardiology, University Heart and Vascular Center Hamburg, Hamburg, Germany.,DZHK (German Center for Cardiovascular Research), Partner site, Hamburg/Kiel/Lübeck, Germany
| | - Karl J Lackner
- DZHK (German Center for Cardiovascular Research), Partner Site Rhine-Main, Mainz, Germany.,Institute for Clinical Chemistry and Laboratory Medicine, University Medical Center Mainz, Mainz, Germany
| | - Mahir Karakas
- Department of General and Interventional Cardiology, University Heart and Vascular Center Hamburg, Hamburg, Germany. .,DZHK (German Center for Cardiovascular Research), Partner site, Hamburg/Kiel/Lübeck, Germany.
| |
Collapse
|
34
|
Leermakers PA, Remels AHV, Zonneveld MI, Rouschop KMA, Schols AMWJ, Gosker HR. Iron deficiency-induced loss of skeletal muscle mitochondrial proteins and respiratory capacity; the role of mitophagy and secretion of mitochondria-containing vesicles. FASEB J 2020; 34:6703-6717. [PMID: 32202346 DOI: 10.1096/fj.201901815r] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2019] [Revised: 02/18/2020] [Accepted: 03/12/2020] [Indexed: 12/13/2022]
Abstract
Iron homeostasis is essential for mitochondrial function, and iron deficiency has been associated with skeletal muscle weakness and decreased exercise capacity in patients with different chronic disorders. We hypothesized that iron deficiency-induced loss of skeletal muscle mitochondria is caused by increased mitochondrial clearance. To study this, C2C12 myotubes were subjected to the iron chelator deferiprone. Mitochondrial parameters and key constituents of mitophagy pathways were studied in presence or absence of pharmacological autophagy inhibition or knockdown of mitophagy-related proteins. Furthermore, it was explored if mitochondria were present in extracellular vesicles (EV). Iron chelation resulted in an increase in BCL2/Adenovirus E1B 19 kDa protein-interacting protein 3 (BNIP3) and BNIP3-like gene and protein levels, and the appearance of mitochondria encapsulated by lysosome-like vesicular structures in myotubes. Moreover, mitochondria were secreted via EV. These changes were associated with cellular mitochondrial impairments. These impairments were unaltered by autophagy inhibition, knockdown of mitophagy-related proteins BNIP3 and BNIP3L, or knockdown of their upstream regulator hypoxia-inducible factor 1 alpha. In conclusion, mitophagy is not essential for development of iron deficiency-induced reductions in mitochondrial proteins or respiratory capacity. The secretion of mitochondria-containing EV could present an additional pathway via which mitochondria can be cleared from iron chelation-exposed myotubes.
Collapse
Affiliation(s)
- Pieter A Leermakers
- Department of Respiratory Medicine, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Centre+, Maastricht, the Netherlands
| | - Alexander H V Remels
- Department of Pharmacology and Toxicology, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Centre+, Maastricht, the Netherlands
| | - Marijke I Zonneveld
- Department of Radiotherapy, GROW School for Oncology and Developmental Biology, Maastricht University Medical Centre+, Maastricht, the Netherlands
| | - Kasper M A Rouschop
- Department of Radiotherapy, GROW School for Oncology and Developmental Biology, Maastricht University Medical Centre+, Maastricht, the Netherlands
| | - Annemie M W J Schols
- Department of Respiratory Medicine, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Centre+, Maastricht, the Netherlands
| | - Harry R Gosker
- Department of Respiratory Medicine, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Centre+, Maastricht, the Netherlands
| |
Collapse
|
35
|
Hernández-Gallardo AK, Missirlis F. Cellular iron sensing and regulation: Nuclear IRP1 extends a classic paradigm. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2020; 1867:118705. [PMID: 32199885 DOI: 10.1016/j.bbamcr.2020.118705] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/21/2020] [Revised: 03/02/2020] [Accepted: 03/16/2020] [Indexed: 01/26/2023]
Abstract
The classic view is that iron regulatory proteins operate at the post-transcriptional level. Iron Regulatory Protein 1 (IRP1) shifts between an apo-form that binds mRNAs and a holo-form that harbors a [4Fe4S] cluster. The latter form is not considered relevant to iron regulation, but rather thought to act as a non-essential cytosolic aconitase. Recent work in Drosophila, however, shows that holo-IRP1 can also translocate to the nucleus, where it appears to downregulate iron metabolism genes, preparing the cell for a decline in iron uptake. The shifting of IRP1 between states requires a functional mitoNEET pathway that includes a glycogen branching enzyme for the repair or disassembly of IRP1's oxidatively damaged [3Fe4S] cluster. The new findings add to the notion that glucose metabolism is modulated by iron metabolism. Furthermore, we propose that ferritin ferroxidase activity participates in the repair of the IRP1 [3Fe4S] cluster leading to the hypothesis that cytosolic ferritin directly contributes to cellular iron sensing.
Collapse
Affiliation(s)
| | - Fanis Missirlis
- Departamento de Fisiología, Biofísica y Neurociencias, Cinvestav, CDMX, Mexico.
| |
Collapse
|
36
|
Santos MCFD, Anderson CP, Neschen S, Zumbrennen-Bullough KB, Romney SJ, Kahle-Stephan M, Rathkolb B, Gailus-Durner V, Fuchs H, Wolf E, Rozman J, de Angelis MH, Cai WM, Rajan M, Hu J, Dedon PC, Leibold EA. Irp2 regulates insulin production through iron-mediated Cdkal1-catalyzed tRNA modification. Nat Commun 2020; 11:296. [PMID: 31941883 PMCID: PMC6962211 DOI: 10.1038/s41467-019-14004-5] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2018] [Accepted: 12/06/2019] [Indexed: 12/12/2022] Open
Abstract
Regulation of cellular iron homeostasis is crucial as both iron excess and deficiency cause hematological and neurodegenerative diseases. Here we show that mice lacking iron-regulatory protein 2 (Irp2), a regulator of cellular iron homeostasis, develop diabetes. Irp2 post-transcriptionally regulates the iron-uptake protein transferrin receptor 1 (TfR1) and the iron-storage protein ferritin, and dysregulation of these proteins due to Irp2 loss causes functional iron deficiency in β cells. This impairs Fe-S cluster biosynthesis, reducing the function of Cdkal1, an Fe-S cluster enzyme that catalyzes methylthiolation of t6A37 in tRNALysUUU to ms2t6A37. As a consequence, lysine codons in proinsulin are misread and proinsulin processing is impaired, reducing insulin content and secretion. Iron normalizes ms2t6A37 and proinsulin lysine incorporation, restoring insulin content and secretion in Irp2-/- β cells. These studies reveal a previously unidentified link between insulin processing and cellular iron deficiency that may have relevance to type 2 diabetes in humans.
Collapse
Affiliation(s)
- Maria C Ferreira Dos Santos
- Department of Medicine, Division of Hematology, University of Utah, Salt Lake City, UT, 84112, USA.,Molecular Medicine Program, University of Utah, Salt Lake City, UT, 84112, USA
| | - Cole P Anderson
- Molecular Medicine Program, University of Utah, Salt Lake City, UT, 84112, USA.,Department of Oncological Sciences, University of Utah, Salt Lake City, UT, 84112, USA.,Landstuhl Regional Medical Center, 66849, Landstuhl, Germany
| | - Susanne Neschen
- German Mouse Clinic, Institute of Experimental Genetics, Helmholtz Zentrum München, Ingolstädter Landstraße 1, 85764, Neuherberg, Germany.,German Center for Diabetes Research (DZD), Ingolstädter Landstraße 1, 85764, Neuherberg, Germany
| | - Kimberly B Zumbrennen-Bullough
- Molecular Medicine Program, University of Utah, Salt Lake City, UT, 84112, USA.,Department of Oncological Sciences, University of Utah, Salt Lake City, UT, 84112, USA
| | - Steven J Romney
- Department of Medicine, Division of Hematology, University of Utah, Salt Lake City, UT, 84112, USA.,Molecular Medicine Program, University of Utah, Salt Lake City, UT, 84112, USA.,Thermo Fisher Scientific, Waltham, MA, 02451, USA
| | - Melanie Kahle-Stephan
- German Mouse Clinic, Institute of Experimental Genetics, Helmholtz Zentrum München, Ingolstädter Landstraße 1, 85764, Neuherberg, Germany.,Medizinische Hochschule Brandenburg Theodor Fontane Institut für Sozialmedizin und Epidemiologie, 14770, Brandenburg an der Havel, Germany
| | - Birgit Rathkolb
- German Mouse Clinic, Institute of Experimental Genetics, Helmholtz Zentrum München, Ingolstädter Landstraße 1, 85764, Neuherberg, Germany.,German Center for Diabetes Research (DZD), Ingolstädter Landstraße 1, 85764, Neuherberg, Germany.,Institute of Molecular Animal Breeding and Biotechnology, Gene Center, Ludwig-Maximilians-Universität München, Feodor-Lynen Strasse 25, 81377, Munich, Germany
| | - Valerie Gailus-Durner
- German Mouse Clinic, Institute of Experimental Genetics, Helmholtz Zentrum München, Ingolstädter Landstraße 1, 85764, Neuherberg, Germany
| | - Helmut Fuchs
- German Mouse Clinic, Institute of Experimental Genetics, Helmholtz Zentrum München, Ingolstädter Landstraße 1, 85764, Neuherberg, Germany
| | - Eckhard Wolf
- Institute of Molecular Animal Breeding and Biotechnology, Gene Center, Ludwig-Maximilians-Universität München, Feodor-Lynen Strasse 25, 81377, Munich, Germany
| | - Jan Rozman
- German Mouse Clinic, Institute of Experimental Genetics, Helmholtz Zentrum München, Ingolstädter Landstraße 1, 85764, Neuherberg, Germany.,German Center for Diabetes Research (DZD), Ingolstädter Landstraße 1, 85764, Neuherberg, Germany.,Czech Centre for Phenogenomics, Institute of Molecular Genetics of the Czech Academy of Sciences, Prumyslova, 595, 252 50 Vestec, Czech Republic
| | - Martin Hrabe de Angelis
- German Mouse Clinic, Institute of Experimental Genetics, Helmholtz Zentrum München, Ingolstädter Landstraße 1, 85764, Neuherberg, Germany.,German Center for Diabetes Research (DZD), Ingolstädter Landstraße 1, 85764, Neuherberg, Germany.,Chair of Experimental Genetics, School of Life Science Weihenstephan, Technische Universität München, Alte Akademie 8, 85354, Freising, Germany
| | - Weiling Maggie Cai
- Department of Microbiology, National University of Singapore, Singapore, Singapore, 119077.,Antimicrobial Resistance Interdisciplinary Research Group (IRG), Singapore-MIT Alliance for Research and Technology, 1 CREATE Way, Singapore, Singapore, 138602.,Agilent Technologies, 1 Yishun Ave 7, Singapore, Singapore, 768923
| | - Malini Rajan
- Department of Medicine, Division of Hematology, University of Utah, Salt Lake City, UT, 84112, USA.,Molecular Medicine Program, University of Utah, Salt Lake City, UT, 84112, USA
| | - Jennifer Hu
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA.,Celgene Corporation, 1616 Eastlake Ave East, Seattle, WA, 98102, USA
| | - Peter C Dedon
- Antimicrobial Resistance Interdisciplinary Research Group (IRG), Singapore-MIT Alliance for Research and Technology, 1 CREATE Way, Singapore, Singapore, 138602.,Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
| | - Elizabeth A Leibold
- Department of Medicine, Division of Hematology, University of Utah, Salt Lake City, UT, 84112, USA. .,Molecular Medicine Program, University of Utah, Salt Lake City, UT, 84112, USA. .,Department of Oncological Sciences, University of Utah, Salt Lake City, UT, 84112, USA.
| |
Collapse
|
37
|
Telser J, Volani C, Hilbe R, Seifert M, Brigo N, Paglia G, Weiss G. Metabolic reprogramming of Salmonella infected macrophages and its modulation by iron availability and the mTOR pathway. MICROBIAL CELL 2019; 6:531-543. [PMID: 31832425 PMCID: PMC6883347 DOI: 10.15698/mic2019.12.700] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Iron is an essential nutrient for immune cells and microbes, therefore the control of its homeostasis plays a decisive role for infections. Moreover, iron affects metabolic pathways by modulating the translational expression of the key tricarboxylic acid cycle (TCA) enzyme mitochondrial aconitase and the energy formation by mitochondria. Recent data provide evidence for metabolic re-programming of immune cells including macrophages during infection which is centrally controlled by mTOR. We herein studied the effects of iron perturbations on metabolic profiles in macrophages upon infection with the intracellular bacterium Salmonella enterica serovar Typhimurium and analysed for a link to the mTOR pathway. Infection of the murine macrophage cell line RAW264.7 with Salmonella resulted in the induction of mTOR activity, anaerobic glycolysis and inhibition of the TCA activity as reflected by reduced pyruvate and increased lactate levels. In contrast, iron supplementation to macrophages not only affected the mRNA expression of TCA and glycolytic enzymes but also resulted in metabolic reprogramming with increased pyruvate accumulation and reduced lactate levels apart from modulating the concentrations of several other metabolites. While mTOR slightly affected cellular iron homeostasis in infected macrophages, mTOR inhibition by rapamycin resulted in a significant growth promotion of bacteria. Importantly, iron further increased bacterial numbers in rapamycin treated macrophages, however, the metabolic profiles induced by iron in the presence or absence of mTOR activity differed in several aspects. Our data indicate, that iron not only serves as a bacterial nutrient but also acts as a metabolic modulator of the TCA cycle, partly reversing the Warburg effect and resulting in a pathogen friendly nutritional environment.
Collapse
Affiliation(s)
- Julia Telser
- Department of Internal Medicine II, Medical University of Innsbruck, Austria.,Christian Doppler Laboratory for Iron Metabolism and Anemia Research, Medical University of Innsbruck, Austria
| | - Chiara Volani
- Department of Internal Medicine II, Medical University of Innsbruck, Austria.,EURAC Research, Institute for Biomedicine, Bolzano/Bozen, Italy
| | - Richard Hilbe
- Department of Internal Medicine II, Medical University of Innsbruck, Austria.,Christian Doppler Laboratory for Iron Metabolism and Anemia Research, Medical University of Innsbruck, Austria
| | - Markus Seifert
- Department of Internal Medicine II, Medical University of Innsbruck, Austria.,Christian Doppler Laboratory for Iron Metabolism and Anemia Research, Medical University of Innsbruck, Austria
| | - Natascha Brigo
- Department of Internal Medicine II, Medical University of Innsbruck, Austria
| | | | - Günter Weiss
- Department of Internal Medicine II, Medical University of Innsbruck, Austria.,Christian Doppler Laboratory for Iron Metabolism and Anemia Research, Medical University of Innsbruck, Austria
| |
Collapse
|
38
|
Peoc'h K, Nicolas G, Schmitt C, Mirmiran A, Daher R, Lefebvre T, Gouya L, Karim Z, Puy H. Regulation and tissue-specific expression of δ-aminolevulinic acid synthases in non-syndromic sideroblastic anemias and porphyrias. Mol Genet Metab 2019; 128:190-197. [PMID: 30737140 DOI: 10.1016/j.ymgme.2019.01.015] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/17/2018] [Revised: 01/21/2019] [Accepted: 01/21/2019] [Indexed: 02/07/2023]
Abstract
Recently, new genes and molecular mechanisms have been identified in patients with porphyrias and sideroblastic anemias (SA). They all modulate either directly or indirectly the δ-aminolevulinic acid synthase (ALAS) activity. ALAS, is encoded by two genes: the erythroid-specific (ALAS2), and the ubiquitously expressed (ALAS1). In the liver, ALAS1 controls the rate-limiting step in the production of heme and hemoproteins that are rapidly turned over in response to metabolic needs. Several heme regulatory targets have been identified as regulators of ALAS1 activity: 1) transcriptional repression via a heme-responsive element, 2) post-transcriptional destabilization of ALAS1 mRNA, 3) post-translational inhibition via a heme regulatory motif, 4) direct inhibition of the activity of the enzyme and 5) breakdown of ALAS1 protein via heme-mediated induction of the protease Lon peptidase 1. In erythroid cells, ALAS2 is a gatekeeper of production of very large amounts of heme necessary for hemoglobin synthesis. The rate of ALAS2 synthesis is transiently increased during the period of active heme synthesis. Its gene expression is determined by trans-activation of nuclear factor GATA1, CACC box and NF-E2-binding sites in the promoter areas. ALAS2 mRNA translation is also regulated by the iron-responsive element (IRE)/iron regulatory proteins (IRP) binding system. In patients, ALAS enzyme activity is affected in most of the mutations causing non-syndromic SA and in several porphyrias. Decreased ALAS2 activity results either directly from loss-of-function ALAS2 mutations as seen in X-linked sideroblastic anemia (XLSA) or from defect in the availability of one of its two mitochondrial substrates: glycine in SLC25A38 mutations and succinyl CoA in GLRX5 mutations. Moreover, ALAS2 gain of function mutations is responsible for X-linked protoporphyria and increased ALAS1 activity lead to acute attacks of hepatic porphyrias. A missense dominant mutation in the Walker A motif of the ATPase binding site in the gene coding for the mitochondrial protein unfoldase CLPX also contributes to increasing ALAS and subsequently protoporphyrinemia. Altogether, these recent data on human ALAS have informed our understanding of porphyrias and sideroblastic anemias pathogeneses and may contribute to new therapeutic strategies.
Collapse
Affiliation(s)
- Katell Peoc'h
- INSERM U1149, CNRS ERL 8252, Centre de Recherche sur l'inflammation, Université Paris Diderot, site Bichat, Sorbonne Paris Cité, France, 16 rue Henri Huchard, 75018 Paris, France; Laboratory of Excellence, GR-Ex, Paris, France.
| | - Gaël Nicolas
- INSERM U1149, CNRS ERL 8252, Centre de Recherche sur l'inflammation, Université Paris Diderot, site Bichat, Sorbonne Paris Cité, France, 16 rue Henri Huchard, 75018 Paris, France; Laboratory of Excellence, GR-Ex, Paris, France.
| | - Caroline Schmitt
- INSERM U1149, CNRS ERL 8252, Centre de Recherche sur l'inflammation, Université Paris Diderot, site Bichat, Sorbonne Paris Cité, France, 16 rue Henri Huchard, 75018 Paris, France; Laboratory of Excellence, GR-Ex, Paris, France; AP-HP, HUPNVS, Centre Français des Porphyries, Hôpital Louis Mourier, Colombes, France.
| | - Arienne Mirmiran
- INSERM U1149, CNRS ERL 8252, Centre de Recherche sur l'inflammation, Université Paris Diderot, site Bichat, Sorbonne Paris Cité, France, 16 rue Henri Huchard, 75018 Paris, France; Laboratory of Excellence, GR-Ex, Paris, France.
| | - Raed Daher
- INSERM U1149, CNRS ERL 8252, Centre de Recherche sur l'inflammation, Université Paris Diderot, site Bichat, Sorbonne Paris Cité, France, 16 rue Henri Huchard, 75018 Paris, France; Laboratory of Excellence, GR-Ex, Paris, France.
| | - Thibaud Lefebvre
- INSERM U1149, CNRS ERL 8252, Centre de Recherche sur l'inflammation, Université Paris Diderot, site Bichat, Sorbonne Paris Cité, France, 16 rue Henri Huchard, 75018 Paris, France; Laboratory of Excellence, GR-Ex, Paris, France; AP-HP, HUPNVS, Centre Français des Porphyries, Hôpital Louis Mourier, Colombes, France.
| | - Laurent Gouya
- INSERM U1149, CNRS ERL 8252, Centre de Recherche sur l'inflammation, Université Paris Diderot, site Bichat, Sorbonne Paris Cité, France, 16 rue Henri Huchard, 75018 Paris, France; Laboratory of Excellence, GR-Ex, Paris, France; AP-HP, HUPNVS, Centre Français des Porphyries, Hôpital Louis Mourier, Colombes, France.
| | - Zoubida Karim
- INSERM U1149, CNRS ERL 8252, Centre de Recherche sur l'inflammation, Université Paris Diderot, site Bichat, Sorbonne Paris Cité, France, 16 rue Henri Huchard, 75018 Paris, France; Laboratory of Excellence, GR-Ex, Paris, France.
| | - Hervé Puy
- INSERM U1149, CNRS ERL 8252, Centre de Recherche sur l'inflammation, Université Paris Diderot, site Bichat, Sorbonne Paris Cité, France, 16 rue Henri Huchard, 75018 Paris, France; Laboratory of Excellence, GR-Ex, Paris, France; AP-HP, HUPNVS, Centre Français des Porphyries, Hôpital Louis Mourier, Colombes, France.
| |
Collapse
|
39
|
Zielonka M, Kölker S, Gleich F, Stützenberger N, Nagamani SCS, Gropman AL, Hoffmann GF, Garbade SF, Posset R. Early prediction of phenotypic severity in Citrullinemia Type 1. Ann Clin Transl Neurol 2019; 6:1858-1871. [PMID: 31469252 PMCID: PMC6764635 DOI: 10.1002/acn3.50886] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2019] [Accepted: 08/14/2019] [Indexed: 12/31/2022] Open
Abstract
Objective Citrullinemia type 1 (CTLN1) is an inherited metabolic disease affecting the brain which is detectable by newborn screening. The clinical spectrum is highly variable including individuals with lethal hyperammonemic encephalopathy in the newborn period and individuals with a mild‐to‐moderate or asymptomatic disease course. Since the phenotypic severity has not been predictable early during the disease course so far, we aimed to design a reliable disease prediction model. Methods We used a newly established mammalian biallelic expression system to determine residual enzymatic activity of argininosuccinate synthetase 1 (ASS1; OMIM #215700) in 71 individuals with CTLN1, representing 48 ASS1 gene variants and 50 different, mostly compound heterozygous combinations in total. Residual enzymatic ASS1 activity was correlated to standardized biochemical and clinical endpoints available from the UCDC and E‐IMD databases. Results Residual enzymatic ASS1 activity correlates with peak plasma ammonium and L‐citrulline concentrations at initial presentation. Individuals with 8% of residual enzymatic ASS1 activity or less had more frequent and more severe hyperammonemic events and lower cognitive function than those above 8%, highlighting that residual enzymatic ASS1 activity allows reliable severity prediction. Noteworthy, empiric clinical practice of affected individuals is in line with the predicted disease severity supporting the notion of a risk stratification‐based guidance of therapeutic decision‐making based on residual enzymatic ASS1 activity in the future. Interpretation Residual enzymatic ASS1 activity reliably predicts the phenotypic severity in CTLN1. We propose a new severity‐adjusted classification system for individuals with CTLN1 based on the activity results of the newly established biallelic expression system.
Collapse
Affiliation(s)
- Matthias Zielonka
- Center for Pediatric and Adolescent Medicine, Division of Pediatric Neurology and Metabolic Medicine, University Hospital Heidelberg, Im Neuenheimer Feld 430, 69120, Heidelberg, Germany.,Heidelberg Research Center for Molecular Medicine (HRCMM), Heidelberg, Germany
| | - Stefan Kölker
- Center for Pediatric and Adolescent Medicine, Division of Pediatric Neurology and Metabolic Medicine, University Hospital Heidelberg, Im Neuenheimer Feld 430, 69120, Heidelberg, Germany
| | - Florian Gleich
- Center for Pediatric and Adolescent Medicine, Division of Pediatric Neurology and Metabolic Medicine, University Hospital Heidelberg, Im Neuenheimer Feld 430, 69120, Heidelberg, Germany
| | - Nicolas Stützenberger
- Center for Pediatric and Adolescent Medicine, Division of Pediatric Neurology and Metabolic Medicine, University Hospital Heidelberg, Im Neuenheimer Feld 430, 69120, Heidelberg, Germany
| | - Sandesh C S Nagamani
- Department of Molecular and Human Genetics, Baylor College of Medicine, Texas Children's Hospital, Houston, Texas
| | - Andrea L Gropman
- Children's National Health System, The George Washington School of Medicine, District of Columbia, Washington
| | - Georg F Hoffmann
- Center for Pediatric and Adolescent Medicine, Division of Pediatric Neurology and Metabolic Medicine, University Hospital Heidelberg, Im Neuenheimer Feld 430, 69120, Heidelberg, Germany
| | - Sven F Garbade
- Center for Pediatric and Adolescent Medicine, Division of Pediatric Neurology and Metabolic Medicine, University Hospital Heidelberg, Im Neuenheimer Feld 430, 69120, Heidelberg, Germany
| | - Roland Posset
- Center for Pediatric and Adolescent Medicine, Division of Pediatric Neurology and Metabolic Medicine, University Hospital Heidelberg, Im Neuenheimer Feld 430, 69120, Heidelberg, Germany
| | | |
Collapse
|
40
|
Yang L, Li X, Wu Y, Zhang J, Li W, Wang Q. Iron regulatory protein is involved in the immune defense of the Chinese mitten crab Eriocheir sinensis. FISH & SHELLFISH IMMUNOLOGY 2019; 89:632-640. [PMID: 30995542 DOI: 10.1016/j.fsi.2019.04.041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/25/2018] [Revised: 04/04/2019] [Accepted: 04/11/2019] [Indexed: 06/09/2023]
Abstract
Iron homeostasis is vital to organismal health; it is maintained by the iron regulatory protein (IRP)-iron-responsive element (IRE) signaling pathway. In the Chinese mitten crab Eriocheir sinensis, EsFer-1 and EsFer-2 reportedly have a putative IRE, but an IRP has not yet been identified. In this study, we successfully amplified the full-length cDNA of EsIRP using gene cloning and rapid amplification of cDNA ends techniques. The length of this cDNA was 4474 bp, and it included a 2682-bp open reading frame encoding 893 amino acids. Using quantitative real-time PCR, mRNA transcripts of EsIRP were detected in various tissues. The highest and lowest expression level was detected in the muscle and gills, respectively. In response to Staphylococcus aureus and Vibrio parahaemolyticus challenge, the transcription level of EsIRP was downregulated and that of EsFer-1 and EsFer-2 was upregulated in hemocytes. EsIRP knockdown resulted in increased expression of both EsFer-1 and EsFer-2. After EsFer-1 and EsFer-2 knockdown, the bacterial clearance ability of E. sinensis against S. aureus and V. parahaemolyticus was impaired. In conclusion, our results suggest that the IRP-IRE signaling pathway plays an important role in the innate immune system response in E. sinensis.
Collapse
Affiliation(s)
- Lei Yang
- Laboratory of Invertebrate Immunological Defense and Reproductive Biology, School of Life Sciences, East China Normal University, Shanghai, China
| | - Xuejie Li
- Laboratory of Invertebrate Immunological Defense and Reproductive Biology, School of Life Sciences, East China Normal University, Shanghai, China
| | - Yaomeng Wu
- Laboratory of Invertebrate Immunological Defense and Reproductive Biology, School of Life Sciences, East China Normal University, Shanghai, China
| | - Jiashun Zhang
- Laboratory of Invertebrate Immunological Defense and Reproductive Biology, School of Life Sciences, East China Normal University, Shanghai, China
| | - Weiwei Li
- Laboratory of Invertebrate Immunological Defense and Reproductive Biology, School of Life Sciences, East China Normal University, Shanghai, China
| | - Qun Wang
- Laboratory of Invertebrate Immunological Defense and Reproductive Biology, School of Life Sciences, East China Normal University, Shanghai, China.
| |
Collapse
|
41
|
Paterek A, Mackiewicz U, Mączewski M. Iron and the heart: A paradigm shift from systemic to cardiomyocyte abnormalities. J Cell Physiol 2019; 234:21613-21629. [DOI: 10.1002/jcp.28820] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2018] [Revised: 04/16/2019] [Accepted: 04/17/2019] [Indexed: 12/13/2022]
Affiliation(s)
- Aleksandra Paterek
- Department of Clinical Physiology Centre of Postgraduate Medical Education Warsaw Poland
| | - Urszula Mackiewicz
- Department of Clinical Physiology Centre of Postgraduate Medical Education Warsaw Poland
| | - Michał Mączewski
- Department of Clinical Physiology Centre of Postgraduate Medical Education Warsaw Poland
| |
Collapse
|
42
|
Iron regulatory protein 2 modulates the switch from aerobic glycolysis to oxidative phosphorylation in mouse embryonic fibroblasts. Proc Natl Acad Sci U S A 2019; 116:9871-9876. [PMID: 31040213 DOI: 10.1073/pnas.1820051116] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
The importance of the role of iron regulatory proteins (IRPs) in mitochondrial iron homeostasis and function has been raised. To understand how an IRP affects mitochondrial function, we used globally Irp2-depleted mouse embryonic fibroblasts (MEFs) and found that Irp2 ablation significantly induced the expression of both hypoxia-inducible factor subunits, Hif1α and Hif2α. The increase of Hif1α up-regulated its targeted genes, enhancing glycolysis, and the increase of Hif2α down-regulated the expression of iron-sulfur cluster (Fe-S) biogenesis-related and electron transport chain (ETC)-related genes, weakening mitochondrial respiration. Inhibition of Hif1α by genetic knockdown or a specific inhibitor prevented Hif1α-targeted gene expression, leading to decreased aerobic glycolysis. Inhibition of Hif2α by genetic knockdown or selective disruption of the heterodimerization of Hif2α and Hif1β restored the mitochondrial ETC and coupled oxidative phosphorylation (OXPHOS) by enhancing Fe-S biogenesis and increasing ETC-related gene expression. Our results indicate that Irp2 modulates the metabolic switch from aerobic glycolysis to OXPHOS that is mediated by Hif1α and Hif2α in MEFs.
Collapse
|
43
|
Ferritin regulates organismal energy balance and thermogenesis. Mol Metab 2019; 24:64-79. [PMID: 30954544 PMCID: PMC6531837 DOI: 10.1016/j.molmet.2019.03.008] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/03/2019] [Revised: 03/13/2019] [Accepted: 03/15/2019] [Indexed: 01/13/2023] Open
Abstract
OBJECTIVE The ferritin heavy/heart chain (FTH) gene encodes the ferroxidase component of the iron (Fe) sequestering ferritin complex, which plays a central role in the regulation of cellular Fe metabolism. Here we tested the hypothesis that ferritin regulates organismal Fe metabolism in a manner that impacts energy balance and thermal homeostasis. METHODS We developed a mouse strain, referred herein as FthR26 fl/fl, expressing a tamoxifen-inducible Cre recombinase under the control of the Rosa26 (R26) promoter and carrying two LoxP (fl) sites: one at the 5'end of the Fth promoter and another the 3' end of the first Fth exon. Tamoxifen administration induces global deletion of Fth in adult FthR26Δ/Δ mice, testing whether FTH is required for maintenance of organismal homeostasis. RESULTS Under standard nutritional Fe supply, Fth deletion in adult FthR26Δ/Δ mice led to a profound deregulation of organismal Fe metabolism, oxidative stress, inflammation, and multi-organ damage, culminating in death. Unexpectedly, Fth deletion was also associated with a profound atrophy of white and brown adipose tissue as well as with collapse of energy expenditure and thermogenesis. This was attributed mechanistically to mitochondrial dysfunction, as assessed in the liver and in adipose tissue. CONCLUSION The FTH component of ferritin acts as a master regulator of organismal Fe homeostasis, coupling nutritional Fe supply to organismal redox homeostasis, energy expenditure and thermoregulation.
Collapse
|
44
|
Greene CJ, Sharma NJ, Fiorica PN, Forrester E, Smith GJ, Gross KW, Kauffman EC. Suppressive effects of iron chelation in clear cell renal cell carcinoma and their dependency on VHL inactivation. Free Radic Biol Med 2019; 133:295-309. [PMID: 30553971 PMCID: PMC10038186 DOI: 10.1016/j.freeradbiomed.2018.12.013] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/20/2018] [Revised: 12/10/2018] [Accepted: 12/12/2018] [Indexed: 02/07/2023]
Abstract
Increasing data implicate iron accumulation in tumorigenesis of the kidney, particularly the clear cell renal cell carcinoma (ccRCC) subtype. The von Hippel Lindau (VHL)/hypoxia inducible factor-α (HIF-α) axis is uniquely dysregulated in ccRCC and is a major regulator and regulatory target of iron metabolism, yet the role of iron in ccRCC tumorigenesis and its potential interplay with VHL inactivation remains unclear. We investigated whether ccRCC iron accumulation occurs due to increased cell dependency on iron for growth and survival as a result of VHL inactivation. Free iron levels were compared between four VHL-mutant ccRCC cell lines (786-0, A704, 769-P, RCC4) and two benign renal tubule epithelial cell lines (RPTEC, HRCEp) using the Phen Green SK fluorescent iron stain. Intracellular iron deprivation was achieved using two clinical iron chelator drugs, deferasirox (DFX) and deferoxamine (DFO), and chelator effects were measured on cell line growth, cell cycle phase, apoptosis, HIF-1α and HIF-2α protein levels and HIF-α transcriptional activity based on expression of target genes CA9, OCT4/POU5F1 and PDGFβ/PDGFB. Similar assays were performed in VHL-mutant ccRCC cells with and without ectopic wild-type VHL expression. Baseline free iron levels were significantly higher in ccRCC cell lines than benign renal cell lines. DFX depleted cellular free iron more rapidly than DFO and led to greater growth suppression of ccRCC cell lines (>90% at ~30-150 µM) than benign renal cell lines (~10-50% at up to 250 µM). Similar growth responses were observed using DFO, with the exception that a prolonged treatment duration was necessary to deplete cellular iron adequately for differential growth suppression of the less susceptible A704 ccRCC cell line relative to benign renal cell lines. Apoptosis and G1-phase cell cycle arrest were identified as potential mechanisms of chelator growth suppression based on their induction in ccRCC cell lines but not benign renal cell lines. Iron chelation in ccRCC cells but not benign renal cells suppressed HIF-1α and HIF-2α protein levels and transcriptional activity, and the degree and timing of HIF-2α suppression correlated with the onset of apoptosis. Restoration of wild-type VHL function in ccRCC cells was sufficient to prevent chelator-induced apoptosis and G1 cell cycle arrest, indicating that ccRCC susceptibility to iron deprivation is VHL inactivation-dependent. In conclusion, ccRCC cells are characterized by high free iron levels and a cancer-specific dependency on iron for HIF-α overexpression, cell cycle progression and apoptotic escape. This iron dependency is introduced by VHL inactivation, revealing a novel interplay between VHL/HIF-α dysregulation and ccRCC iron metabolism. Future study is warranted to determine if iron deprivation using chelator drugs provides an effective therapeutic strategy for targeting HIF-2α and suppressing tumor progression in ccRCC patients.
Collapse
Affiliation(s)
- Christopher J Greene
- Department of Urology, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, United States
| | - Nitika J Sharma
- Department of Urology, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, United States
| | - Peter N Fiorica
- Department of Urology, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, United States
| | - Emily Forrester
- Department of Urology, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, United States
| | - Gary J Smith
- Department of Urology, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, United States
| | - Kenneth W Gross
- Department of Molecular and Cellular Biology, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, United States
| | - Eric C Kauffman
- Department of Urology, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, United States; Department of Cancer Genetics, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, United States; Jacobs School of Medicine and Biomedical Sciences, State University of New York at Buffalo, Buffalo, NY 14214, United States.
| |
Collapse
|
45
|
Kober KM, Olshen A, Conley YP, Schumacher M, Topp K, Smoot B, Mazor M, Chesney M, Hammer M, Paul SM, Levine JD, Miaskowski C. Expression of mitochondrial dysfunction-related genes and pathways in paclitaxel-induced peripheral neuropathy in breast cancer survivors. Mol Pain 2018; 14:1744806918816462. [PMID: 30426838 PMCID: PMC6293373 DOI: 10.1177/1744806918816462] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Background Paclitaxel is one of the most commonly used drugs to treat breast cancer. Its
major dose-limiting toxicity is paclitaxel-induced peripheral neuropathy
(PIPN). PIPN persists into survivorship and has a negative impact on
patient’s mood, functional status, and quality of life. No interventions are
available to treat PIPN. A critical barrier to the development of
efficacious interventions is the lack of understanding of the mechanisms
that underlie PIPN. Mitochondrial dysfunction has been evaluated in
preclinical studies as a hypothesized mechanism for PIPN, but clinical data
to support this hypothesis are limited. The purpose of this pilot study was
to evaluate for differential gene expression and perturbed pathways between
breast cancer survivors with and without PIPN. Methods Gene expression in peripheral blood was assayed using RNA-seq. Differentially
expressed genes (DEG) and pathways associated with mitochondrial dysfunction
were identified between survivors who received paclitaxel and did (n = 25)
and did not (n = 25) develop PIPN. Results Breast cancer survivors with PIPN were significantly older; more likely to be
unemployed; reported lower alcohol use; had a higher body mass index and
poorer functional status; and had a higher number of lower extremity sites
with loss of light touch, cold, and pain sensations and higher vibration
thresholds. No between-group differences were found in the cumulative dose
of paclitaxel received or in the percentage of patients who had a dose
reduction or delay due to PIPN. Five DEGs and nine perturbed pathways were
associated with mitochondrial dysfunction related to oxidative stress, iron
homeostasis, mitochondrial fission, apoptosis, and autophagy. Conclusions This study is the first to provide molecular evidence that a number of
mitochondrial dysfunction mechanisms identified in preclinical models of
various types of neuropathic pain including chemotherapy-induced peripheral
neuropathy are found in breast cancer survivors with persistent PIPN and
suggest genes for validation and as potential therapeutic targets.
Collapse
Affiliation(s)
- Kord M Kober
- 1 School of Nursing, University of California, San Francisco, San Francisco, CA, USA
| | - Adam Olshen
- 2 School of Medicine, University of California, San Francisco, San Francisco, CA, USA
| | - Yvettte P Conley
- 3 School of Nursing, University of Pittsburgh, Pittsburgh, PA, USA
| | - Mark Schumacher
- 2 School of Medicine, University of California, San Francisco, San Francisco, CA, USA
| | - Kimberly Topp
- 2 School of Medicine, University of California, San Francisco, San Francisco, CA, USA
| | - Betty Smoot
- 2 School of Medicine, University of California, San Francisco, San Francisco, CA, USA
| | - Melissa Mazor
- 1 School of Nursing, University of California, San Francisco, San Francisco, CA, USA
| | - Margaret Chesney
- 2 School of Medicine, University of California, San Francisco, San Francisco, CA, USA
| | - Marilyn Hammer
- 4 Department of Nursing, Mount Sinai Medical Center, New York, NY, USA
| | - Steven M Paul
- 1 School of Nursing, University of California, San Francisco, San Francisco, CA, USA
| | - Jon D Levine
- 2 School of Medicine, University of California, San Francisco, San Francisco, CA, USA
| | - Christine Miaskowski
- 1 School of Nursing, University of California, San Francisco, San Francisco, CA, USA
| |
Collapse
|
46
|
Haschka D, Volani C, Stefani A, Tymoszuk P, Mitterling T, Holzknecht E, Heidbreder A, Coassin S, Sumbalova Z, Seifert M, Dichtl S, Theurl I, Gnaiger E, Kronenberg F, Frauscher B, Högl B, Weiss G. Association of mitochondrial iron deficiency and dysfunction with idiopathic restless legs syndrome. Mov Disord 2018; 34:114-123. [DOI: 10.1002/mds.27482] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2018] [Revised: 07/11/2018] [Accepted: 07/16/2018] [Indexed: 12/11/2022] Open
Affiliation(s)
- David Haschka
- Department of Internal Medicine II Medical University of Innsbruck Innsbruck Austria
| | - Chiara Volani
- Department of Internal Medicine II Medical University of Innsbruck Innsbruck Austria
| | - Ambra Stefani
- Department of Neurology Medical University of Innsbruck Innsbruck Austria
| | - Piotr Tymoszuk
- Department of Internal Medicine II Medical University of Innsbruck Innsbruck Austria
| | - Thomas Mitterling
- Department of Neurology Medical University of Innsbruck Innsbruck Austria
- Department of Neurology Wagner‐Jauregg Hospital Linz Linz Austria
| | - Evi Holzknecht
- Department of Neurology Medical University of Innsbruck Innsbruck Austria
| | - Anna Heidbreder
- Department of Neurology Medical University of Innsbruck Innsbruck Austria
- Department of Neurology, Division of Sleep Medicine and Neuromuscular Disorders University Hospital Muenster Muenster Germany
| | - Stefan Coassin
- Department of Medical Genetics, Division of Genetic Epidemiology, Molecular and Clinical Pharmacology Medical University of Innsbruck Innsbruck Austria
| | - Zuzana Sumbalova
- Pharmacobiochemical Laboratory of the 3rd Department of Internal Medicine, Faculty of Medicine in Bratislava Comenius University Bratislava Slovakia
| | - Markus Seifert
- Department of Internal Medicine II Medical University of Innsbruck Innsbruck Austria
| | - Stefanie Dichtl
- Department of Internal Medicine II Medical University of Innsbruck Innsbruck Austria
| | - Igor Theurl
- Department of Internal Medicine II Medical University of Innsbruck Innsbruck Austria
| | - Erich Gnaiger
- Department of General and Transplant Surgery, D. Swarovski Research Laboratory Medical University of Innsbruck Innsbruck Austria
| | - Florian Kronenberg
- Department of Medical Genetics, Division of Genetic Epidemiology, Molecular and Clinical Pharmacology Medical University of Innsbruck Innsbruck Austria
| | - Birgit Frauscher
- Department of Neurology Medical University of Innsbruck Innsbruck Austria
| | - Birgit Högl
- Department of Neurology Medical University of Innsbruck Innsbruck Austria
| | - Guenter Weiss
- Department of Internal Medicine II Medical University of Innsbruck Innsbruck Austria
- Christian Doppler Laboratory for Iron Metabolism and Anemia Research Medical University of Innsbruck Innsbruck Austria
| |
Collapse
|
47
|
Dziegala M, Josiak K, Kasztura M, Kobak K, von Haehling S, Banasiak W, Anker SD, Ponikowski P, Jankowska E. Iron deficiency as energetic insult to skeletal muscle in chronic diseases. J Cachexia Sarcopenia Muscle 2018; 9:802-815. [PMID: 30178922 PMCID: PMC6204587 DOI: 10.1002/jcsm.12314] [Citation(s) in RCA: 70] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/08/2018] [Revised: 04/05/2018] [Accepted: 04/22/2018] [Indexed: 12/19/2022] Open
Abstract
Specific skeletal myopathy constitutes a common feature of heart failure, chronic obstructive pulmonary disease, and type 2 diabetes mellitus, where it can be characterized by the loss of skeletal muscle oxidative capacity. There is evidence from in vitro and animal studies that iron deficiency affects skeletal muscle functioning mainly in the context of its energetics by limiting oxidative metabolism in favour of glycolysis and by alterations in both carbohydrate and fat catabolic processing. In this review, we depict the possible molecular pathomechanisms of skeletal muscle energetic impairment and postulate iron deficiency as an important factor causally linked to loss of muscle oxidative capacity that contributes to skeletal myopathy seen in patients with heart failure, chronic obstructive pulmonary disease, and type 2 diabetes mellitus.
Collapse
Affiliation(s)
- Magdalena Dziegala
- Laboratory for Applied Research on Cardiovascular System, Department of Heart DiseasesWroclaw Medical University50‐981WroclawPoland
| | - Krystian Josiak
- Centre for Heart DiseasesMilitary Hospital50‐981WroclawPoland
- Department of Heart DiseasesWroclaw Medical University50‐367WroclawPoland
| | - Monika Kasztura
- Laboratory for Applied Research on Cardiovascular System, Department of Heart DiseasesWroclaw Medical University50‐981WroclawPoland
| | - Kamil Kobak
- Laboratory for Applied Research on Cardiovascular System, Department of Heart DiseasesWroclaw Medical University50‐981WroclawPoland
| | - Stephan von Haehling
- Department of Cardiology and PneumologyUniversity Medicine Göttingen (UMG)37075GöttingenGermany
| | | | - Stefan D. Anker
- Department of Cardiology and PneumologyUniversity Medicine Göttingen (UMG)37075GöttingenGermany
- Division of Cardiology and MetabolismCharité Universitätsmedizin10117BerlinGermany
- Department of Cardiology (CVK)Charité Universitätsmedizin10117BerlinGermany
- Berlin‐Brandenburg Center for Regenerative Therapies (BCRT)Charité Universitätsmedizin10117BerlinGermany
- German Centre for Cardiovascular Research (DZHK) partner site BerlinCharité Universitätsmedizin10117BerlinGermany
| | - Piotr Ponikowski
- Centre for Heart DiseasesMilitary Hospital50‐981WroclawPoland
- Department of Heart DiseasesWroclaw Medical University50‐367WroclawPoland
| | - Ewa Jankowska
- Laboratory for Applied Research on Cardiovascular System, Department of Heart DiseasesWroclaw Medical University50‐981WroclawPoland
- Centre for Heart DiseasesMilitary Hospital50‐981WroclawPoland
| |
Collapse
|
48
|
Ruhe J, Waldeyer C, Ojeda F, Altay A, Schnabel RB, Schäfer S, Lackner KJ, Blankenberg S, Zeller T, Karakas M. Intrinsic Iron Release Is Associated with Lower Mortality in Patients with Stable Coronary Artery Disease-First Report on the Prospective Relevance of Intrinsic Iron Release. Biomolecules 2018; 8:biom8030072. [PMID: 30096922 PMCID: PMC6164542 DOI: 10.3390/biom8030072] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2018] [Revised: 08/03/2018] [Accepted: 08/03/2018] [Indexed: 12/20/2022] Open
Abstract
Intrinsic iron release is discussed to have favorable effects in coronary artery disease (CAD). The aim of this study was to evaluate the prognostic relevance of intrinsic iron release in patients with CAD. Intrinsic iron release was based on a definition including hepcidin and soluble transferrin receptor (sTfR). In a cohort of 811 patients with angiographically documented CAD levels of hepcidin and sTfR were measured at baseline. Systemic body iron release was defined as low levels of hepcidin (<24 ng/mL) and high levels of sTfR (≥2 mg/L). A commercially available ELISA (DRG) was used for measurements of serum hepcidin. Serum sTfR was determined by using an automated immunoassay (). Cardiovascular mortality was the main outcome measure. The criteria of intrinsic iron release were fulfilled in 32.6% of all patients. Significantly lower cardiovascular mortality rates were observed in CAD patients with systemic iron release. After adjustment for body mass index, smoking status, hypertension, diabetes, dyslipidemia, sex, and age, the hazard ratio for future cardiovascular death was 0.41. After an additional adjustment for surrogates of the size of myocardial necrosis (troponin I), anemia (hemoglobin), and cardiac function and heart failure severity (N-terminal pro B-type natriuretic peptide), this association did not change (Hazard ratio 0.37 (95% confidence interval 0.14⁻0.99), p = 0.047). In conclusion, significantly lower cardiovascular mortality rates were observed in CAD patients with intrinsic iron release shown during follow-up.
Collapse
Affiliation(s)
- Julia Ruhe
- Department of General and Interventional Cardiology, University Heart Center Hamburg, 20246 Hamburg, Germany.
| | - Christoph Waldeyer
- Department of General and Interventional Cardiology, University Heart Center Hamburg, 20246 Hamburg, Germany.
| | - Francisco Ojeda
- Department of General and Interventional Cardiology, University Heart Center Hamburg, 20246 Hamburg, Germany.
| | - Alev Altay
- Department of General and Interventional Cardiology, University Heart Center Hamburg, 20246 Hamburg, Germany.
| | - Renate B Schnabel
- Department of General and Interventional Cardiology, University Heart Center Hamburg, 20246 Hamburg, Germany.
- German Center for Cardiovascular Research (DZHK), Partner Site Hamburg, Lübeck, Kiel, 20246 Hamburg, Germany.
| | - Sarina Schäfer
- Department of General and Interventional Cardiology, University Heart Center Hamburg, 20246 Hamburg, Germany.
- German Center for Cardiovascular Research (DZHK), Partner Site Hamburg, Lübeck, Kiel, 20246 Hamburg, Germany.
| | - Karl J Lackner
- German Center for Cardiovascular Research (DZHK), Partner Site Rhein-Main, 55131 Mainz, Germany.
- Department of Laboratory Medicine, University Medical Center, Johannes Gutenberg University Mainz, 55131 Mainz, Germany.
| | - Stefan Blankenberg
- Department of General and Interventional Cardiology, University Heart Center Hamburg, 20246 Hamburg, Germany.
- German Center for Cardiovascular Research (DZHK), Partner Site Hamburg, Lübeck, Kiel, 20246 Hamburg, Germany.
| | - Tanja Zeller
- Department of General and Interventional Cardiology, University Heart Center Hamburg, 20246 Hamburg, Germany.
- German Center for Cardiovascular Research (DZHK), Partner Site Hamburg, Lübeck, Kiel, 20246 Hamburg, Germany.
| | - Mahir Karakas
- Department of General and Interventional Cardiology, University Heart Center Hamburg, 20246 Hamburg, Germany.
- German Center for Cardiovascular Research (DZHK), Partner Site Hamburg, Lübeck, Kiel, 20246 Hamburg, Germany.
| |
Collapse
|
49
|
Motonishi S, Tanaka K, Ozawa T. Iron deficiency associates with deterioration in several symptoms independently from hemoglobin level among chronic hemodialysis patients. PLoS One 2018; 13:e0201662. [PMID: 30071093 PMCID: PMC6072073 DOI: 10.1371/journal.pone.0201662] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2018] [Accepted: 07/19/2018] [Indexed: 12/22/2022] Open
Abstract
Background While iron deficiency (ID) is a frequent cause of anemia in hemodialysis patients, the clinical impact of ID without anemic level of hemoglobin remains unclear. As such, this study was designed to clarify the manifestations of ID itself in subjects on hemodialysis. Methods Maintenance hemodialysis patients achieving target hemoglobin levels (≥ 10.0g/dL) under treatment in our clinic were stratified for comparison from three perspectives: ID (transferrin saturation [TSAT] < 20% or ferritin < 100ng/mL) vs non-ID, level of TSAT (< or ≥ 20%), and level of serum ferritin concentration (< or ≥ 100ng/mL). The severity of frequent symptoms was determined by a self-rating symptom score questionnaire, and the rate of those with severe manifestations was calculated for each symptom. Significant difference was examined between groups; univariate and adjusted multivariate odds ratios and 95% confidence intervals were obtained by logistic regression. Results Among 154 subjects selected for analysis, the ratio of severe arthralgia and fatigue was significantly higher in the ID group (n = 94) compared to the non-ID group (n = 60), in both univariate and adjusted multivariate analyses. Moreover, in multivariate analysis, low TSAT was significantly associated with exacerbation of pain during vascular access puncture and intradialytic leg cramps, while low serum ferritin concentration was related to significant increase in severe arthralgia, fatigue, intradialytic headache and leg cramps. Conclusions ID was identified as a risk factor regarding severity of several symptoms even without low hemoglobin level among chronic hemodialysis patients, and supplementation of iron was considered efficacious for improving critical symptoms affecting those undergoing maintenance dialysis.
Collapse
|
50
|
Varga E, Pandur E, Abrahám H, Horváth A, Ács P, Komoly S, Miseta A, Sipos K. Cuprizone Administration Alters the Iron Metabolism in the Mouse Model of Multiple Sclerosis. Cell Mol Neurobiol 2018; 38:1081-1097. [PMID: 29464444 DOI: 10.1007/s10571-018-0578-5] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2017] [Accepted: 02/13/2018] [Indexed: 01/01/2023]
Abstract
Cuprizone (CZ) is a widely used copper chelating agent to develop non-autoimmune animal model of multiple sclerosis, characterized by demyelination of the corpus callosum (CC) and other brain regions. The exact mechanisms of CZ action are still arguable, but it seems that the only affected cells are the mature oligodendrocytes, possibly via metabolic disturbances caused by copper deficiency. During the pathogenesis of multiple sclerosis, high amount of deposited iron can be found throughout the demyelinated areas of the brain in the form of extracellular iron deposits and intracellularly accumulated iron in microglia. In the present study, we used the accepted experimental model of 0.2% CZ-containing diet with standard iron concentration to induce demyelination in the brain of C57BL/6 mice. Our aim was to examine the changes of iron homeostasis in the CC and as a part of the systemic iron regulation, in the liver. Our data showed that CZ treatment changed the iron metabolism of both tissues; however, it had more impact on the liver. Besides the alterations in the expressions of iron storage and import proteins, we detected reduced serum iron concentration and iron stores in the liver, together with elevated hepcidin levels and feasible disturbances in the Fe-S cluster biosynthesis. Our results revealed that the CZ-containing diet influences the systemic iron metabolism in mice, particularly the iron homeostasis of the liver. This inadequate systemic iron regulation may affect the iron homeostasis of the brain, eventually indicating a relationship among CZ treatment, iron metabolism, and neurodegeneration.
Collapse
Affiliation(s)
- E Varga
- Department of Pharmaceutical Biology, Faculty of Pharmacy, University of Pécs, Rókus str. 2., Pécs, 7624, Hungary
| | - E Pandur
- Department of Pharmaceutical Biology, Faculty of Pharmacy, University of Pécs, Rókus str. 2., Pécs, 7624, Hungary
| | - H Abrahám
- Department of Medical Biology and Central Electron Microscope Laboratory, Medical School, University of Pécs, Szigeti str. 12., Pécs, 7624, Hungary
| | - A Horváth
- Department of Neurosurgery, Medical School, University of Pécs, Rét str. 2., Pécs, 7623, Hungary
| | - P Ács
- Department of Neurology, Medical School, University of Pécs, Rét str. 2., Pécs, 7623, Hungary
| | - S Komoly
- Department of Neurology, Medical School, University of Pécs, Rét str. 2., Pécs, 7623, Hungary
| | - A Miseta
- Department of Laboratory Medicine, Medical School, University of Pécs, Ifjúság str. 13., Pécs, 7624, Hungary
| | - K Sipos
- Department of Pharmaceutical Biology, Faculty of Pharmacy, University of Pécs, Rókus str. 2., Pécs, 7624, Hungary.
- , Szigeti str. 12., Pécs, 7624, Hungary.
| |
Collapse
|