51
|
Neonatal Lead (Pb) Exposure and DNA Methylation Profiles in Dried Bloodspots. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2020; 17:ijerph17186775. [PMID: 32957503 PMCID: PMC7559513 DOI: 10.3390/ijerph17186775] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Revised: 09/12/2020] [Accepted: 09/14/2020] [Indexed: 12/21/2022]
Abstract
Lead (Pb) exposure remains a major concern in the United States (US) and around the world, even following the removal of Pb from gasoline and other products. Environmental Pb exposures from aging infrastructure and housing stock are of particular concern to pregnant women, children, and other vulnerable populations. Exposures during sensitive periods of development are known to influence epigenetic modifications which are thought to be one mechanism of the Developmental Origins of Health and Disease (DOHaD) paradigm. To gain insights into early life Pb exposure-induced health risks, we leveraged neonatal dried bloodspots in a cohort of children from Michigan, US to examine associations between blood Pb levels and concomitant DNA methylation profiles (n = 96). DNA methylation analysis was conducted via the Infinium MethylationEPIC array and Pb levels were assessed via high resolution inductively coupled plasma mass spectrometry (HR-ICP-MS). While at-birth Pb exposure levels were relatively low (average 0.78 µg/dL, maximum of 5.27 ug/dL), we identified associations between DNA methylation and Pb at 33 CpG sites, with the majority (82%) exhibiting reduced methylation with increasing Pb exposure (q < 0.2). Biological pathways related to development and neurological function were enriched amongst top differentially methylated genes by p-value. In addition to increases/decreases in methylation, we also demonstrate that Pb exposure is related to increased variability in DNA methylation at 16 CpG sites. More work is needed to assess the accuracy and precision of metals assessment using bloodspots, but this study highlights the utility of this unique resource to enhance environmental epigenetics research around the world.
Collapse
|
52
|
Huang M, Yang L, Peng X, Wei S, Fan Q, Yang S, Li X, Li B, Jin H, Wu B, Liu J, Li H. Autonomous glucose metabolic reprogramming of tumour cells under hypoxia: opportunities for targeted therapy. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2020; 39:185. [PMID: 32928258 PMCID: PMC7491117 DOI: 10.1186/s13046-020-01698-5] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/16/2020] [Accepted: 09/03/2020] [Indexed: 12/11/2022]
Abstract
Molecular oxygen (O2) is a universal electron acceptor that is eventually synthesized into ATP in the mitochondrial respiratory chain of all metazoans. Therefore, hypoxia biology has become an organizational principle of cell evolution, metabolism and pathology. Hypoxia-inducible factor (HIF) mediates tumour cells to produce a series of glucose metabolism adaptations including the regulation of glucose catabolism, glycogen metabolism and the biological oxidation of glucose to hypoxia. Since HIF can regulate the energy metabolism of cancer cells and promote the survival of cancer cells, targeting HIF or HIF mediated metabolic enzymes may become one of the potential treatment methods for cancer. In this review, we summarize the established and recently discovered autonomous molecular mechanisms that can induce cell reprogramming of hypoxic glucose metabolism in tumors and explore opportunities for targeted therapy.
Collapse
Affiliation(s)
- Mingyao Huang
- Department of General Surgery, the Fourth Affiliated Hospital, China Medical University, Shenyang, 110032, China
| | - Liang Yang
- Department of General Surgery, the Fourth Affiliated Hospital, China Medical University, Shenyang, 110032, China
| | - Xueqiang Peng
- Department of General Surgery, the Fourth Affiliated Hospital, China Medical University, Shenyang, 110032, China
| | - Shibo Wei
- Department of General Surgery, the Fourth Affiliated Hospital, China Medical University, Shenyang, 110032, China
| | - Qing Fan
- Department of General Surgery, the Fourth Affiliated Hospital, China Medical University, Shenyang, 110032, China
| | - Shuo Yang
- Department of General Surgery, the Fourth Affiliated Hospital, China Medical University, Shenyang, 110032, China
| | - Xinyu Li
- Department of General Surgery, the Fourth Affiliated Hospital, China Medical University, Shenyang, 110032, China
| | - Bowen Li
- Department of General Surgery, the Fourth Affiliated Hospital, China Medical University, Shenyang, 110032, China
| | - Hongyuan Jin
- Department of General Surgery, the Fourth Affiliated Hospital, China Medical University, Shenyang, 110032, China
| | - Bo Wu
- Department of General Surgery, the Fourth Affiliated Hospital, China Medical University, Shenyang, 110032, China
| | - Jingang Liu
- Department of General Surgery, the Fourth Affiliated Hospital, China Medical University, Shenyang, 110032, China
| | - Hangyu Li
- Department of General Surgery, the Fourth Affiliated Hospital, China Medical University, Shenyang, 110032, China.
| |
Collapse
|
53
|
Wu H, Wei H, Zhang D, Sehgal SA, Zhang D, Wang X, Qin Y, Liu L, Chen Q. Defective mitochondrial ISCs biogenesis switches on IRP1 to fine tune selective mitophagy. Redox Biol 2020; 36:101661. [PMID: 32795936 PMCID: PMC7426581 DOI: 10.1016/j.redox.2020.101661] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2020] [Revised: 07/21/2020] [Accepted: 07/24/2020] [Indexed: 12/26/2022] Open
Abstract
Both iron metabolism and mitophagy, a selective mitochondrial degradation process via autolysosomal pathway, are fundamental for the cellular well-being. Mitochondria are the major site for iron metabolism, especially the biogenesis of iron-sulfur clusters (ISCs) via the mitochondria-localized ISCs assembly machinery. Here we report that mitochondrial ISCs biogenesis is coupled with receptor-mediated mitophagy in mammalian cells. Perturbation of mitochondrial ISCs biogenesis, either by depleting iron with the iron chelator or by knocking down the core components of the mitochondrial ISCs assembly machinery, triggers FUNDC1-dependent mitophagy. IRP1, one of the cellular iron sensors to maintain iron homeostasis, is crucial for iron stresses induced mitophagy. Knockdown of IRP1 disturbed iron stresses induced mitophagy. Furthermore, IRP1 could bind to a newly characterized IRE in the 5’ untranslated region of the Bcl-xL mRNA and suppress its translation. Bcl-xL is an intrinsic inhibitory protein of the mitochondrial phosphatase PGAM5, which catalyzes the dephosphorylation of FUNDC1 for mitophagy activation. Alterations of the IRP1/Bcl-xL axis navigate iron stresses induced mitophagy. We conclude that ISCs serve as physiological signals for mitophagy activation, thus coupling mitophagy with iron metabolism. Perturbation of ISCs biogenesis triggers FUNDC1 dependent mitophagy. IRP1 targets a newly characterized IRE in Bcl-xL mRNA to suppress its translation. IRP1/Bcl-xL axis navigates iron stresses induced mitophagy and dominates mitochondrial redox response.
Collapse
Affiliation(s)
- Hao Wu
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, China; State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China.
| | - Huifang Wei
- China-US (Henan) Hormel Cancer Institute, Zhengzhou, 450003, China; Department of Pathophysiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450001, Henan, China
| | - Di Zhang
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China; University of Chinese Academy of Sciences, Beijing, 100101, China
| | - Sheikh Arslan Sehgal
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China; University of Chinese Academy of Sciences, Beijing, 100101, China; COMSATS University, Islamabad, Sahiwal Campus, Pakistan
| | - Dejiu Zhang
- Key Laboratory of RNA Biology, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China
| | - Xiaohui Wang
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China
| | - Yan Qin
- Key Laboratory of RNA Biology, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China
| | - Lei Liu
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China.
| | - Quan Chen
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China; University of Chinese Academy of Sciences, Beijing, 100101, China; Tianjin Key Laboratory of Protein Science, College of Life Sciences, Nankai University, Tianjin, 300071, China.
| |
Collapse
|
54
|
Seguin A, Jia X, Earl AM, Li L, Wallace J, Qiu A, Bradley T, Shrestha R, Troadec MB, Hockin M, Titen S, Warner DE, Dowdle PT, Wohlfahrt ME, Hillas E, Firpo MA, Phillips JD, Kaplan J, Paw BH, Barasch J, Ward DM. The mitochondrial metal transporters mitoferrin1 and mitoferrin2 are required for liver regeneration and cell proliferation in mice. J Biol Chem 2020; 295:11002-11020. [PMID: 32518166 DOI: 10.1074/jbc.ra120.013229] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2020] [Revised: 06/04/2020] [Indexed: 01/31/2023] Open
Abstract
Mitochondrial iron import is essential for iron-sulfur cluster formation and heme biosynthesis. Two nuclear-encoded vertebrate mitochondrial high-affinity iron importers, mitoferrin1 (Mfrn1) and Mfrn2, have been identified in mammals. In mice, the gene encoding Mfrn1, solute carrier family 25 member 37 (Slc25a37), is highly expressed in sites of erythropoiesis, and whole-body Slc25a37 deletion leads to lethality. Here, we report that mice with a deletion of Slc25a28 (encoding Mfrn2) are born at expected Mendelian ratios, but show decreased male fertility due to reduced sperm numbers and sperm motility. Mfrn2 -/- mice placed on a low-iron diet exhibited reduced mitochondrial manganese, cobalt, and zinc levels, but not reduced iron. Hepatocyte-specific loss of Slc25a37 (encoding Mfrn1) in Mfrn2 -/- mice did not affect animal viability, but resulted in a 40% reduction in mitochondrial iron and reduced levels of oxidative phosphorylation proteins. Placing animals on a low-iron diet exaggerated the reduction in mitochondrial iron observed in liver-specific Mfrn1/2-knockout animals. Mfrn1 -/-/Mfrn2 -/- bone marrow-derived macrophages or skin fibroblasts in vitro were unable to proliferate, and overexpression of Mfrn1-GFP or Mfrn2-GFP prevented this proliferation defect. Loss of both mitoferrins in hepatocytes dramatically reduced regeneration in the adult mouse liver, further supporting the notion that both mitoferrins transport iron and that their absence limits proliferative capacity of mammalian cells. We conclude that Mfrn1 and Mfrn2 contribute to mitochondrial iron homeostasis and are required for high-affinity iron import during active proliferation of mammalian cells.
Collapse
Affiliation(s)
- Alexandra Seguin
- Department of Pathology, University of Utah School of Medicine, Salt Lake City, Utah, USA
| | - Xuan Jia
- Department of Pathology, University of Utah School of Medicine, Salt Lake City, Utah, USA
| | - Aubree M Earl
- Department of Pathology, University of Utah School of Medicine, Salt Lake City, Utah, USA
| | - Liangtao Li
- Department of Pathology, University of Utah School of Medicine, Salt Lake City, Utah, USA
| | - Jared Wallace
- Department of Pathology, University of Utah School of Medicine, Salt Lake City, Utah, USA
| | - Andong Qiu
- Columbia University, New York, New York, USA
| | - Thomas Bradley
- Department of Pathology, University of Utah School of Medicine, Salt Lake City, Utah, USA
| | - Rishna Shrestha
- Department of Pathology, University of Utah School of Medicine, Salt Lake City, Utah, USA
| | - Marie-Bérengère Troadec
- University Brest, Inserm, EFS, UMR 1078, GGB, F-29200, Brest, France.,CHRU Brest, Service of Genetics, Laboratory of Chromosome Genetics, Brest, France
| | - Matt Hockin
- Department of Human Genetics, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
| | - Simon Titen
- Department of Human Genetics, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
| | - Dave E Warner
- Department of Pathology, University of Utah School of Medicine, Salt Lake City, Utah, USA
| | - P Tom Dowdle
- Department of Pathology, University of Utah School of Medicine, Salt Lake City, Utah, USA
| | - Martin E Wohlfahrt
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
| | - Elaine Hillas
- Department of General Surgery, University of Utah School of Medicine, Salt Lake City, Utah, USA
| | - Matthew A Firpo
- Department of General Surgery, University of Utah School of Medicine, Salt Lake City, Utah, USA
| | - John D Phillips
- Department of Medicine, University of Utah School of Medicine, Salt Lake City, Utah
| | - Jerry Kaplan
- Department of Pathology, University of Utah School of Medicine, Salt Lake City, Utah, USA
| | - Barry H Paw
- Harvard Medical School, Children's Hospital, Boston, Massachusetts, USA
| | | | - Diane M Ward
- Department of Pathology, University of Utah School of Medicine, Salt Lake City, Utah, USA
| |
Collapse
|
55
|
Chiabrando D, Bertino F, Tolosano E. Hereditary Ataxia: A Focus on Heme Metabolism and Fe-S Cluster Biogenesis. Int J Mol Sci 2020; 21:ijms21113760. [PMID: 32466579 PMCID: PMC7312568 DOI: 10.3390/ijms21113760] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Revised: 05/21/2020] [Accepted: 05/22/2020] [Indexed: 02/07/2023] Open
Abstract
Heme and Fe-S clusters regulate a plethora of essential biological processes ranging from cellular respiration and cell metabolism to the maintenance of genome integrity. Mutations in genes involved in heme metabolism and Fe-S cluster biogenesis cause different forms of ataxia, like posterior column ataxia and retinitis pigmentosa (PCARP), Friedreich's ataxia (FRDA) and X-linked sideroblastic anemia with ataxia (XLSA/A). Despite great efforts in the elucidation of the molecular pathogenesis of these disorders several important questions still remain to be addressed. Starting with an overview of the biology of heme metabolism and Fe-S cluster biogenesis, the review discusses recent progress in the understanding of the molecular pathogenesis of PCARP, FRDA and XLSA/A, and highlights future line of research in the field. A better comprehension of the mechanisms leading to the degeneration of neural circuity responsible for balance and coordinated movement will be crucial for the therapeutic management of these patients.
Collapse
|
56
|
Glia maturation factor-γ regulates murine macrophage iron metabolism and M2 polarization through mitochondrial ROS. Blood Adv 2020; 3:1211-1225. [PMID: 30971398 DOI: 10.1182/bloodadvances.2018026070] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2018] [Accepted: 02/16/2019] [Indexed: 12/19/2022] Open
Abstract
In macrophages, cellular iron metabolism status is tightly integrated with macrophage phenotype and associated with mitochondrial function. However, how molecular events regulate mitochondrial activity to integrate regulation of iron metabolism and macrophage phenotype remains unclear. Here, we explored the important role of the actin-regulatory protein glia maturation factor-γ (GMFG) in the regulation of cellular iron metabolism and macrophage phenotype. We found that GMFG was downregulated in murine macrophages by exposure to iron and hydrogen peroxide. GMFG knockdown altered the expression of iron metabolism proteins and increased iron levels in murine macrophages and concomitantly promoted their polarization toward an anti-inflammatory M2 phenotype. GMFG-knockdown macrophages exhibited moderately increased levels of mitochondrial reactive oxygen species (mtROS), which were accompanied by decreased expression of some mitochondrial respiration chain components, including the iron-sulfur cluster assembly scaffold protein ISCU as well as the antioxidant enzymes SOD1 and SOD2. Importantly, treatment of GMFG-knockdown macrophages with the antioxidant N-acetylcysteine reversed the altered expression of iron metabolism proteins and significantly inhibited the enhanced gene expression of M2 macrophage markers, suggesting that mtROS is mechanistically linked to cellular iron metabolism and macrophage phenotype. Finally, GMFG interacted with the mitochondrial membrane ATPase ATAD3A, suggesting that GMFG knockdown-induced mtROS production might be attributed to alteration of mitochondrial function in macrophages. Our findings suggest that GMFG is an important regulator in cellular iron metabolism and macrophage phenotype and could be a novel therapeutic target for modulating macrophage function in immune and metabolic disorders.
Collapse
|
57
|
MiR-210-3p Inhibits Proliferation and Migration of C6 Cells by Targeting Iscu. Neurochem Res 2020; 45:1813-1824. [PMID: 32388695 DOI: 10.1007/s11064-020-03043-w] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2020] [Revised: 04/27/2020] [Accepted: 04/29/2020] [Indexed: 12/20/2022]
Abstract
Glioma is the most common primary brain tumor and the most malignant type of glioma is glioblastoma with the character of high mortality, high recurrence rate and poor prognosis. MicroRNAs act as an important component in glioma development and thus may be a potential target for the treatment of glioma. There were some researches indicated that miR-210-3p played a role in glioma development, but if it can inhibit glioma growth, as well as the underlying mechanism, is still uncertain. In the present study, we investigated the effects of miR-210-3p and its potential target gene Iscu on glioma (C6) cells proliferation and migration in vitro as well as the influence of miR-210-3p on glioma growth in vivo. The results showed that miR-210-3p inhibited the proliferation and migration of C6 cells by regulating the expression of its target gene Iscu in vitro. We also demonstrated that glioma growth was suppressed in immunodeficient mice when they were implanted with C6 cells overexpressing miR-210-3p. Our data indicated that miR-210-3p played an important role in the prevention of glioma growth by targeting Iscu and so miR-210-3p/Iscu axis might be a potential target for the treatment of glioma.
Collapse
|
58
|
Abedi M, Rahgozar S, Esmaeili A. Iron protects childhood acute lymphoblastic leukemia cells from methotrexate cytotoxicity. Cancer Med 2020; 9:3537-3550. [PMID: 32176452 PMCID: PMC7221302 DOI: 10.1002/cam4.2982] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2019] [Revised: 02/19/2020] [Accepted: 02/22/2020] [Indexed: 12/14/2022] Open
Abstract
Drug resistance is a fundamental clinical concern in pediatric acute lymphoblastic leukemia (pALL), and methotrexate (MTX) is an essential chemotherapy drug administered for the treatment. In the current study, the effect of iron in response to methotrexate and its underlying mechanisms were investigated in pALL cells. CCRF-CEM and Nalm6 cell lines were selected as T and B-ALL subtypes. Cells were pretreated with ferric ammonium citrate, exposed to the IC50 concentration of MTX and cell viability was assessed using MTT, colony formation, and flow cytometry assays. Iron-loaded cells were strongly resistant to MTX cytotoxicity. The inhibitory effect of N-acetyl cysteine to reverse the acquired MTX resistance was greater than that of the iron chelator, deferasirox, highlighting the importance of iron-mediated ROS in MTX resistance. Subsequently, the upregulation of BCL2, SOD2, NRF2, and MRP1 was confirmed using quantitative RT-PCR. Moreover, a positive correlation was demonstrated between the MRP1 expression levels and bone marrow iron storage in pALL patients. Further supporting our findings were the hematoxylin and eosin-stained histological sections showing that iron-treated nude mice xenografts demonstrated significantly more liver damage than those unexposed to iron. Overall, iron is introduced as a player with a novel role contributing to methotrexate resistance in pALL. Our findings suggest that the patients' bone marrow iron stores are necessary to be assessed during the chemotherapy, and transfusions should be carefully administrated.
Collapse
Affiliation(s)
- Marjan Abedi
- Department of Cell and Molecular biology & Microbiology, Faculty of Biological Science and Technology, University of Isfahan, Isfahan, Iran
| | - Soheila Rahgozar
- Department of Cell and Molecular biology & Microbiology, Faculty of Biological Science and Technology, University of Isfahan, Isfahan, Iran
| | - Abolghasem Esmaeili
- Department of Cell and Molecular biology & Microbiology, Faculty of Biological Science and Technology, University of Isfahan, Isfahan, Iran
| |
Collapse
|
59
|
Kan C, Song F, Shao X, Wu L, Zhu J. Fe(III) induced fluorescent probe based on triamine and rhodamine derivatives and its applications in biological imaging. J Photochem Photobiol A Chem 2020. [DOI: 10.1016/j.jphotochem.2019.112306] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
|
60
|
Management of Iron Overload in Resource Poor Nations: A Systematic Review of Phlebotomy and Natural Chelators. J Toxicol 2020; 2020:4084538. [PMID: 32399029 PMCID: PMC7204175 DOI: 10.1155/2020/4084538] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2019] [Revised: 09/13/2019] [Accepted: 09/20/2019] [Indexed: 12/29/2022] Open
Abstract
Iron is an essential element and the most abundant trace metal in the body involved in oxygen transport and oxygen sensing, electron transfer, energy metabolism, and DNA synthesis. Excess labile and unchelated iron can catalyze the formation of tissue-damaging radicals and induce oxidative stress. English abstracts were identified in PubMed and Google Scholar using multiple and various search terms based on defined inclusion and exclusion criteria. Full-length articles were selected for systematic review, and secondary and tertiary references were developed. Although bloodletting or phlebotomy remains the gold standard in the management of iron overload, this systematic review is an updated account of the pitfalls of phlebotomy and classical synthetic chelators with scientific justification for the use of natural iron chelators of dietary origin in resource-poor nations.
Collapse
|
61
|
Birjiniuk A, Glinton KE, Villafranco N, Boyer S, Laufman J, Mizerik E, Scott D, Elsea SH, Galambos C, Varghese NP, Scaglia F. Multiple mitochondrial dysfunctions syndrome 1: An unusual cause of developmental pulmonary hypertension. Am J Med Genet A 2020; 182:755-761. [PMID: 31970900 DOI: 10.1002/ajmg.a.61491] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2019] [Revised: 12/30/2019] [Accepted: 01/05/2020] [Indexed: 11/06/2022]
Abstract
Pulmonary hypertension (pHTN) is a severe, life-threatening disease, which can be idiopathic or associated with an underlying syndrome or genetic diagnosis. Here we discuss a patient who presented with severe pHTN and was later found to be compound heterozygous for pathogenic variants in the NFU1 gene causing multiple mitochondrial dysfunctions syndrome 1 (MMDS1). Review of autopsy slides from an older sibling revealed the same diagnosis along with pulmonary findings consistent with a developmental lung disorder. In particular, these postmortem, autopsy findings have not been described previously in humans with this mitochondrial syndrome and suggest a possible developmental basis for the severe pHTN seen in this disease. Given the rarity of patients reported with MMDS1, we review the current state of knowledge of this disease and our novel management strategies for pHTN and MMDS1-associated complications in this population.
Collapse
Affiliation(s)
- Alona Birjiniuk
- Department of Pediatrics, Baylor College of Medicine, Houston, Texas
| | - Kevin E Glinton
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas
| | - Natalie Villafranco
- Department of Pulmonary Medicine, Texas Children's Hospital, Houston, Texas.,Department of Pediatrics, Section of Pediatric Pulmonology, Baylor College of Medicine, Houston, Texas
| | - Suzanne Boyer
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas
| | - Jason Laufman
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas
| | - Elizabeth Mizerik
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas
| | - Daryl Scott
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas
| | - Sarah H Elsea
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas
| | - Csaba Galambos
- Department of Pathology and Laboratory Medicine, University of Colorado School of Medicine and Children's Hospital Colorado, Aurora, Colorado.,Pediatric Heart Lung Center, University of Colorado School of Medicine and Children's Hospital Colorado, Aurora, Colorado
| | - Nidhy P Varghese
- Department of Pulmonary Medicine, Texas Children's Hospital, Houston, Texas.,Department of Pediatrics, Section of Pediatric Pulmonology, Baylor College of Medicine, Houston, Texas
| | - Fernando Scaglia
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas.,BCM-CUHK Center of Medical Genetics, Prince of Wales Hospital, Shatin, Hong Kong.,Texas Children's Hospital, Houston, Texas
| |
Collapse
|
62
|
Huang L, Li L, Luo X, Huang S, Hou Q, Ge X, Lv Y, Mo Z, Yang X. The association between serum iron status and risk of asthma: a 2-sample Mendelian randomization study in descendants of Europeans. Am J Clin Nutr 2019; 110:959-968. [PMID: 31380560 DOI: 10.1093/ajcn/nqz162] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2019] [Accepted: 06/27/2019] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND Observational studies present conflicting results about a possible association of iron status with asthma risk, pointing to potential modifiable targets for prevention. OBJECTIVE The aim of this study was to use Mendelian randomization (MR) to estimate associations between iron status and asthma risk. METHODS We used the Genetics of Iron Status consortium to identify genetic variants that could be used as instrumental variables for the effect of systemic iron status. The following sets of instruments were used: a conservative set (instruments restricted to variants with concordant relations to 4 iron status biomarkers) and a liberal set (instruments selected using variants associated with at least 1 of 4 iron status biomarkers). Associations of these genetic variants with asthma risk were estimated in data from the Trans-National Asthma Genetics Consortium (TAGC) and the GABRIEL consortium (A Multidisciplinary Study to Identify the Genetic and Environmental Causes of Asthma in the European Community). Data on the association of genetic variants with iron status and with asthma were combined to assess the influence of iron status on asthma risk. RESULTS In the conservative approach, the MR OR of asthma was 1.00 (95% CI: 0.91, 1.10) per SD increase in iron, 0.96 (95% CI: 0.78, 1.18) in log-transformed ferritin, 0.99 (95% CI: 0.93, 1.06) in transferrin saturation, and 1.03 (95% CI: 0.93, 1.14) in transferrin in the TAGC dataset (none of the values were statistically significant). An age at onset-stratified analysis in the GABRIEL dataset suggested no effect of iron status in childhood onset, later onset, or unknown age at onset asthma. Findings from the liberal approach were similar, and the results persisted in sensitivity analyses (all P > 0.05). CONCLUSIONS This MR study does not provide evidence of an effect of iron status on asthma, suggesting that efforts to change iron concentrations will probably not result in decreased risk of asthma.
Collapse
Affiliation(s)
- Lulu Huang
- Center for Genomic and Personalized Medicine, Guangxi Medical University, Nanning, Guangxi, China.,School of Public Health, Guangxi Medical University, Nanning, Guangxi, China.,School of Nursing, Guangxi Medical University, Nanning, Guangxi, China
| | - Longman Li
- Center for Genomic and Personalized Medicine, Guangxi Medical University, Nanning, Guangxi, China.,School of Public Health, Guangxi Medical University, Nanning, Guangxi, China
| | - Xiaoyu Luo
- Center for Genomic and Personalized Medicine, Guangxi Medical University, Nanning, Guangxi, China.,School of Public Health, Guangxi Medical University, Nanning, Guangxi, China
| | - Sifang Huang
- Center for Genomic and Personalized Medicine, Guangxi Medical University, Nanning, Guangxi, China.,School of Public Health, Guangxi Medical University, Nanning, Guangxi, China
| | - Qingzhi Hou
- Center for Genomic and Personalized Medicine, Guangxi Medical University, Nanning, Guangxi, China.,School of Public Health, Guangxi Medical University, Nanning, Guangxi, China
| | - Xiaoting Ge
- Center for Genomic and Personalized Medicine, Guangxi Medical University, Nanning, Guangxi, China.,School of Public Health, Guangxi Medical University, Nanning, Guangxi, China
| | - Yingnan Lv
- Center for Genomic and Personalized Medicine, Guangxi Medical University, Nanning, Guangxi, China.,School of Public Health, Guangxi Medical University, Nanning, Guangxi, China
| | - Zengnan Mo
- Center for Genomic and Personalized Medicine, Guangxi Medical University, Nanning, Guangxi, China
| | - Xiaobo Yang
- Center for Genomic and Personalized Medicine, Guangxi Medical University, Nanning, Guangxi, China.,Department of Occupational Health and Environmental Health, School of Public Health, Guangxi Medical University, Nanning, Guangxi, China
| |
Collapse
|
63
|
Ferreira GC, Oberstaller J, Fonseca R, Keller TE, Adapa SR, Gibbons J, Wang C, Liu X, Li C, Pham M, Dayhoff Ii GW, Duong LM, Reyes LT, Laratelli LE, Franz D, Fatumo S, Bari AG, Freischel A, Fiedler L, Dokur O, Sharma K, Cragun D, Busby B, Jiang RHY. Iron Hack - A symposium/hackathon focused on porphyrias, Friedreich's ataxia, and other rare iron-related diseases. F1000Res 2019; 8:1135. [PMID: 31824661 PMCID: PMC6894363 DOI: 10.12688/f1000research.19140.1] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 06/05/2019] [Indexed: 01/14/2023] Open
Abstract
Background: Basic and clinical scientific research at the University of South Florida (USF) have intersected to support a multi-faceted approach around a common focus on rare iron-related diseases. We proposed a modified version of the National Center for Biotechnology Information’s (NCBI) Hackathon-model to take full advantage of local expertise in building “Iron Hack”, a rare disease-focused hackathon. As the collaborative, problem-solving nature of hackathons tends to attract participants of highly-diverse backgrounds, organizers facilitated a symposium on rare iron-related diseases, specifically porphyrias and Friedreich’s ataxia, pitched at general audiences. Methods: The hackathon was structured to begin each day with presentations by expert clinicians, genetic counselors, researchers focused on molecular and cellular biology, public health/global health, genetics/genomics, computational biology, bioinformatics, biomolecular science, bioengineering, and computer science, as well as guest speakers from the American Porphyria Foundation (APF) and Friedreich’s Ataxia Research Alliance (FARA) to inform participants as to the human impact of these diseases. Results: As a result of this hackathon, we developed resources that are relevant not only to these specific disease-models, but also to other rare diseases and general bioinformatics problems. Within two and a half days, “Iron Hack” participants successfully built collaborative projects to visualize data, build databases, improve rare disease diagnosis, and study rare-disease inheritance. Conclusions: The purpose of this manuscript is to demonstrate the utility of a hackathon model to generate prototypes of generalizable tools for a given disease and train clinicians and data scientists to interact more effectively.
Collapse
Affiliation(s)
- Gloria C Ferreira
- Department of Molecular Medicine, Morsani College of Medicine, University of South Florida, MDC 7, Tampa, FL, 33612, USA
| | - Jenna Oberstaller
- Global and Planetary Health, College of Public Health, University of South Florida, USF Genomics Program, 3720 Spectrum Blvd, Tampa, FL, 33612, USA
| | - Renée Fonseca
- Morsani College of Medicine, University of South Florida, 12901 Bruce B Downs Blvd, Tampa, FL, 33612, USA
| | - Thomas E Keller
- University of South Florida, USF Genomics Program, 3720 Spectrum Blvd, Tampa, FL, 33612, USA
| | - Swamy Rakesh Adapa
- Global and Planetary Health, College of Public Health, University of South Florida, USF Genomics Program, 3720 Spectrum Blvd, Tampa, FL, 33612, USA
| | - Justin Gibbons
- Morsani College of Medicine, University of South Florida, 12901 Bruce B Downs Blvd, Tampa, FL, 33612, USA
| | - Chengqi Wang
- Global and Planetary Health, College of Public Health, University of South Florida, USF Genomics Program, 3720 Spectrum Blvd, Tampa, FL, 33612, USA
| | - Xiaoming Liu
- Global and Planetary Health, College of Public Health, University of South Florida, USF Genomics Program, 3720 Spectrum Blvd, Tampa, FL, 33612, USA
| | - Chang Li
- Global and Planetary Health, College of Public Health, University of South Florida, USF Genomics Program, 3720 Spectrum Blvd, Tampa, FL, 33612, USA
| | - Minh Pham
- Center for Urban Transportation Research, University of South Florida, 4202 E. Fowler Avenue, CUT100, Tampa, FL, 33620, USA
| | - Guy W Dayhoff Ii
- Department of Chemistry, University of South Florida, 4202 E. Fowler Avenue, CHE 205, Tampa, FL, 33620-5250, USA
| | - Linh M Duong
- College of Public Health, University of South Florida, 13201 Bruce B. Downs Blvd., MDC 56, Tampa, FL, 33612, USA.,Moffitt Cancer Center, Tampa, FL, 33612, USA
| | - Luis Tañón Reyes
- Department of Cell Biology, Microbiology and Molecular Biology, University of South Florida, 4202 East Fowler Ave, ISA 2015 Tampa, FL, 33620, USA
| | - Luciano Enrique Laratelli
- Department of Chemistry, University of South Florida, 4202 E. Fowler Avenue, CHE 205, Tampa, FL, 33620-5250, USA
| | - Douglas Franz
- Department of Chemistry, University of South Florida, 4202 E. Fowler Avenue, CHE 205, Tampa, FL, 33620-5250, USA
| | - Segun Fatumo
- MRC/UVRI and LSHTM (Uganda Research Unit), Entebbe, Uganda
| | - Atm Golam Bari
- Department of Computer Science and Engineering, University of South Florida, Tampa, FL, USA
| | | | - Lindsey Fiedler
- College of Public Health, University of South Florida, 13201 Bruce B. Downs Blvd., MDC 56, Tampa, FL, 33612, USA
| | - Omkar Dokur
- Department of Computer Science and Engineering, University of South Florida, Tampa, FL, USA
| | | | - Deborah Cragun
- Global and Planetary Health, College of Public Health, University of South Florida, USF Genomics Program, 3720 Spectrum Blvd, Tampa, FL, 33612, USA
| | - Ben Busby
- National Library of Medicine, 8600 Rockville Pike, Bethesda, MD, 20894-6075, USA
| | - Rays H Y Jiang
- Global and Planetary Health, College of Public Health, University of South Florida, USF Genomics Program, 3720 Spectrum Blvd, Tampa, FL, 33612, USA
| |
Collapse
|
64
|
Alfadhel M, Nashabat M, Abu Ali Q, Hundallah K. Mitochondrial iron-sulfur cluster biogenesis from molecular understanding to clinical disease. ACTA ACUST UNITED AC 2019; 22:4-13. [PMID: 28064324 PMCID: PMC5726836 DOI: 10.17712/nsj.2017.1.20160542] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/04/2022]
Abstract
Iron–sulfur clusters (ISCs) are known to play a major role in various protein functions. Located in the mitochondria, cytosol, endoplasmic reticulum and nucleus, they contribute to various core cellular functions. Until recently, only a few human diseases related to mitochondrial ISC biogenesis defects have been described. Such diseases include Friedreich ataxia, combined oxidative phosphorylation deficiency 19, infantile complex II/III deficiency defect, hereditary myopathy with lactic acidosis and mitochondrial muscle myopathy, lipoic acid biosynthesis defects, multiple mitochondrial dysfunctions syndromes and non ketotic hyperglycinemia due to glutaredoxin 5 gene defect. Disorders of mitochondrial import, export and translation, including sideroblastic anemia with ataxia, EVEN-PLUS syndrome and mitochondrial complex I deficiency due to nucleotide-binding protein-like protein gene defect, have also been implicated in ISC biogenesis defects. With advances in next generation sequencing technologies, more disorders related to ISC biogenesis defects are expected to be elucidated. In this article, we aim to shed the light on mitochondrial ISC biogenesis, related proteins and their function, pathophysiology, clinical phenotypes of related disorders, diagnostic approach, and future implications.
Collapse
Affiliation(s)
- Majid Alfadhel
- Division of Genetics, Department of Pediatrics, King Saud bin Abdulaziz University for Health Sciences, King Abdulaziz Medical City, Riyadh, Kingdom of Saudi Arabia
| | | | | | | |
Collapse
|
65
|
Abstract
Organisms from all kingdoms of life use iron-proteins in a multitude of functional processes. We applied a bioinformatics approach to investigate the human portfolio of iron-proteins. We separated iron-proteins based on the chemical nature of their metal-containing cofactors: individual iron ions, heme cofactors and iron-sulfur clusters. We found that about 2% of human genes encode an iron-protein. Of these, 35% are proteins binding individual iron ions, 48% are heme-binding proteins and 17% are iron-sulfur proteins. More than half of the human iron-proteins have a catalytic function. Indeed, we predict that 6.5% of all human enzymes are iron-dependent. This percentage is quite different for the various enzyme classes. Human oxidoreductases feature the largest fraction of iron-dependent family members (about 37%). The distribution of iron proteins in the various cellular compartments is uneven. In particular, the mitochondrion and the endoplasmic reticulum are enriched in iron-proteins with respect to the average content of the cell. Finally, we observed that genes encoding iron-proteins are more frequently associated to pathologies than the all other human genes on average. The present research provides an extensive overview of iron usage by the human proteome, and highlights several specific features of the physiological role of iron ions in human cells.
Collapse
Affiliation(s)
- Claudia Andreini
- Magnetic Resonance Center, University of Florence, Via Luigi Sacconi 6, Sesto Fiorentino 50019, Italy.
| | | | | | | |
Collapse
|
66
|
Eidi M, Garshasbi M. A novel ISCA2 variant responsible for an early-onset neurodegenerative mitochondrial disorder: a case report of multiple mitochondrial dysfunctions syndrome 4. BMC Neurol 2019; 19:153. [PMID: 31279336 PMCID: PMC6612116 DOI: 10.1186/s12883-019-1387-2] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2018] [Accepted: 07/01/2019] [Indexed: 12/30/2022] Open
Abstract
BACKGROUND Multiple Mitochondrial Dysfunctions Syndrome 4 (MMDS4) is manifested as a result of ISCA2 mutations. ISCA2 is a vital component of 4Fe-4S clusters assembly machine. Therefore, in MMDS4 patients, deficient mitochondrial respiratory chain complexes I and II, Aconitase and Succinate dehydrogenase of Kerbs cycle and Lipoic Acid Synthetase in the biosynthesis of lipoic acid are expected. CASE PRESENTATIONS A 7 months boy in an Iranian consanguineous family with progressive neurodegenerative problems was referred to us. Primarily, general laboratory tests, Abdomen ultrasonography and brain magnetic resonance imaging were performed. In order to find out the genetic problem in this case Whole Exome Sequencing (WES) following by Sanger sequencing was carried out. A novel variant (c.355G > A, p.Ala119Thr) in ISCA2 gene was identified by WES in the proband. Confirmation and segregation in the family for this variant was performed by Sanger sequencing. In-Silico prediction of the ISCA2 secondary structure showed that a helix motif in the Fe-S biosynthesis domain of ISCA2 protein will be eliminated as a result of this variant. CONCLUSIONS We reported the first patient with ISCA2 variant in Iranian population and the third one in the world reported for ISCA2 gene, so far associated with early-onset mitochondrial neurodegeneration. However further functional studies on this variant or finding it in other patients with similar clinical problems are needed to confirm the pathogenicity of this variant.
Collapse
Affiliation(s)
- Milad Eidi
- Department of Medical Genetics, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Masoud Garshasbi
- Department of Medical Genetics, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran.
| |
Collapse
|
67
|
Ast T, Meisel JD, Patra S, Wang H, Grange RMH, Kim SH, Calvo SE, Orefice LL, Nagashima F, Ichinose F, Zapol WM, Ruvkun G, Barondeau DP, Mootha VK. Hypoxia Rescues Frataxin Loss by Restoring Iron Sulfur Cluster Biogenesis. Cell 2019; 177:1507-1521.e16. [PMID: 31031004 PMCID: PMC6911770 DOI: 10.1016/j.cell.2019.03.045] [Citation(s) in RCA: 97] [Impact Index Per Article: 16.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2018] [Revised: 02/11/2019] [Accepted: 03/22/2019] [Indexed: 12/16/2022]
Abstract
Friedreich's ataxia (FRDA) is a devastating, multisystemic disorder caused by recessive mutations in the mitochondrial protein frataxin (FXN). FXN participates in the biosynthesis of Fe-S clusters and is considered to be essential for viability. Here we report that when grown in 1% ambient O2, FXN null yeast, human cells, and nematodes are fully viable. In human cells, hypoxia restores steady-state levels of Fe-S clusters and normalizes ATF4, NRF2, and IRP2 signaling events associated with FRDA. Cellular studies and in vitro reconstitution indicate that hypoxia acts through HIF-independent mechanisms that increase bioavailable iron as well as directly activate Fe-S synthesis. In a mouse model of FRDA, breathing 11% O2 attenuates the progression of ataxia, whereas breathing 55% O2 hastens it. Our work identifies oxygen as a key environmental variable in the pathogenesis associated with FXN depletion, with important mechanistic and therapeutic implications.
Collapse
Affiliation(s)
- Tslil Ast
- Broad Institute, Cambridge, MA 02142, USA; Howard Hughes Medical Institute, Howard Hughes Medical Institute, Massachusetts General Hospital, Boston, MA 02114, USA; Department of Molecular Biology, Massachusetts General Hospital, Boston, MA 02114, USA; Harvard Medical School, Boston, MA 02115, USA
| | - Joshua D Meisel
- Broad Institute, Cambridge, MA 02142, USA; Howard Hughes Medical Institute, Howard Hughes Medical Institute, Massachusetts General Hospital, Boston, MA 02114, USA; Department of Molecular Biology, Massachusetts General Hospital, Boston, MA 02114, USA; Harvard Medical School, Boston, MA 02115, USA
| | - Shachin Patra
- Department of Chemistry, Texas A&M University, College Station, TX 77843, USA
| | - Hong Wang
- Broad Institute, Cambridge, MA 02142, USA; Howard Hughes Medical Institute, Howard Hughes Medical Institute, Massachusetts General Hospital, Boston, MA 02114, USA; Department of Molecular Biology, Massachusetts General Hospital, Boston, MA 02114, USA; Harvard Medical School, Boston, MA 02115, USA
| | - Robert M H Grange
- Department of Anesthesia, Critical Care, and Pain Medicine, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Sharon H Kim
- Broad Institute, Cambridge, MA 02142, USA; Howard Hughes Medical Institute, Howard Hughes Medical Institute, Massachusetts General Hospital, Boston, MA 02114, USA; Department of Molecular Biology, Massachusetts General Hospital, Boston, MA 02114, USA; Harvard Medical School, Boston, MA 02115, USA
| | - Sarah E Calvo
- Broad Institute, Cambridge, MA 02142, USA; Howard Hughes Medical Institute, Howard Hughes Medical Institute, Massachusetts General Hospital, Boston, MA 02114, USA; Department of Molecular Biology, Massachusetts General Hospital, Boston, MA 02114, USA; Harvard Medical School, Boston, MA 02115, USA
| | - Lauren L Orefice
- Department of Molecular Biology, Massachusetts General Hospital, Boston, MA 02114, USA; Harvard Medical School, Boston, MA 02115, USA
| | - Fumiaki Nagashima
- Department of Anesthesia, Critical Care, and Pain Medicine, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Fumito Ichinose
- Department of Anesthesia, Critical Care, and Pain Medicine, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Warren M Zapol
- Department of Anesthesia, Critical Care, and Pain Medicine, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Gary Ruvkun
- Department of Molecular Biology, Massachusetts General Hospital, Boston, MA 02114, USA; Harvard Medical School, Boston, MA 02115, USA
| | - David P Barondeau
- Department of Chemistry, Texas A&M University, College Station, TX 77843, USA
| | - Vamsi K Mootha
- Broad Institute, Cambridge, MA 02142, USA; Howard Hughes Medical Institute, Howard Hughes Medical Institute, Massachusetts General Hospital, Boston, MA 02114, USA; Department of Molecular Biology, Massachusetts General Hospital, Boston, MA 02114, USA; Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|
68
|
Do E, Park S, Li MH, Wang JM, Ding C, Kronstad JW, Jung WH. The mitochondrial ABC transporter Atm1 plays a role in iron metabolism and virulence in the human fungal pathogen Cryptococcus neoformans. Med Mycol 2019; 56:458-468. [PMID: 29420779 DOI: 10.1093/mmy/myx073] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2017] [Accepted: 08/17/2017] [Indexed: 12/19/2022] Open
Abstract
Iron-sulfur clusters (ISC) are indispensable cofactors for essential enzymes in various cellular processes. In the model yeast Saccharomyces cerevisiae, the precursor of ISCs is exported from mitochondria via a mitochondrial ABC transporter Atm1 and used for cytosolic and nuclear ISC protein assembly. Although iron homeostasis has been implicated in the virulence of the human fungal pathogen Cryptococcus neoformans, the key components of the ISC biosynthesis pathway need to be fully elucidated. In the current study, a homolog of S. cerevisiae Atm1 was identified in C. neoformans, and its function was characterized. We constructed C. neoformans mutants lacking ATM1 and found that deletion of ATM1 affected mitochondrial functions. Furthermore, we observed diminished activity of the cytosolic ISC-containing protein Leu1 and the heme-containing protein catalase in the atm1 mutant. These results suggested that Atm1 is required for the biosynthesis of ISCs in the cytoplasm as well as heme metabolism in C. neoformans. In addition, the atm1 mutants were avirulent in a murine model of cryptococcosis. Overall, our results demonstrated that Atm1 plays a critical role in iron metabolism and virulence for C. neoformans.
Collapse
Affiliation(s)
- Eunsoo Do
- Department of Systems Biotechnology, Chung-Ang University, Anseong 17546, Korea
| | - Seho Park
- Department of Systems Biotechnology, Chung-Ang University, Anseong 17546, Korea
| | - Ming-Hui Li
- College of Life and Health Sciences, Northeastern University, Shenyang, Liaoning 110015, China
| | - Jia-Mei Wang
- College of Life and Health Sciences, Northeastern University, Shenyang, Liaoning 110015, China
| | - Chen Ding
- College of Life and Health Sciences, Northeastern University, Shenyang, Liaoning 110015, China
| | - James W Kronstad
- Michael Smith Laboratories, University of British Columbia, Vancouver, BC V6T 1Z4, Canada
| | - Won Hee Jung
- Department of Systems Biotechnology, Chung-Ang University, Anseong 17546, Korea
| |
Collapse
|
69
|
The biogenesis of iron-sulfur proteins: from cellular biology to molecular aspects. J Biol Inorg Chem 2019; 23:493-494. [PMID: 29637286 DOI: 10.1007/s00775-018-1555-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
|
70
|
Differentially Expressed miRNAs Influence Metabolic Processes in Pituitary Oncocytoma. Neurochem Res 2019; 44:2360-2371. [PMID: 30945144 PMCID: PMC6776564 DOI: 10.1007/s11064-019-02789-2] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2018] [Revised: 03/25/2019] [Accepted: 03/28/2019] [Indexed: 12/18/2022]
Abstract
Spindle cell oncocytomas (SCO) of the pituitary are rare tumors accounting for 0.1–0.4% of all sellar tumors. Due to their rarity, little information is available regarding their pathogenesis. Our aim was to investigate miRNA expression profile of pituitary oncocytomas. Total RNA was extracted from 9 formalin-fixed paraffin embedded pituitary samples (4 primary, 3 recurrent oncocytomas and 2 normal tissues). Next-generation sequencing was performed for miRNA profiling. Transcriptome data of additional 6 samples’ were obtained from NBCI GEO database for gene expression reanalysis and tissue-specific target prediction. Bioinformatical analysis, in vitro miRNA mimics transfection, luciferase reporter system and AlamarBlue assay were applied to characterize miRNA’s function. 54 differentially expressed miRNAs and 485 genes in pituitary SCO vs. normal tissue and 8 miRNAs in recurrent vs. primary SCO were determined. Global miRNA downregulation and decreased level of DROSHA were detected in SCO samples vs. normal tissue. Transcriptome analysis revealed cell cycle alterations while miRNAs influenced mainly metabolic processes (tricarboxylic acid cycle-TCA, carbohydrate, lipid metabolism). Through miRNA-target interaction network the overexpressed Aconitase 2 potentially targeted by two downregulated miRNAs (miR-744-5p, miR-127-3p) was revealed. ACO2 and miR-744-5p interaction was validated by luciferase assay. MiR-127-3p and miR-744-5p significantly decreased cell proliferation in vitro. Our study firstly reported miRNA profile of pituitary oncocytoma. Our results suggest that tumor suppressor miRNAs may have an essential role in the pathogenesis of pituitary oncocytoma. Earlier reports showed downregulated TCA cycle in SCO which is extended by our results adding the role of miR-744-5p targeting ACO2.
Collapse
|
71
|
Patnaik MM, Tefferi A. Refractory anemia with ring sideroblasts (RARS) and RARS with thrombocytosis: "2019 Update on Diagnosis, Risk-stratification, and Management". Am J Hematol 2019; 94:475-488. [PMID: 30618061 DOI: 10.1002/ajh.25397] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2018] [Accepted: 01/02/2019] [Indexed: 12/17/2022]
Abstract
DISEASE OVERVIEW Ring sideroblasts (RS) are erythroid precursors with abnormal perinuclear mitochondrial iron accumulation. Two myeloid neoplasms defined by the presence of RS, include refractory anemia with ring sideroblasts (RARS), now classified under myelodysplastic syndromes with RS (MDS-RS) and RARS with thrombocytosis (RARS-T); now called myelodysplastic/myeloproliferative neoplasm with RS and thrombocytosis (MDS/MPN-RS-T). DIAGNOSIS MDS-RS is a lower-risk MDS, with single or multilineage dysplasia (SLD/MLD), <5% bone marrow (BM) blasts and ≥ 15% BM RS (≥5% in the presence of SF3B1 mutations). MDS/MPN-RS-T, now a formal entity in the MDS/MPN overlap syndromes, has diagnostic features of MDS-RS-SLD, along with a platelet count ≥450 × 10(9)/L and large atypical megakaryocytes. MUTATIONS AND KARYOTYPE Mutations in SF3B1 are seen in ≥80% of patients with MDS-RS-SLD and MDS/MPN-RS-T, and strongly correlate with the presence of BM RS; MDS/MPN-RS-T patients also demonstrate JAK2V617F, ASXL1, DNMT3A, SETBP1, and TET2 mutations. Cytogenetic abnormalities are uncommon in both. RISK STRATIFICATION Most patients with MDS-RS-SLD are stratified into lower-risk groups by the revised-IPSS. Disease outcome in MDS/MPN-RS-T is better than that of MDS-RS-SLD, but worse than that of essential thrombocythemia. Both diseases have a low risk of leukemic transformation. TREATMENT Anemia and iron overload are complications seen in both and are managed similar to lower-risk MDS and MPN. The advent of luspatercept, a first-in-class erythroid maturation agent will tremendously boost the ability to manage anemia. Aspirin therapy is reasonable in MDS/MPN-RS-T, especially in the presence of JAK2V617F, but the value of platelet-lowering drugs remains uncertain.
Collapse
Affiliation(s)
- Mrinal M. Patnaik
- Division of Hematology, Department of Internal Medicine Mayo Clinic Rochester Minnesota
| | - Ayalew Tefferi
- Division of Hematology, Department of Internal Medicine Mayo Clinic Rochester Minnesota
| |
Collapse
|
72
|
Rouault TA. The indispensable role of mammalian iron sulfur proteins in function and regulation of multiple diverse metabolic pathways. Biometals 2019; 32:343-353. [PMID: 30923992 PMCID: PMC6584224 DOI: 10.1007/s10534-019-00191-7] [Citation(s) in RCA: 58] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2019] [Accepted: 03/18/2019] [Indexed: 02/07/2023]
Abstract
In recent years, iron sulfur (Fe–S) proteins have been identified as key players in mammalian metabolism, ranging from long-known roles in the respiratory complexes and the citric acid cycle, to more recently recognized roles in RNA and DNA metabolism. Fe–S cofactors have often been missed because of their intrinsic lability and oxygen sensitivity. More Fe–S proteins have now been identified owing to detection of their direct interactions with components of the Fe–S biogenesis machinery, and through use of informatics to detect a motif that binds the co-chaperone responsible for transferring nascent Fe–S clusters to domains of recipient proteins. Dissection of the molecular steps involved in Fe–S transfer to Fe–S proteins has revealed that direct and shielded transfer occurs through highly conserved pathways that operate in parallel in the mitochondrial matrix and in the cytosolic/nuclear compartments of eukaryotic cells. Because Fe–S clusters have the unusual ability to accept or donate single electrons in chemical reactions, their presence renders complex chemical reactions possible. In addition, Fe–S clusters may function as sensors that interconnect activity of metabolic pathways with cellular redox status. Presence in pathways that control growth and division may enable cells to regulate their growth according to sufficiency of energy stores represented by redox capacity, and oxidation of such proteins could diminish anabolic activities to give cells an opportunity to restore energy supplies. This review will discuss mechanisms of Fe–S biogenesis and delivery, and methods that will likely reveal important roles of Fe–S proteins in proteins not yet recognized as Fe–S proteins.
Collapse
|
73
|
Iwai K. Regulation of cellular iron metabolism: Iron-dependent degradation of IRP by SCF FBXL5 ubiquitin ligase. Free Radic Biol Med 2019; 133:64-68. [PMID: 30218771 DOI: 10.1016/j.freeradbiomed.2018.09.011] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/22/2018] [Revised: 09/10/2018] [Accepted: 09/12/2018] [Indexed: 12/30/2022]
Abstract
Because of essentiality and toxicity of iron in our body, iron metabolism is tightly regulated in cells. In mammalian cells, iron regulatory protein 1 and 2 (IRP1 and IRP2) are the central regulators of cellular iron metabolism. IRPs regulate iron metabolism by interacting with the RNA stem-loop structures, iron-responsive elements (IREs), found on the transcripts encoding proteins involved in iron metabolism only in iron depleted condition. It is also well-known that the ubiquitin system plays central roles in cellular iron regulation because both IRPs having the IRE binding activity are recognized and ubiquitinated by the SCFFBXL5 ubiquitin ligase in condition of iron-replete. FBXL5, which is a substrate recognition subunit of SCFFBXL5, senses iron availability via its hemerythrin-like domain. In this small article, current understanding of the roles of SCFFBXL5-mediated degradation of IRPs played in cellular iron metabolism is discussed.
Collapse
Affiliation(s)
- Kazuhiro Iwai
- Department of Molecular and Cellular Physiology, Graduate School of Medicine, Kyoto University, Yoshida-konoe-cho, Sakyo-ku, Kyoto 606-8501, Japan.
| |
Collapse
|
74
|
Ma Q, Dasgupta C, Li Y, Huang L, Zhang L. MicroRNA-210 Downregulates ISCU and Induces Mitochondrial Dysfunction and Neuronal Death in Neonatal Hypoxic-Ischemic Brain Injury. Mol Neurobiol 2019; 56:5608-5625. [PMID: 30656514 DOI: 10.1007/s12035-019-1491-8] [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] [Received: 11/20/2018] [Accepted: 01/10/2019] [Indexed: 02/01/2023]
Abstract
Neonatal hypoxic-ischemic (HI) brain injury causes significant mortality and long-term neurologic sequelae. We previously demonstrated that HI significantly increased microRNA-210 (miR-210) in the neonatal rat brain and inhibition of brain endogenous miR-210 was neuroprotective in HI brain injury. However, the molecular mechanisms underpinning this neuroprotection remain unclear. Using both in vivo and in vitro models, herein we uncover a novel mechanism mediating oxidative brain injury after neonatal HI, in which miR-210 induces mitochondrial dysfunction via downregulation of iron-sulfur cluster assembly protein (ISCU). Inhibition of miR-210 significantly ameliorates mitochondrial dysfunction, oxidative stress, and neuronal loss in the neonatal brain subjected to HI, as well as in primary cortical neurons exposed to oxygen-glucose deprivation (OGD). These effects are mediated through ISCU, in that miR-210 mimic decreases ISCU abundance in the brains of rat pups and primary cortical neurons, and inhibition of miR-210 protects ISCU against HI in vivo or OGD in vitro. Deletion of miR-210 binding sequences at the 3'UTR of ISCU transcript ablates miR-210-induced downregulation of ISCU protein abundance in PC12 cells. In primary cortical neurons, miR-210 mimic or silencing ISCU results in mitochondrial dysfunction, reactive oxygen species production, and activation of caspase-dependent death pathways. Of importance, knockdown of ISCU increases HI-induced injury in the neonatal rat brain and counteracts the neuroprotection of miR-210 inhibition. Therefore, miR-210 by downregulating ISCU and inducing mitochondrial dysfunction in neurons is a potent contributor of oxidative brain injury after neonatal HI.
Collapse
Affiliation(s)
- Qingyi Ma
- The Lawrence D. Longo Center for Perinatal Biology, Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, CA, 92350, USA.
| | - Chiranjib Dasgupta
- The Lawrence D. Longo Center for Perinatal Biology, Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, CA, 92350, USA
| | - Yong Li
- The Lawrence D. Longo Center for Perinatal Biology, Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, CA, 92350, USA
| | - Lei Huang
- The Lawrence D. Longo Center for Perinatal Biology, Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, CA, 92350, USA
| | - Lubo Zhang
- The Lawrence D. Longo Center for Perinatal Biology, Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, CA, 92350, USA.
| |
Collapse
|
75
|
Ciofi-Baffoni S, Nasta V, Banci L. Protein networks in the maturation of human iron-sulfur proteins. Metallomics 2019; 10:49-72. [PMID: 29219157 DOI: 10.1039/c7mt00269f] [Citation(s) in RCA: 82] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
The biogenesis of iron-sulfur (Fe-S) proteins in humans is a multistage process occurring in different cellular compartments. The mitochondrial iron-sulfur cluster (ISC) assembly machinery composed of at least 17 proteins assembles mitochondrial Fe-S proteins. A cytosolic iron-sulfur assembly (CIA) machinery composed of at least 13 proteins has been more recently identified and shown to be responsible for the Fe-S cluster incorporation into cytosolic and nuclear Fe-S proteins. Cytosolic and nuclear Fe-S protein maturation requires not only the CIA machinery, but also the components of the mitochondrial ISC assembly machinery. An ISC export machinery, composed of a protein transporter located in the mitochondrial inner membrane, has been proposed to act in mediating the export process of a still unknown component that is required for the CIA machinery. Several functional and molecular aspects of the protein networks operative in the three machineries are still largely obscure. This Review focuses on the Fe-S protein maturation processes in humans with the specific aim of providing a molecular picture of the currently known protein-protein interaction networks. The human ISC and CIA machineries are presented, and the ISC export machinery is discussed with respect to possible molecules being the substrates of the mitochondrial protein transporter.
Collapse
Affiliation(s)
- Simone Ciofi-Baffoni
- Magnetic Resonance Center-CERM, University of Florence, Via Luigi Sacconi 6, 50019, Sesto Fiorentino, Florence, Italy.
| | | | | |
Collapse
|
76
|
McCarthy EL, Rankin AN, Dill ZR, Booker SJ. The A-type domain in Escherichia coli NfuA is required for regenerating the auxiliary [4Fe-4S] cluster in Escherichia coli lipoyl synthase. J Biol Chem 2018; 294:1609-1617. [PMID: 30538130 DOI: 10.1074/jbc.ra118.006171] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2018] [Revised: 12/10/2018] [Indexed: 11/06/2022] Open
Abstract
The lipoyl cofactor plays an integral role in several essential biological processes. The last step in its de novo biosynthetic pathway, the attachment of two sulfur atoms at C6 and C8 of an n-octanoyllysyl chain, is catalyzed by lipoyl synthase (LipA), a member of the radical SAM superfamily. In addition to the [4Fe-4S] cluster common to all radical SAM enzymes, LipA contains a second [4Fe-4S] auxiliary cluster, which is sacrificed during catalysis to supply the requisite sulfur atoms, rendering the protein inactive for further turnovers. Recently, it was shown that the Fe-S cluster carrier protein NfuA from Escherichia coli can regenerate the auxiliary cluster of E. coli LipA after each turnover, but the molecular mechanism is incompletely understood. Herein, using protein-protein interaction and kinetic assays as well as site-directed mutagenesis, we provide further insight into the mechanism of NfuA-mediated cluster regeneration. In particular, we show that the N-terminal A-type domain of E. coli NfuA is essential for its tight interaction with LipA. Further, we demonstrate that NfuA from Mycobacterium tuberculosis can also regenerate the auxiliary cluster of E. coli LipA. However, an Nfu protein from Staphylococcus aureus, which lacks the A-type domain, was severely diminished in facilitating cluster regeneration. Of note, addition of the N-terminal domain of E. coli NfuA to S. aureus Nfu, fully restored cluster-regenerating activity. These results expand our understanding of the newly discovered mechanism by which the auxiliary cluster of LipA is restored after each turnover.
Collapse
Affiliation(s)
- Erin L McCarthy
- Department of Biochemistry and Molecular Biology, The Pennsylvania State University, University Park, Pennsylvania 16802
| | - Ananda N Rankin
- Department of Chemistry, The Pennsylvania State University, University Park, Pennsylvania 16802
| | - Zerick R Dill
- Department of Chemistry, The Pennsylvania State University, University Park, Pennsylvania 16802
| | - Squire J Booker
- Department of Biochemistry and Molecular Biology, The Pennsylvania State University, University Park, Pennsylvania 16802; Department of Chemistry, The Pennsylvania State University, University Park, Pennsylvania 16802; Howard Hughes Medical Institute, The Pennsylvania State University, University Park, Pennsylvania 16802.
| |
Collapse
|
77
|
Rawcliffe DFR, Österman L, Nordin A, Holmberg M. PTBP1 acts as a dominant repressor of the aberrant tissue-specific splicing of ISCU in hereditary myopathy with lactic acidosis. Mol Genet Genomic Med 2018; 6:887-897. [PMID: 30209894 PMCID: PMC6305642 DOI: 10.1002/mgg3.413] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2018] [Revised: 03/23/2018] [Accepted: 04/17/2018] [Indexed: 01/23/2023] Open
Abstract
BACKGROUND Hereditary myopathy with lactic acidosis (HML) is an autosomal recessive disease caused by an intron mutation in the iron-sulfur cluster assembly (ISCU) gene. The mutation results in aberrant splicing, where part of the intron is retained in the final mRNA transcript, giving rise to a truncated nonfunctional ISCU protein. Using an ISCU mini-gene system, we have previously shown that PTBP1 can act as a repressor of the mis-splicing of ISCU, where overexpression of PTBP1 resulted in a decrease of the incorrect splicing. In this study, we wanted to, in more detail, analyze the role of PTBP1 in the regulation of endogenous ISCU mis-splicing. METHODS Overexpression and knockdown of PTBP1 was performed in myoblasts from two HML patients and a healthy control. Quantification of ISCU mis-splicing was done by qRTPCR. Biotinylated ISCU RNA, representing wildtype and mutant intron sequence, was used in a pull-down assay with nuclear extracts from myoblasts. Levels of PTBP1 in human cell lines and mice tissues were analyzed by qRTPCR and western blot. RESULTS PTBP1 overexpression in HML patient myoblasts resulted in a substantial decrease of ISCU mis-splicing while knockdown of PTBP1 resulted in a drastic increase. The effect could be observed in both patient and control myoblasts. We could also show that PTBP1 interacts with both the mutant and wild-type ISCU intron sequence, but with a higher affinity to the mutant sequence. Furthermore, low levels of PTBP1 among examined mouse tissues correlated with high levels of incorrect splicing of ISCU. CONCLUSION Our results show that PTBP1 acts as a dominant repressor of ISCU mis-splicing. We also show an inverse correlation between the levels of PTBP1 and ISCU mis-splicing, suggesting that the high level of mis-splicing in the skeletal muscle is primarily due to the low levels of PTBP1.
Collapse
Affiliation(s)
- Denise F. R. Rawcliffe
- Unit for Medical and Clinical GeneticsDepartment of Medical BiosciencesUmeå UniversityUmeåSweden
| | - Lennart Österman
- Unit for Medical and Clinical GeneticsDepartment of Medical BiosciencesUmeå UniversityUmeåSweden
| | - Angelica Nordin
- Unit for Medical and Clinical GeneticsDepartment of Medical BiosciencesUmeå UniversityUmeåSweden
| | - Monica Holmberg
- Unit for Medical and Clinical GeneticsDepartment of Medical BiosciencesUmeå UniversityUmeåSweden
| |
Collapse
|
78
|
Torgovnick A, Schiavi A, Shaik A, Kassahun H, Maglioni S, Rea SL, Johnson TE, Reinhardt HC, Honnen S, Schumacher B, Nilsen H, Ventura N. BRCA1 and BARD1 mediate apoptotic resistance but not longevity upon mitochondrial stress in Caenorhabditis elegans. EMBO Rep 2018; 19:embr.201845856. [PMID: 30366941 DOI: 10.15252/embr.201845856] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2018] [Revised: 09/14/2018] [Accepted: 09/26/2018] [Indexed: 02/05/2023] Open
Abstract
Interventions that promote healthy aging are typically associated with increased stress resistance. Paradoxically, reducing the activity of core biological processes such as mitochondrial or insulin metabolism promotes the expression of adaptive responses, which in turn increase animal longevity and resistance to stress. In this study, we investigated the relation between the extended Caenorhabditis elegans lifespan elicited by reduction in mitochondrial functionality and resistance to genotoxic stress. We find that reducing mitochondrial activity during development confers germline resistance to DNA damage-induced cell cycle arrest and apoptosis in a cell-non-autonomous manner. We identified the C. elegans homologs of the BRCA1/BARD1 tumor suppressor genes, brc-1/brd-1, as mediators of the anti-apoptotic effect but dispensable for lifespan extension upon mitochondrial stress. Unexpectedly, while reduced mitochondrial activity only in the soma was not sufficient to promote longevity, its reduction only in the germline or in germline-less strains still prolonged lifespan. Thus, in animals with partial reduction in mitochondrial functionality, the mechanisms activated during development to safeguard the germline against genotoxic stress are uncoupled from those required for somatic robustness and animal longevity.
Collapse
Affiliation(s)
- Alessandro Torgovnick
- Leibniz Research Institute for Environmental Medicine (IUF), Düsseldorf, Germany.,Clinic I of Internal Medicine, Center for Integrated Oncology, Center for Molecular Medicine and the CECAD Research Center, University of Cologne, Cologne, Germany.,Medical Faculty, Institute for Genome Stability in Aging and Disease, CECAD Research Center, University of Cologne, Cologne, Germany
| | - Alfonso Schiavi
- Leibniz Research Institute for Environmental Medicine (IUF), Düsseldorf, Germany.,Institute for Clinical Chemistry and Laboratory Diagnostic, Medical Faculty, Heinrich Heine University of Düsseldorf, Düsseldorf, Germany
| | - Anjumara Shaik
- Leibniz Research Institute for Environmental Medicine (IUF), Düsseldorf, Germany
| | - Henok Kassahun
- Department of Clinical Molecular Biology, University of Oslo, Oslo, Norway.,Akershus University, Akershus, Norway
| | - Silvia Maglioni
- Leibniz Research Institute for Environmental Medicine (IUF), Düsseldorf, Germany
| | - Shane L Rea
- Department of Pathology, University of Washington, Seattle, WA, USA
| | - Thomas E Johnson
- Institute for Behavioral Genetics & Department of Integrative Physiology, University of Colorado at Boulder, Boulder, CO, USA
| | - Hans C Reinhardt
- Clinic I of Internal Medicine, Center for Integrated Oncology, Center for Molecular Medicine and the CECAD Research Center, University of Cologne, Cologne, Germany
| | - Sebastian Honnen
- Medical Faculty, Institute of Toxicology, Heinrich Heine University of Düsseldorf, Düsseldorf, Germany
| | - Björn Schumacher
- Medical Faculty, Institute for Genome Stability in Aging and Disease, CECAD Research Center, University of Cologne, Cologne, Germany
| | - Hilde Nilsen
- Department of Clinical Molecular Biology, University of Oslo, Oslo, Norway.,Akershus University, Akershus, Norway
| | - Natascia Ventura
- Leibniz Research Institute for Environmental Medicine (IUF), Düsseldorf, Germany .,Institute for Clinical Chemistry and Laboratory Diagnostic, Medical Faculty, Heinrich Heine University of Düsseldorf, Düsseldorf, Germany
| |
Collapse
|
79
|
Wang Y, Song F, Zhu J, Zhang Y, Du L, Kan C. Highly selective fluorescent probe based on a rhodamine B and furan-2-carbonyl chloride conjugate for detection of Fe3+ in cells. Tetrahedron Lett 2018. [DOI: 10.1016/j.tetlet.2018.09.003] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
|
80
|
Ziemka-Nalecz M, Jaworska J, Sypecka J, Zalewska T. Histone Deacetylase Inhibitors: A Therapeutic Key in Neurological Disorders? J Neuropathol Exp Neurol 2018; 77:855-870. [DOI: 10.1093/jnen/nly073] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Affiliation(s)
- Malgorzata Ziemka-Nalecz
- NeuroRepair Department, Mossakowski Medical Research Centre, Polish Academy of Sciences, Warsaw, Poland
| | - Joanna Jaworska
- NeuroRepair Department, Mossakowski Medical Research Centre, Polish Academy of Sciences, Warsaw, Poland
| | - Joanna Sypecka
- NeuroRepair Department, Mossakowski Medical Research Centre, Polish Academy of Sciences, Warsaw, Poland
| | - Teresa Zalewska
- NeuroRepair Department, Mossakowski Medical Research Centre, Polish Academy of Sciences, Warsaw, Poland
| |
Collapse
|
81
|
Volani C, Doerrier C, Demetz E, Haschka D, Paglia G, Lavdas AA, Gnaiger E, Weiss G. Dietary iron loading negatively affects liver mitochondrial function. Metallomics 2018; 9:1634-1644. [PMID: 29026901 DOI: 10.1039/c7mt00177k] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Iron is an essential co-factor for several metabolic processes, including mitochondrial respiration, and mitochondria are the major sites of iron-utilization. Cellular iron homeostasis must be tightly regulated, as intracellular iron deficiency can lead to insufficient energy production, whereas iron overload triggers ROS (reactive oxygen species) formation via the Fenton reaction. So far little is known on how iron imbalances affect mitochondrial function in vivo and the impact of the genotype on that, we studied the effects of dietary iron loading on mitochondrial respiratory capacity in liver by comparing two genetically divergent mouse strains, namely C57BL/6N and FVB mice. Both mouse strains differed in their basal iron levels and their metabolic responses to iron loading as determined by expression of iron trafficking proteins (ferritin was increased in livers of animals receiving high iron diet) as well as tissue iron content (2-fold increase, FVB p = 0.0013; C57BL/6N p = 0.0022). Dietary iron exposure caused a significant impairment of mitochondrial oxidative phosphorylation, especially regarding OXPHOS capacity (FVB p = 0.0006; C57BL/6N p = 0.0087) and S-ETS capacity (FVB p = 0.0281; C57BL/6N p = 0.0159). These effects were more pronounced in C57BL/6N than in FVB mice and were paralleled by an iron mediated induction of oxidative stress in mitochondria. The increased susceptibility of C57BL6/N mice to iron loading may be due to reduced expression of anti-oxidant defense mechanisms and altered iron trafficking upon dietary challenge pointing to a role of genetic modifiers for cellular and mitochondrial iron trafficking. Finally, iron-mediated induction of mitochondrial oxidative stress and reduction of oxidative phosphorylation may underlie fatigue in subjects with iron loading diseases.
Collapse
Affiliation(s)
- Chiara Volani
- Department of Internal Medicine II (Infectious Diseases, Immunology, Rheumatology and Pneumology), Medical University of Innsbruck, Anichstr. 35, A-6020 Innsbruck, Austria.
| | | | | | | | | | | | | | | |
Collapse
|
82
|
Dos Santos PC. B. subtilis as a Model for Studying the Assembly of Fe-S Clusters in Gram-Positive Bacteria. Methods Enzymol 2018; 595:185-212. [PMID: 28882201 DOI: 10.1016/bs.mie.2017.07.009] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Complexes of iron and sulfur (Fe-S clusters) are widely distributed in nature and participate in essential biochemical reactions. The biological formation of Fe-S clusters involves dedicated pathways responsible for the mobilization of sulfur, the assembly of Fe-S clusters, and the transfer of these clusters to target proteins. Genomic analysis of Bacillus subtilis and other Gram-positive bacteria indicated the presence of only one Fe-S cluster biosynthesis pathway, which is distinct in number of components and organization from previously studied systems. B. subtilis has been used as a model system for the characterization of cysteine desulfurases responsible for sulfur mobilization reactions in the biogenesis of Fe-S clusters and other sulfur-containing cofactors. Cysteine desulfurases catalyze the cleavage of the C-S bond from the amino acid cysteine and subsequent transfer of sulfur to acceptor molecules. These reactions can be monitored by the rate of alanine formation, the first product in the reaction, and sulfide formation, a byproduct of reactions performed under reducing conditions. The assembly of Fe-S clusters on protein scaffolds and the transfer of these clusters to target acceptors are determined through a combination of spectroscopic methods probing the rate of cluster assembly and transfer. This chapter provides a description of reactions promoting the assembly of Fe-S clusters in bacteria as well as methods used to study functions of each biosynthetic component and identify mechanistic differences employed by these enzymes across different pathways.
Collapse
|
83
|
Zohora F, Bidad K, Pourpak Z, Moin M. Biological and Immunological Aspects of Iron Deficiency Anemia in Cancer Development: A Narrative Review. Nutr Cancer 2018; 70:546-556. [PMID: 29697284 DOI: 10.1080/01635581.2018.1460685] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Iron Deficiency Anemia (IDA) is a universal health problem and a risk factor for the development of cancer. IDA changes the microenvironment of the human body by affecting both the biological and immunological systems. It increases DNA damage and genomic instability by different mechanisms. IDA is one of the leading causes of the imbalance between different antioxidant enzymes as well as enzymes involved in DNA damage and DNA repair systems of the body. It can affect the biogenesis/expression of microRNAs. IDA interrupts the oxidative phosphorylation energy metabolism and intestinal Cytochrome-P450 systems. It also disturbs multicellular signaling pathways involved in cell survival and helps in tumor angiogenesis. Moreover, IDA is also responsible for the functional deterioration of innate and adaptive immune systems that lead to immunological dysfunctions against invading pathogens. Genomic instability and immunological dysfunctions are the hallmarks of cancer development. In this review, we will review the evidence linking IDA to increased cancer risk.
Collapse
Affiliation(s)
- Fatema Zohora
- a Immunology, Asthma & Allergy Research Institute (IAARI), Tehran University of Medical Sciences (TUMS) , Tehran , Iran
| | - Katayoon Bidad
- a Immunology, Asthma & Allergy Research Institute (IAARI), Tehran University of Medical Sciences (TUMS) , Tehran , Iran
| | - Zahra Pourpak
- a Immunology, Asthma & Allergy Research Institute (IAARI), Tehran University of Medical Sciences (TUMS) , Tehran , Iran
| | - Mostafa Moin
- a Immunology, Asthma & Allergy Research Institute (IAARI), Tehran University of Medical Sciences (TUMS) , Tehran , Iran
| |
Collapse
|
84
|
The unique fold and lability of the [2Fe-2S] clusters of NEET proteins mediate their key functions in health and disease. J Biol Inorg Chem 2018; 23:599-612. [PMID: 29435647 PMCID: PMC6006223 DOI: 10.1007/s00775-018-1538-8] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2017] [Accepted: 01/26/2018] [Indexed: 02/08/2023]
Abstract
NEET proteins comprise a new class of [2Fe-2S] cluster proteins. In human, three genes encode for NEET proteins: cisd1 encodes mitoNEET (mNT), cisd2 encodes the Nutrient-deprivation autophagy factor-1 (NAF-1) and cisd3 encodes MiNT (Miner2). These recently discovered proteins play key roles in many processes related to normal metabolism and disease. Indeed, NEET proteins are involved in iron, Fe-S, and reactive oxygen homeostasis in cells and play an important role in regulating apoptosis and autophagy. mNT and NAF-1 are homodimeric and reside on the outer mitochondrial membrane. NAF-1 also resides in the membranes of the ER associated mitochondrial membranes (MAM) and the ER. MiNT is a monomer with distinct asymmetry in the molecular surfaces surrounding the clusters. Unlike its paralogs mNT and NAF-1, it resides within the mitochondria. NAF-1 and mNT share similar backbone folds to the plant homodimeric NEET protein (At-NEET), while MiNT's backbone fold resembles a bacterial MiNT protein. Despite the variation of amino acid composition among these proteins, all NEET proteins retained their unique CDGSH domain harboring their unique 3Cys:1His [2Fe-2S] cluster coordination through evolution. The coordinating exposed His was shown to convey the lability to the NEET proteins' [2Fe-2S] clusters. In this minireview, we discuss the NEET fold and its structural elements. Special attention is given to the unique lability of the NEETs' [2Fe-2S] cluster and the implication of the latter to the NEET proteins' cellular and systemic function in health and disease.
Collapse
|
85
|
Alfadhel M, Nashabat M, Alrifai MT, Alshaalan H, Al Mutairi F, Al-Shahrani SA, Plecko B, Almass R, Alsagob M, Almutairi FB, Al-Rumayyan A, Al-Twaijri W, Al-Owain M, Taylor RW, Kaya N. Further delineation of the phenotypic spectrum of ISCA2 defect: A report of ten new cases. Eur J Paediatr Neurol 2018; 22:46-55. [PMID: 29122497 DOI: 10.1016/j.ejpn.2017.10.003] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/03/2017] [Revised: 10/01/2017] [Accepted: 10/06/2017] [Indexed: 11/26/2022]
Abstract
Iron-Sulfur Cluster (ISC) biogenesis is a vital cellular process required to produce various ISC-containing proteins. These ISC proteins are responsible for essential functions such as glycine cleavage and the formation of lipoic acid, an essential cofactor of respiratory chain complexes. Defects in ISC biogenesis lead to multiple mitochondrial dysfunction syndromes including: ISCA2 with infantile onset leukodystrophy. Recently, a founder mutation, c.229G > A, p.Gly77Ser in ISCA2 was reported to cause Multiple Mitochondrial Dysfunction Syndrome type 4. In a retrospective review of children diagnosed with the ISCA2 defect, we were able to identify ten new patients who were not reported previously with the identical founder mutation. High CSF glycine levels and elevated glycine peaks on MR spectroscopy were demonstrated in all tested probands. All patients were between 3 and 7 months of age with a triad of neurodevelopmental regression, nystagmus and optic atrophy and leukodystrophy. MRI findings were typical in the patients with diffuse, abnormal white matter signal in the cerebrum, cerebellum, brain stem and spinal cord. The patients ended up in a vegetative state, and often premature death due to respiratory infections. We alert clinicians to consider the ISCA2 defect as a differential diagnosis of infantile onset leukodystrophies affecting the brain as well as the spinal cord, especially in the presence of elevated CSF glycine or elevated glycine peaks in MR spectroscopy.
Collapse
Affiliation(s)
- Majid Alfadhel
- Genetics Division, Department of Pediatrics, King Saud bin Abdulaziz University for Health Science, King Abdullah International Medical Research Centre, King Abdulaziz Medical City, Riyadh, Saudi Arabia.
| | - Marwan Nashabat
- Genetics Division, Department of Pediatrics, King Saud bin Abdulaziz University for Health Science, King Abdullah International Medical Research Centre, King Abdulaziz Medical City, Riyadh, Saudi Arabia
| | - Muhammad Talal Alrifai
- Neurology Division, Department of Pediatrics, King Saud bin Abdulaziz University for Health Science, King Abdullah International Medical Research Centre, King Abdulaziz Medical City, Riyadh, Saudi Arabia
| | - Hesham Alshaalan
- Medical Imaging Department, King Abdullah Specialized Children Hospital, King Saud bin Abdulaziz University for Health Sciences, King Abdulaziz Medical City, Ministry of National Guard-Health Affairs (NGHA), Riyadh, Saudi Arabia
| | - Fuad Al Mutairi
- Genetics Division, Department of Pediatrics, King Saud bin Abdulaziz University for Health Science, King Abdullah International Medical Research Centre, King Abdulaziz Medical City, Riyadh, Saudi Arabia
| | - Saif A Al-Shahrani
- Department of Medical Genetics, King Faisal Specialist Hospital and Research Center, Alfaisal University, Riyadh, Saudi Arabia
| | - Barbara Plecko
- Division of Child Neurology, University Children's Hospital, Zurich, Switzerland
| | - Rawan Almass
- Department of Genetics, King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia
| | - Maysoon Alsagob
- Department of Genetics, King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia
| | - Faten B Almutairi
- Department of Genetics, King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia
| | - Ahmed Al-Rumayyan
- Neurology Division, Department of Pediatrics, King Saud bin Abdulaziz University for Health Science, King Abdullah International Medical Research Centre, King Abdulaziz Medical City, Riyadh, Saudi Arabia
| | - Waleed Al-Twaijri
- Neurology Division, Department of Pediatrics, King Saud bin Abdulaziz University for Health Science, King Abdullah International Medical Research Centre, King Abdulaziz Medical City, Riyadh, Saudi Arabia
| | - Mohammed Al-Owain
- Department of Medical Genetics, King Faisal Specialist Hospital and Research Center, Alfaisal University, Riyadh, Saudi Arabia
| | - Robert W Taylor
- Wellcome Centre for Mitochondrial Research, Institute of Neuroscience, The Medical School, Newcastle University, Newcastle Upon Tyne, NE2 4HH, UK
| | - Namik Kaya
- Department of Genetics, King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia.
| |
Collapse
|
86
|
Bernardo-Castiñeira C, Valdés N, Sierra MI, Sáenz-de-Santa-María I, Bayón GF, Perez RF, Fernández AF, Fraga MF, Astudillo A, Menéndez R, Fernández B, Del Olmo M, Suarez C, Chiara MD. SDHC Promoter Methylation, a Novel Pathogenic Mechanism in Parasympathetic Paragangliomas. J Clin Endocrinol Metab 2018; 103:295-305. [PMID: 29126304 DOI: 10.1210/jc.2017-01702] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/31/2017] [Accepted: 10/31/2017] [Indexed: 02/07/2023]
Abstract
CONTEXT Germline mutations in the succinate dehydrogenase A, B, C, and D genes (collectively, SDHx) predispose to the development of paragangliomas (PGLs) arising at the parasympathetic or sympathetic neuroendocrine systems. SDHx mutations cause absence of tumoral immunostaining for SDHB. However, negative SDHB immunostaining has also been found in a subset of PGLs that lack SDHx mutations. SETTINGS Here, we report the comprehensive molecular characterization of one such a tumor of parasympathetic origin compared with healthy paraganglia and other PGLs with or without SDHx mutations. RESULTS Integration of multiplatform data revealed somatic SDHC methylation and loss of the 1q23.3 region containing the SDHC gene. This correlated with decreased SDHC messenger RNA (mRNA) and protein levels. Furthermore, another genetic event found affected the VHL gene, which showed a decreased DNA copy number, associated with low VHL mRNA levels, and an absence of VHL protein detected by immunohistochemistry. In addition, the tumor displayed a pseudohypoxic phenotype consisting in overexpression of the hypoxia-inducible factor (HIF)-1α and miR-210, as well as downregulation of the iron-sulfur cluster assembly enzyme (ISCU) involved in SDHB maturation. This profile resembles that of SDHx- or VHL-mutated PGLs but not of PGLs with decreased VHL copy number, pointing to SDHC rather than VHL as the pathogenic driver. CONCLUSIONS Collectively, these findings demonstrate the potential importance of both the SDHC epigenomic event and the activation of the HIF-1α/miR-210/ISCU axis in the pathogenesis of SDHx wild-type/SDHB-negative PGLs. To our knowledge, this is the first case of a sporadic parasympathetic PGL that carries silencing of SDHC, fulfilling the two-hit Knudson's model for tumorigenesis.
Collapse
Affiliation(s)
- Cristóbal Bernardo-Castiñeira
- Head and Neck Oncology Laboratory, Institute of Sanitary Research of Asturias (ISPA), Hospital Universitario Central de Asturias, Institute of Oncology of Asturias (IUOPA), CIBERONC, Oviedo, Spain
| | - Nuria Valdés
- Service of Endocrinology and Nutrition, Hospital Universitario Central de Asturias, Oviedo, Spain
| | - Marta I Sierra
- Cancer Epigenetics Laboratory, Institute of Oncology of Asturias (IUOPA), Institute of Sanitary Research of Asturias (ISPA), Hospital Universitario Central de Asturias, Universidad de Oviedo, Oviedo, Spain
| | - Inés Sáenz-de-Santa-María
- Head and Neck Oncology Laboratory, Institute of Sanitary Research of Asturias (ISPA), Hospital Universitario Central de Asturias, Institute of Oncology of Asturias (IUOPA), CIBERONC, Oviedo, Spain
| | - Gustavo F Bayón
- Cancer Epigenetics Laboratory, Institute of Oncology of Asturias (IUOPA), Institute of Sanitary Research of Asturias (ISPA), Hospital Universitario Central de Asturias, Universidad de Oviedo, Oviedo, Spain
| | - Raúl F Perez
- Cancer Epigenetics Laboratory, Institute of Oncology of Asturias (IUOPA), Institute of Sanitary Research of Asturias (ISPA), Hospital Universitario Central de Asturias, Universidad de Oviedo, Oviedo, Spain
- Nanomaterials and Nanotechnology Research Center (CINN-CSIC), Universidad de Oviedo-Principado de Asturias, Oviedo, Spain
| | - Agustín F Fernández
- Cancer Epigenetics Laboratory, Institute of Oncology of Asturias (IUOPA), Institute of Sanitary Research of Asturias (ISPA), Hospital Universitario Central de Asturias, Universidad de Oviedo, Oviedo, Spain
| | - Mario F Fraga
- Nanomaterials and Nanotechnology Research Center (CINN-CSIC), Universidad de Oviedo-Principado de Asturias, Oviedo, Spain
| | - Aurora Astudillo
- Service of Pathology, Hospital Universitario Central de Asturias, Oviedo, Spain
| | - Rafael Menéndez
- Service of Radiology, Hospital Universitario Central de Asturias, Oviedo, Spain
| | - Belén Fernández
- Service of Nuclear Medicine, Hospital Universitario Central de Asturias, Oviedo, Spain
| | - Maribel Del Olmo
- Service of Endocrinology and Nutrition, Hospital Universitario la Fé, Valencia, Spain
| | - Carlos Suarez
- Head and Neck Oncology Laboratory, Institute of Sanitary Research of Asturias (ISPA), Hospital Universitario Central de Asturias, Institute of Oncology of Asturias (IUOPA), CIBERONC, Oviedo, Spain
| | - María-Dolores Chiara
- Head and Neck Oncology Laboratory, Institute of Sanitary Research of Asturias (ISPA), Hospital Universitario Central de Asturias, Institute of Oncology of Asturias (IUOPA), CIBERONC, Oviedo, Spain
| |
Collapse
|
87
|
Ranchoux B, Harvey LD, Ayon RJ, Babicheva A, Bonnet S, Chan SY, Yuan JXJ, Perez VDJ. Endothelial dysfunction in pulmonary arterial hypertension: an evolving landscape (2017 Grover Conference Series). Pulm Circ 2018; 8:2045893217752912. [PMID: 29283043 PMCID: PMC5798691 DOI: 10.1177/2045893217752912] [Citation(s) in RCA: 105] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/23/2017] [Accepted: 12/18/2017] [Indexed: 02/06/2023] Open
Abstract
Endothelial dysfunction is a major player in the development and progression of vascular pathology in pulmonary arterial hypertension (PAH), a disease associated with small vessel loss and obstructive vasculopathy that leads to increased pulmonary vascular resistance, subsequent right heart failure, and premature death. Over the past ten years, there has been tremendous progress in our understanding of pulmonary endothelial biology as it pertains to the genetic and molecular mechanisms that orchestrate the endothelial response to direct or indirect injury, and how their dysregulation can contribute to the pathogenesis of PAH. As one of the major topics included in the 2017 Grover Conference Series, discussion centered on recent developments in four areas of pulmonary endothelial biology: (1) angiogenesis; (2) endothelial-mesenchymal transition (EndMT); (3) epigenetics; and (4) biology of voltage-gated ion channels. The present review will summarize the content of these discussions and provide a perspective on the most promising aspects of endothelial dysfunction that may be amenable for therapeutic development.
Collapse
Affiliation(s)
| | - Lloyd D. Harvey
- University of Pittsburgh Vascular Medicine Institute Division of Cardiology, Pittsburgh, PA, USA
| | - Ramon J. Ayon
- Division of Translational and Regenerative Medicine, The University of Arizona College of Medicine, Tucson, AZ, USA
| | - Aleksandra Babicheva
- Division of Translational and Regenerative Medicine, The University of Arizona College of Medicine, Tucson, AZ, USA
| | | | - Stephen Y. Chan
- University of Pittsburgh Vascular Medicine Institute Division of Cardiology, Pittsburgh, PA, USA
| | - Jason X.-J. Yuan
- Division of Translational and Regenerative Medicine, The University of Arizona College of Medicine, Tucson, AZ, USA
| | - Vinicio de Jesus Perez
- Division of Pulmonary and Critical Care Medicine, Stanford University Medical Center, Stanford, CA, USA
- The Vera Moulton Wall Center for Pulmonary Vascular Medicine, Stanford University Medical Center, Stanford, CA, USA
- Stanford Cardiovascular Institute, Stanford University Medical Center, Stanford, CA, USA
| |
Collapse
|
88
|
Hu H, Ding Y, Wang Y, Geng S, Liu J, He J, Lu Y, Li X, Yuan M, Zhu S, Zhao S. MitoK ATP channels promote the proliferation of hypoxic human pulmonary artery smooth muscle cells via the ROS/HIF/miR-210/ISCU signaling pathway. Exp Ther Med 2017; 14:6105-6112. [PMID: 29285165 DOI: 10.3892/etm.2017.5322] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2016] [Accepted: 05/16/2017] [Indexed: 12/18/2022] Open
Abstract
Previous results have indicated that mitochondrial ATP-sensitive potassium (mitoKATP) channels are associated with the hypoxic proliferation of pulmonary artery smooth muscle cells (PASMCs). However, the mechanism underlying the promotive effects of mitoKATP channels on cell proliferation in response to hypoxia remains unknown. mitoKATP channel opening results in a collapse of mitochondrial membrane potential and generation of mitochondrial reactive oxygen species (ROS). As hypoxia-inducible factor-1α (HIF-1α) is a critical oxygen sensor and major transcriptional regulator of the hypoxic adaptive response, the current study assessed whether mitoKATP opening contributes to the chronic proliferation of human PASMCs (hPASMCs) in collaboration with HIF-1α and its downstream targets under hypoxic conditions. The present study demonstrated that there was crosstalk between mitoKATP channels and HIF-1α signaling in PASMCs under hypoxic conditions. The results suggest that mitoKATP channels are involved in the proliferation of PASMCs during hypoxia through upregulation of the ROS/HIF/microRNA-210/iron-sulfur cluster protein signaling pathway.
Collapse
Affiliation(s)
- Hongling Hu
- Department of Respiratory Medicine, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430014, P.R. China
| | - Yu Ding
- Key Laboratory for Molecular Diagnosis of Hubei, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430014, P.R. China.,Central Laboratory, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430014, P.R. China
| | - Yang Wang
- Department of Respiratory Medicine, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430014, P.R. China
| | - Shuang Geng
- Department of Respiratory Medicine, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430014, P.R. China
| | - Jue Liu
- Department of Clinical Pharmacy, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430014, P.R. China
| | - Jinrong He
- Key Laboratory for Molecular Diagnosis of Hubei, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430014, P.R. China.,Central Laboratory, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430014, P.R. China
| | - Yang Lu
- Department of Respiratory Medicine, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430014, P.R. China
| | - Xueying Li
- Department of Respiratory Medicine, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430014, P.R. China
| | - Mingli Yuan
- Department of Respiratory Medicine, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430014, P.R. China
| | - Shan Zhu
- Department of Respiratory Medicine, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430014, P.R. China
| | - Su Zhao
- Department of Respiratory Medicine, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430014, P.R. China
| |
Collapse
|
89
|
Ishiyama A, Sakai C, Matsushima Y, Noguchi S, Mitsuhashi S, Endo Y, Hayashi YK, Saito Y, Nakagawa E, Komaki H, Sugai K, Sasaki M, Sato N, Nonaka I, Goto YI, Nishino I. IBA57 mutations abrogate iron-sulfur cluster assembly leading to cavitating leukoencephalopathy. NEUROLOGY-GENETICS 2017; 3:e184. [PMID: 28913435 PMCID: PMC5591399 DOI: 10.1212/nxg.0000000000000184] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/09/2017] [Accepted: 07/27/2017] [Indexed: 12/30/2022]
Abstract
OBJECTIVE To determine the molecular factors contributing to progressive cavitating leukoencephalopathy (PCL) to help resolve the underlying genotype-phenotype associations in the mitochondrial iron-sulfur cluster (ISC) assembly system. METHODS The subjects were 3 patients from 2 families who showed no inconsistencies in either clinical or brain MRI findings as PCL. We used exome sequencing, immunoblotting, and enzyme activity assays to establish a molecular diagnosis and determine the roles of ISC-associated factors in PCL. RESULTS We performed genetic analyses on these 3 patients and identified compound heterozygosity for the IBA57 gene, which encodes the mitochondrial iron-sulfur protein assembly factor. Protein expression analysis revealed substantial decreases in IBA57 protein expression in myoblasts and fibroblasts. Immunoblotting revealed substantially reduced expression of SDHB, a subunit of complex II, and lipoic acid synthetase (LIAS). Levels of pyruvate dehydrogenase complex-E2 and α-ketoglutarate dehydrogenase-E2, which use lipoic acid as a cofactor, were also reduced. In activity staining, SDH activity was clearly reduced, but it was ameliorated in mitochondrial fractions from rescued myoblasts. In addition, NFU1 protein expression was also decreased, which is required for the assembly of a subset of iron-sulfur proteins to SDH and LIAS in the mitochondrial ISC assembly system. CONCLUSIONS Defects in IBA57 essentially regulate NFU1 expression, and aberrant NFU1 ultimately affects SDH activity and LIAS expression in the ISC biogenesis pathway. This study provides new insights into the role of the iron-sulfur protein assembly system in disorders related to mitochondrial energy metabolism associated with leukoencephalopathy with cavities.
Collapse
Affiliation(s)
- Akihiko Ishiyama
- Department of Child Neurology (A.I., Y.S., E.N., H.K, K.S., M.S.), National Center Hospital; Department of Neuromuscular Research (A.I., S.N., S.M., Y.E., Y.K.H., I. Nonaka, I. Nishino.), National Institute of Neuroscience; Department of Mental Retardation and Birth Defect Research (C.S., Y.M., Y.-i.G.), National Institute of Neuroscience; Department of Radiology (N.S.), National Center Hospital, National Center of Neurology and Psychiatry, Tokyo; Department of Pharmacology (A.I.), Interdisciplinary Graduate School of Medicine and Engineering, University of Yamanashi; and Department of Pathophysiology (Y.K.H), Tokyo Medical University, Japan
| | - Chika Sakai
- Department of Child Neurology (A.I., Y.S., E.N., H.K, K.S., M.S.), National Center Hospital; Department of Neuromuscular Research (A.I., S.N., S.M., Y.E., Y.K.H., I. Nonaka, I. Nishino.), National Institute of Neuroscience; Department of Mental Retardation and Birth Defect Research (C.S., Y.M., Y.-i.G.), National Institute of Neuroscience; Department of Radiology (N.S.), National Center Hospital, National Center of Neurology and Psychiatry, Tokyo; Department of Pharmacology (A.I.), Interdisciplinary Graduate School of Medicine and Engineering, University of Yamanashi; and Department of Pathophysiology (Y.K.H), Tokyo Medical University, Japan
| | - Yuichi Matsushima
- Department of Child Neurology (A.I., Y.S., E.N., H.K, K.S., M.S.), National Center Hospital; Department of Neuromuscular Research (A.I., S.N., S.M., Y.E., Y.K.H., I. Nonaka, I. Nishino.), National Institute of Neuroscience; Department of Mental Retardation and Birth Defect Research (C.S., Y.M., Y.-i.G.), National Institute of Neuroscience; Department of Radiology (N.S.), National Center Hospital, National Center of Neurology and Psychiatry, Tokyo; Department of Pharmacology (A.I.), Interdisciplinary Graduate School of Medicine and Engineering, University of Yamanashi; and Department of Pathophysiology (Y.K.H), Tokyo Medical University, Japan
| | - Satoru Noguchi
- Department of Child Neurology (A.I., Y.S., E.N., H.K, K.S., M.S.), National Center Hospital; Department of Neuromuscular Research (A.I., S.N., S.M., Y.E., Y.K.H., I. Nonaka, I. Nishino.), National Institute of Neuroscience; Department of Mental Retardation and Birth Defect Research (C.S., Y.M., Y.-i.G.), National Institute of Neuroscience; Department of Radiology (N.S.), National Center Hospital, National Center of Neurology and Psychiatry, Tokyo; Department of Pharmacology (A.I.), Interdisciplinary Graduate School of Medicine and Engineering, University of Yamanashi; and Department of Pathophysiology (Y.K.H), Tokyo Medical University, Japan
| | - Satomi Mitsuhashi
- Department of Child Neurology (A.I., Y.S., E.N., H.K, K.S., M.S.), National Center Hospital; Department of Neuromuscular Research (A.I., S.N., S.M., Y.E., Y.K.H., I. Nonaka, I. Nishino.), National Institute of Neuroscience; Department of Mental Retardation and Birth Defect Research (C.S., Y.M., Y.-i.G.), National Institute of Neuroscience; Department of Radiology (N.S.), National Center Hospital, National Center of Neurology and Psychiatry, Tokyo; Department of Pharmacology (A.I.), Interdisciplinary Graduate School of Medicine and Engineering, University of Yamanashi; and Department of Pathophysiology (Y.K.H), Tokyo Medical University, Japan
| | - Yukari Endo
- Department of Child Neurology (A.I., Y.S., E.N., H.K, K.S., M.S.), National Center Hospital; Department of Neuromuscular Research (A.I., S.N., S.M., Y.E., Y.K.H., I. Nonaka, I. Nishino.), National Institute of Neuroscience; Department of Mental Retardation and Birth Defect Research (C.S., Y.M., Y.-i.G.), National Institute of Neuroscience; Department of Radiology (N.S.), National Center Hospital, National Center of Neurology and Psychiatry, Tokyo; Department of Pharmacology (A.I.), Interdisciplinary Graduate School of Medicine and Engineering, University of Yamanashi; and Department of Pathophysiology (Y.K.H), Tokyo Medical University, Japan
| | - Yukiko K Hayashi
- Department of Child Neurology (A.I., Y.S., E.N., H.K, K.S., M.S.), National Center Hospital; Department of Neuromuscular Research (A.I., S.N., S.M., Y.E., Y.K.H., I. Nonaka, I. Nishino.), National Institute of Neuroscience; Department of Mental Retardation and Birth Defect Research (C.S., Y.M., Y.-i.G.), National Institute of Neuroscience; Department of Radiology (N.S.), National Center Hospital, National Center of Neurology and Psychiatry, Tokyo; Department of Pharmacology (A.I.), Interdisciplinary Graduate School of Medicine and Engineering, University of Yamanashi; and Department of Pathophysiology (Y.K.H), Tokyo Medical University, Japan
| | - Yoshiaki Saito
- Department of Child Neurology (A.I., Y.S., E.N., H.K, K.S., M.S.), National Center Hospital; Department of Neuromuscular Research (A.I., S.N., S.M., Y.E., Y.K.H., I. Nonaka, I. Nishino.), National Institute of Neuroscience; Department of Mental Retardation and Birth Defect Research (C.S., Y.M., Y.-i.G.), National Institute of Neuroscience; Department of Radiology (N.S.), National Center Hospital, National Center of Neurology and Psychiatry, Tokyo; Department of Pharmacology (A.I.), Interdisciplinary Graduate School of Medicine and Engineering, University of Yamanashi; and Department of Pathophysiology (Y.K.H), Tokyo Medical University, Japan
| | - Eiji Nakagawa
- Department of Child Neurology (A.I., Y.S., E.N., H.K, K.S., M.S.), National Center Hospital; Department of Neuromuscular Research (A.I., S.N., S.M., Y.E., Y.K.H., I. Nonaka, I. Nishino.), National Institute of Neuroscience; Department of Mental Retardation and Birth Defect Research (C.S., Y.M., Y.-i.G.), National Institute of Neuroscience; Department of Radiology (N.S.), National Center Hospital, National Center of Neurology and Psychiatry, Tokyo; Department of Pharmacology (A.I.), Interdisciplinary Graduate School of Medicine and Engineering, University of Yamanashi; and Department of Pathophysiology (Y.K.H), Tokyo Medical University, Japan
| | - Hirofumi Komaki
- Department of Child Neurology (A.I., Y.S., E.N., H.K, K.S., M.S.), National Center Hospital; Department of Neuromuscular Research (A.I., S.N., S.M., Y.E., Y.K.H., I. Nonaka, I. Nishino.), National Institute of Neuroscience; Department of Mental Retardation and Birth Defect Research (C.S., Y.M., Y.-i.G.), National Institute of Neuroscience; Department of Radiology (N.S.), National Center Hospital, National Center of Neurology and Psychiatry, Tokyo; Department of Pharmacology (A.I.), Interdisciplinary Graduate School of Medicine and Engineering, University of Yamanashi; and Department of Pathophysiology (Y.K.H), Tokyo Medical University, Japan
| | - Kenji Sugai
- Department of Child Neurology (A.I., Y.S., E.N., H.K, K.S., M.S.), National Center Hospital; Department of Neuromuscular Research (A.I., S.N., S.M., Y.E., Y.K.H., I. Nonaka, I. Nishino.), National Institute of Neuroscience; Department of Mental Retardation and Birth Defect Research (C.S., Y.M., Y.-i.G.), National Institute of Neuroscience; Department of Radiology (N.S.), National Center Hospital, National Center of Neurology and Psychiatry, Tokyo; Department of Pharmacology (A.I.), Interdisciplinary Graduate School of Medicine and Engineering, University of Yamanashi; and Department of Pathophysiology (Y.K.H), Tokyo Medical University, Japan
| | - Masayuki Sasaki
- Department of Child Neurology (A.I., Y.S., E.N., H.K, K.S., M.S.), National Center Hospital; Department of Neuromuscular Research (A.I., S.N., S.M., Y.E., Y.K.H., I. Nonaka, I. Nishino.), National Institute of Neuroscience; Department of Mental Retardation and Birth Defect Research (C.S., Y.M., Y.-i.G.), National Institute of Neuroscience; Department of Radiology (N.S.), National Center Hospital, National Center of Neurology and Psychiatry, Tokyo; Department of Pharmacology (A.I.), Interdisciplinary Graduate School of Medicine and Engineering, University of Yamanashi; and Department of Pathophysiology (Y.K.H), Tokyo Medical University, Japan
| | - Noriko Sato
- Department of Child Neurology (A.I., Y.S., E.N., H.K, K.S., M.S.), National Center Hospital; Department of Neuromuscular Research (A.I., S.N., S.M., Y.E., Y.K.H., I. Nonaka, I. Nishino.), National Institute of Neuroscience; Department of Mental Retardation and Birth Defect Research (C.S., Y.M., Y.-i.G.), National Institute of Neuroscience; Department of Radiology (N.S.), National Center Hospital, National Center of Neurology and Psychiatry, Tokyo; Department of Pharmacology (A.I.), Interdisciplinary Graduate School of Medicine and Engineering, University of Yamanashi; and Department of Pathophysiology (Y.K.H), Tokyo Medical University, Japan
| | - Ikuya Nonaka
- Department of Child Neurology (A.I., Y.S., E.N., H.K, K.S., M.S.), National Center Hospital; Department of Neuromuscular Research (A.I., S.N., S.M., Y.E., Y.K.H., I. Nonaka, I. Nishino.), National Institute of Neuroscience; Department of Mental Retardation and Birth Defect Research (C.S., Y.M., Y.-i.G.), National Institute of Neuroscience; Department of Radiology (N.S.), National Center Hospital, National Center of Neurology and Psychiatry, Tokyo; Department of Pharmacology (A.I.), Interdisciplinary Graduate School of Medicine and Engineering, University of Yamanashi; and Department of Pathophysiology (Y.K.H), Tokyo Medical University, Japan
| | - Yu-Ichi Goto
- Department of Child Neurology (A.I., Y.S., E.N., H.K, K.S., M.S.), National Center Hospital; Department of Neuromuscular Research (A.I., S.N., S.M., Y.E., Y.K.H., I. Nonaka, I. Nishino.), National Institute of Neuroscience; Department of Mental Retardation and Birth Defect Research (C.S., Y.M., Y.-i.G.), National Institute of Neuroscience; Department of Radiology (N.S.), National Center Hospital, National Center of Neurology and Psychiatry, Tokyo; Department of Pharmacology (A.I.), Interdisciplinary Graduate School of Medicine and Engineering, University of Yamanashi; and Department of Pathophysiology (Y.K.H), Tokyo Medical University, Japan
| | - Ichizo Nishino
- Department of Child Neurology (A.I., Y.S., E.N., H.K, K.S., M.S.), National Center Hospital; Department of Neuromuscular Research (A.I., S.N., S.M., Y.E., Y.K.H., I. Nonaka, I. Nishino.), National Institute of Neuroscience; Department of Mental Retardation and Birth Defect Research (C.S., Y.M., Y.-i.G.), National Institute of Neuroscience; Department of Radiology (N.S.), National Center Hospital, National Center of Neurology and Psychiatry, Tokyo; Department of Pharmacology (A.I.), Interdisciplinary Graduate School of Medicine and Engineering, University of Yamanashi; and Department of Pathophysiology (Y.K.H), Tokyo Medical University, Japan
| |
Collapse
|
90
|
Abstract
Hypoxia and dysregulated metabolism are defining features of solid tumors. How cancer cells adapt to low O2 has been illuminated by numerous studies, with "reprogrammed" metabolism being one of the most important mechanisms. This metabolic reprogramming not only promotes cancer cell plasticity, but also provides novel insights for treatment strategies. As the most studied O2 "sensor," hypoxia-inducible factor (HIF) is regarded as an important regulator of hypoxia-induced transcriptional responses. This minireview will summarize our current understanding of hypoxia-induced changes in cancer cell metabolism, with an initial focus on HIF-mediated effects, and will highlight how these metabolic alterations affect malignant phenotypes.
Collapse
Affiliation(s)
- Hong Xie
- From the Abramson Family Cancer Research Institute and.,Departments of Cancer Biology and
| | - M Celeste Simon
- From the Abramson Family Cancer Research Institute and .,Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104
| |
Collapse
|
91
|
|
92
|
Habarou F, Hamel Y, Haack TB, Feichtinger RG, Lebigot E, Marquardt I, Busiah K, Laroche C, Madrange M, Grisel C, Pontoizeau C, Eisermann M, Boutron A, Chrétien D, Chadefaux-Vekemans B, Barouki R, Bole-Feysot C, Nitschke P, Goudin N, Boddaert N, Nemazanyy I, Delahodde A, Kölker S, Rodenburg RJ, Korenke GC, Meitinger T, Strom TM, Prokisch H, Rotig A, Ottolenghi C, Mayr JA, de Lonlay P. Biallelic Mutations in LIPT2 Cause a Mitochondrial Lipoylation Defect Associated with Severe Neonatal Encephalopathy. Am J Hum Genet 2017; 101:283-290. [PMID: 28757203 DOI: 10.1016/j.ajhg.2017.07.001] [Citation(s) in RCA: 57] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2017] [Accepted: 07/05/2017] [Indexed: 11/25/2022] Open
Abstract
Lipoate serves as a cofactor for the glycine cleavage system (GCS) and four 2-oxoacid dehydrogenases functioning in energy metabolism (α-oxoglutarate dehydrogenase [α-KGDHc] and pyruvate dehydrogenase [PDHc]), or amino acid metabolism (branched-chain oxoacid dehydrogenase, 2-oxoadipate dehydrogenase). Mitochondrial lipoate synthesis involves three enzymatic steps catalyzed sequentially by lipoyl(octanoyl) transferase 2 (LIPT2), lipoic acid synthetase (LIAS), and lipoyltransferase 1 (LIPT1). Mutations in LIAS have been associated with nonketotic hyperglycinemia-like early-onset convulsions and encephalopathy combined with a defect in mitochondrial energy metabolism. LIPT1 deficiency spares GCS deficiency and has been associated with a biochemical signature of combined 2-oxoacid dehydrogenase deficiency leading to early death or Leigh-like encephalopathy. We report on the identification of biallelic LIPT2 mutations in three affected individuals from two families with severe neonatal encephalopathy. Brain MRI showed major cortical atrophy with white matter abnormalities and cysts. Plasma glycine was mildly increased. Affected individuals' fibroblasts showed reduced oxygen consumption rates, PDHc, α-KGDHc activities, leucine catabolic flux, and decreased protein lipoylation. A normalization of lipoylation was observed after expression of wild-type LIPT2, arguing for LIPT2 requirement in intramitochondrial lipoate synthesis. Lipoic acid supplementation did not improve clinical condition nor activities of PDHc, α-KGDHc, or leucine metabolism in fibroblasts and was ineffective in yeast deleted for the orthologous LIP2.
Collapse
|
93
|
Imam MU, Zhang S, Ma J, Wang H, Wang F. Antioxidants Mediate Both Iron Homeostasis and Oxidative Stress. Nutrients 2017; 9:E671. [PMID: 28657578 PMCID: PMC5537786 DOI: 10.3390/nu9070671] [Citation(s) in RCA: 143] [Impact Index Per Article: 17.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2017] [Revised: 06/20/2017] [Accepted: 06/22/2017] [Indexed: 12/18/2022] Open
Abstract
Oxidative stress is a common denominator in the pathogenesis of many chronic diseases. Therefore, antioxidants are often used to protect cells and tissues and reverse oxidative damage. It is well known that iron metabolism underlies the dynamic interplay between oxidative stress and antioxidants in many pathophysiological processes. Both iron deficiency and iron overload can affect redox state, and these conditions can be restored to physiological conditions using iron supplementation and iron chelation, respectively. Similarly, the addition of antioxidants to these treatment regimens has been suggested as a viable therapeutic approach for attenuating tissue damage induced by oxidative stress. Notably, many bioactive plant-derived compounds have been shown to regulate both iron metabolism and redox state, possibly through interactive mechanisms. This review summarizes our current understanding of these mechanisms and discusses compelling preclinical evidence that bioactive plant-derived compounds can be both safe and effective for managing both iron deficiency and iron overload conditions.
Collapse
Affiliation(s)
- Mustapha Umar Imam
- Department of Nutrition, Precision Nutrition Innovation Center, School of Public Health, Zhengzhou University, Zhengzhou 450001, China.
| | - Shenshen Zhang
- Department of Nutrition, Precision Nutrition Innovation Center, School of Public Health, Zhengzhou University, Zhengzhou 450001, China.
| | - Jifei Ma
- Department of Nutrition, Precision Nutrition Innovation Center, School of Public Health, Zhengzhou University, Zhengzhou 450001, China.
| | - Hao Wang
- Department of Nutrition, Precision Nutrition Innovation Center, School of Public Health, Zhengzhou University, Zhengzhou 450001, China.
- Department of Nutrition, Nutrition Discovery Innovation Center, School of Public Health, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, School of Medicine, Zhejiang University, Hangzhou 310058, China.
| | - Fudi Wang
- Department of Nutrition, Precision Nutrition Innovation Center, School of Public Health, Zhengzhou University, Zhengzhou 450001, China.
- Department of Nutrition, Nutrition Discovery Innovation Center, School of Public Health, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, School of Medicine, Zhejiang University, Hangzhou 310058, China.
| |
Collapse
|
94
|
Ali MK, Kim RY, Karim R, Mayall JR, Martin KL, Shahandeh A, Abbasian F, Starkey MR, Loustaud-Ratti V, Johnstone D, Milward EA, Hansbro PM, Horvat JC. Role of iron in the pathogenesis of respiratory disease. Int J Biochem Cell Biol 2017; 88:181-195. [PMID: 28495571 DOI: 10.1016/j.biocel.2017.05.003] [Citation(s) in RCA: 70] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2016] [Revised: 05/01/2017] [Accepted: 05/03/2017] [Indexed: 12/13/2022]
Abstract
Iron is essential for many biological processes, however, too much or too little iron can result in a wide variety of pathological consequences, depending on the organ system, tissue or cell type affected. In order to reduce pathogenesis, iron levels are tightly controlled in throughout the body by regulatory systems that control iron absorption, systemic transport and cellular uptake and storage. Altered iron levels and/or dysregulated homeostasis have been associated with several lung diseases, including chronic obstructive pulmonary disease, lung cancer, cystic fibrosis, idiopathic pulmonary fibrosis and asthma. However, the mechanisms that underpin these associations and whether iron plays a key role in the pathogenesis of lung disease are yet to be fully elucidated. Furthermore, in order to survive and replicate, pathogenic micro-organisms have evolved strategies to source host iron, including freeing iron from cells and proteins that store and transport iron. To counter these microbial strategies, mammals have evolved immune-mediated defence mechanisms that reduce iron availability to pathogens. This interplay between iron, infection and immunity has important ramifications for the pathogenesis and management of human respiratory infections and diseases. An increased understanding of the role that iron plays in the pathogenesis of lung disease and respiratory infections may help inform novel therapeutic strategies. Here we review the clinical and experimental evidence that highlights the potential importance of iron in respiratory diseases and infections.
Collapse
Affiliation(s)
- Md Khadem Ali
- School of Biomedical Sciences and Pharmacy, Faculty of Health and Medicine, The University of Newcastle, Callaghan NSW 2308, Australia
| | - Richard Y Kim
- School of Biomedical Sciences and Pharmacy, Faculty of Health and Medicine, The University of Newcastle, Callaghan NSW 2308, Australia
| | - Rafia Karim
- School of Biomedical Sciences and Pharmacy, Faculty of Health and Medicine, The University of Newcastle, Callaghan NSW 2308, Australia
| | - Jemma R Mayall
- School of Biomedical Sciences and Pharmacy, Faculty of Health and Medicine, The University of Newcastle, Callaghan NSW 2308, Australia
| | - Kristy L Martin
- School of Biomedical Sciences and Pharmacy, Faculty of Health and Medicine, The University of Newcastle, Callaghan NSW 2308, Australia
| | - Ali Shahandeh
- School of Biomedical Sciences and Pharmacy, Faculty of Health and Medicine, The University of Newcastle, Callaghan NSW 2308, Australia
| | - Firouz Abbasian
- Global Centre for Environmental Remediation, Faculty of Science, the University of Newcastle, Callaghan, NSW 2308, Australia
| | - Malcolm R Starkey
- School of Biomedical Sciences and Pharmacy, Faculty of Health and Medicine, The University of Newcastle, Callaghan NSW 2308, Australia
| | | | - Daniel Johnstone
- Bosch Institute and Discipline of Physiology, The University of Sydney, Sydney NSW 2000, Australia
| | - Elizabeth A Milward
- School of Biomedical Sciences and Pharmacy, Faculty of Health and Medicine, The University of Newcastle, Callaghan NSW 2308, Australia
| | - Philip M Hansbro
- School of Biomedical Sciences and Pharmacy, Faculty of Health and Medicine, The University of Newcastle, Callaghan NSW 2308, Australia
| | - Jay C Horvat
- School of Biomedical Sciences and Pharmacy, Faculty of Health and Medicine, The University of Newcastle, Callaghan NSW 2308, Australia.
| |
Collapse
|
95
|
Bertero T, Rezzonico R, Pottier N, Mari B. Impact of MicroRNAs in the Cellular Response to Hypoxia. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2017; 333:91-158. [PMID: 28729029 DOI: 10.1016/bs.ircmb.2017.03.006] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
In mammalian cells, hypoxia, or inadequate oxygen availability, regulates the expression of a specific set of MicroRNAs (MiRNAs), termed "hypoxamiRs." Over the past 10 years, the appreciation of the importance of hypoxamiRs in regulating the cellular adaptation to hypoxia has grown dramatically. At the cellular level, each hypoxamiR, including the master hypoxamiR MiR-210, can simultaneously regulate expression of multiple target genes in order to fine-tune the adaptive response of cells to hypoxia. This review addresses the complex molecular regulation of MiRNAs in both physiological and pathological conditions of low oxygen adaptation and the multiple functions of hypoxamiRs in various hypoxia-associated biological processes, including apoptosis, survival, proliferation, angiogenesis, inflammation, and metabolism. From a clinical perspective, we also discuss the potential use of hypoxamiRs as new biomarkers and/or therapeutic targets in cancer and aging-associated diseases including cardiovascular and fibroproliferative disorders.
Collapse
Affiliation(s)
- Thomas Bertero
- Université Côte d'Azur, CNRS, INSERM, IRCAN, FHU-OncoAge, Nice, France
| | - Roger Rezzonico
- Université Côte d'Azur, CNRS, IPMC, FHU-OncoAge, Sophia-Antipolis, France
| | | | - Bernard Mari
- Université Côte d'Azur, CNRS, IPMC, FHU-OncoAge, Sophia-Antipolis, France.
| |
Collapse
|
96
|
Harvey LD, Chan SY. Emerging Metabolic Therapies in Pulmonary Arterial Hypertension. J Clin Med 2017; 6:jcm6040043. [PMID: 28375184 PMCID: PMC5406775 DOI: 10.3390/jcm6040043] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2017] [Revised: 03/28/2017] [Accepted: 03/29/2017] [Indexed: 12/26/2022] Open
Abstract
Pulmonary hypertension (PH) is an enigmatic vascular disorder characterized by pulmonary vascular remodeling and increased pulmonary vascular resistance, ultimately resulting in pressure overload, dysfunction, and failure of the right ventricle. Current medications for PH do not reverse or prevent disease progression, and current diagnostic strategies are suboptimal for detecting early-stage disease. Thus, there is a substantial need to develop new diagnostics and therapies that target the molecular origins of PH. Emerging investigations have defined metabolic aberrations as fundamental and early components of disease manifestation in both pulmonary vasculature and the right ventricle. As such, the elucidation of metabolic dysregulation in pulmonary hypertension allows for greater therapeutic insight into preventing, halting, or even reversing disease progression. This review will aim to discuss (1) the reprogramming and dysregulation of metabolic pathways in pulmonary hypertension; (2) the emerging therapeutic interventions targeting these metabolic pathways; and (3) further innovation needed to overcome barriers in the treatment of this devastating disease.
Collapse
Affiliation(s)
- Lloyd D Harvey
- Medical Scientist Training Program, University of Pittsburgh School of Medicine and University of Pittsburgh Medical Center, Pittsburgh, PA 15213, USA.
| | - Stephen Y Chan
- Division of Cardiology, Center for Pulmonary Vascular Biology and Medicine, Pittsburgh Heart, Lung, Blood, and Vascular Medicine Institute, University of Pittsburgh School of Medicine and University of Pittsburgh Medical Center, Pittsburgh, PA 15213, USA.
| |
Collapse
|
97
|
Zuo J, Wu Z, Li Y, Shen Z, Feng X, Zhang M, Ye H. Mitochondrial ABC Transporter ATM3 Is Essential for Cytosolic Iron-Sulfur Cluster Assembly. PLANT PHYSIOLOGY 2017; 173:2096-2109. [PMID: 28250070 PMCID: PMC5373059 DOI: 10.1104/pp.16.01760] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/16/2016] [Accepted: 02/28/2017] [Indexed: 05/18/2023]
Abstract
The mitochondrial ATP-binding cassette transporter ATM3 has been studied in Arabidopsis. Its function, however, is poorly understood in other model plant species. This study reports that the ATM3 is required for cytosolic iron-sulfur cluster assembly and is essential for meristem maintenance in rice (Oryza sativa). The loss of function of OsATM3 is lethal in rice at the four-leaf stage. In the osatm3 T-DNA insertion mutant, the fourth leaf fails to develop and the lateral roots are short. Cytosolic iron-sulfur protein activities were significantly reduced in both osatm3 and RNA interference transgenic lines. The expression profiles of many iron metabolism genes were altered in the osatm3 and RNA interference lines. Glutathione metabolism was impaired and reactive oxygen species, particularly superoxide, accumulated in osatm3 Promoter-β-glucuronidase staining of the transgenic line indicated that OsATM3 is highly expressed in lateral root primordia, root tip meristem zones, and shoot apical meristem regions. The average cell size was significantly greater in osatm3 than in the wild type. Massive cell death occurred in the osatm3 root tip meristem zone. Quantitative RT-PCR revealed transcriptional reprogramming of the genes in the osatm3 and RNAi lines involved in DNA repair and cell cycle arrest. Our results suggest that the mitochondrial ATM3 is essential for iron homeostasis in rice.
Collapse
Affiliation(s)
- Jia Zuo
- Key Laboratory of South China Agricultural Plant Molecular Analysis and Genetic Improvement, South China Botanical Garden, Chinese Academy of Sciences, Guangzhou 510650, China (J.Z., Z.W., Y.L., Z.S., X.F., M.Z., H.Y.); and
- University of the Chinese Academy of Sciences, Beijing 100049, China (J.Z., Z.W., Y.L.)
| | - Zhigeng Wu
- Key Laboratory of South China Agricultural Plant Molecular Analysis and Genetic Improvement, South China Botanical Garden, Chinese Academy of Sciences, Guangzhou 510650, China (J.Z., Z.W., Y.L., Z.S., X.F., M.Z., H.Y.); and
- University of the Chinese Academy of Sciences, Beijing 100049, China (J.Z., Z.W., Y.L.)
| | - Ying Li
- Key Laboratory of South China Agricultural Plant Molecular Analysis and Genetic Improvement, South China Botanical Garden, Chinese Academy of Sciences, Guangzhou 510650, China (J.Z., Z.W., Y.L., Z.S., X.F., M.Z., H.Y.); and
- University of the Chinese Academy of Sciences, Beijing 100049, China (J.Z., Z.W., Y.L.)
| | - Zedan Shen
- Key Laboratory of South China Agricultural Plant Molecular Analysis and Genetic Improvement, South China Botanical Garden, Chinese Academy of Sciences, Guangzhou 510650, China (J.Z., Z.W., Y.L., Z.S., X.F., M.Z., H.Y.); and
- University of the Chinese Academy of Sciences, Beijing 100049, China (J.Z., Z.W., Y.L.)
| | - Xiangyang Feng
- Key Laboratory of South China Agricultural Plant Molecular Analysis and Genetic Improvement, South China Botanical Garden, Chinese Academy of Sciences, Guangzhou 510650, China (J.Z., Z.W., Y.L., Z.S., X.F., M.Z., H.Y.); and
- University of the Chinese Academy of Sciences, Beijing 100049, China (J.Z., Z.W., Y.L.)
| | - Mingyong Zhang
- Key Laboratory of South China Agricultural Plant Molecular Analysis and Genetic Improvement, South China Botanical Garden, Chinese Academy of Sciences, Guangzhou 510650, China (J.Z., Z.W., Y.L., Z.S., X.F., M.Z., H.Y.); and
- University of the Chinese Academy of Sciences, Beijing 100049, China (J.Z., Z.W., Y.L.)
| | - Hong Ye
- Key Laboratory of South China Agricultural Plant Molecular Analysis and Genetic Improvement, South China Botanical Garden, Chinese Academy of Sciences, Guangzhou 510650, China (J.Z., Z.W., Y.L., Z.S., X.F., M.Z., H.Y.); and
- University of the Chinese Academy of Sciences, Beijing 100049, China (J.Z., Z.W., Y.L.)
| |
Collapse
|
98
|
Nambot S, Gavrilov D, Thevenon J, Bruel A, Bainbridge M, Rio M, Goizet C, Rötig A, Jaeken J, Niu N, Xia F, Vital A, Houcinat N, Mochel F, Kuentz P, Lehalle D, Duffourd Y, Rivière J, Thauvin-Robinet C, Beaudet A, Faivre L. Further delineation of a rare recessive encephalomyopathy linked to mutations in GFER thanks to data sharing of whole exome sequencing data. Clin Genet 2017; 92:188-198. [DOI: 10.1111/cge.12985] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2016] [Revised: 12/24/2016] [Accepted: 01/25/2017] [Indexed: 02/06/2023]
Affiliation(s)
- S. Nambot
- Centre de Génétique et Centre de référence «Anomalies du Développement et Syndromes Malformatifs», Hôpital d'Enfants; Centre Hospitalier Universitaire de Dijon; Dijon France
- Laboratoire de Génétique Moléculaire, Plateau Technique de Biologie; Centre Hospitalier Universitaire de Dijon; Dijon France
| | - D. Gavrilov
- Biochemical Genetics Laboratory, Department of Laboratory Medicine and Pathology; Mayo Clinic College of Medicine; Rochester Minnesota
- Department of Genetics and Genomics; Mayo Clinic College of Medicine; Rochester Minnesota
| | - J. Thevenon
- Centre de Génétique et Centre de référence «Anomalies du Développement et Syndromes Malformatifs», Hôpital d'Enfants; Centre Hospitalier Universitaire de Dijon; Dijon France
- Fédération Hospitalo-Universitaire Médecine Translationnelle et Anomalies du Développement (FHU TRANSLAD); Centre Hospitalier Universitaire de Dijon et Université de Bourgogne-Franche Comté; Dijon France
- Génétique des Anomalies du Développement; Université de Bourgogne; Dijon France
| | - A.L. Bruel
- Laboratoire de Génétique Moléculaire, Plateau Technique de Biologie; Centre Hospitalier Universitaire de Dijon; Dijon France
- Génétique des Anomalies du Développement; Université de Bourgogne; Dijon France
| | - M. Bainbridge
- Human Genome Sequencing Center; Baylor College of Medicine; Houston Texas
| | - M. Rio
- Service de Génétique Médicale; Hôpital Necker Enfants Malades; Paris France
| | - C. Goizet
- Service de Génétique Médicale; Centre Hospitalier Universitaire de Bordeaux-GH Pellegrin; Bordeaux France
| | - A. Rötig
- Laboratoire de Génétique Moléculaire, Institut de Recherche Necker Enfants Malades; Hôpital Necker Enfants Malades; Paris France
| | - J. Jaeken
- Center for Metabolic Diseases; University Hospital Gasthuisberg; Leuven Belgium
| | - N. Niu
- Department of Molecular and Human Genetics; Baylor College of Medicine; Houston Texas
| | - F. Xia
- Department of Molecular and Human Genetics; Baylor College of Medicine; Houston Texas
| | - A. Vital
- Service de Pathologie, Pôle Biologie et Pathologie; Centre Hospitalier Universitaire de Bordeaux-GH Pellegrin; Bordeaux France
| | - N. Houcinat
- Centre de Génétique et Centre de référence «Anomalies du Développement et Syndromes Malformatifs», Hôpital d'Enfants; Centre Hospitalier Universitaire de Dijon; Dijon France
| | - F. Mochel
- Service de Génétique médicale; Centre Hospitalier Universitaire La Pitié Salpêtrière-Charles Foix; Paris France
| | - P. Kuentz
- Laboratoire de Génétique Moléculaire, Plateau Technique de Biologie; Centre Hospitalier Universitaire de Dijon; Dijon France
| | - D. Lehalle
- Centre de Génétique et Centre de référence «Anomalies du Développement et Syndromes Malformatifs», Hôpital d'Enfants; Centre Hospitalier Universitaire de Dijon; Dijon France
| | - Y. Duffourd
- Fédération Hospitalo-Universitaire Médecine Translationnelle et Anomalies du Développement (FHU TRANSLAD); Centre Hospitalier Universitaire de Dijon et Université de Bourgogne-Franche Comté; Dijon France
- Génétique des Anomalies du Développement; Université de Bourgogne; Dijon France
| | - J.B. Rivière
- Laboratoire de Génétique Moléculaire, Plateau Technique de Biologie; Centre Hospitalier Universitaire de Dijon; Dijon France
- Fédération Hospitalo-Universitaire Médecine Translationnelle et Anomalies du Développement (FHU TRANSLAD); Centre Hospitalier Universitaire de Dijon et Université de Bourgogne-Franche Comté; Dijon France
- Génétique des Anomalies du Développement; Université de Bourgogne; Dijon France
| | - C. Thauvin-Robinet
- Centre de Génétique et Centre de référence «Anomalies du Développement et Syndromes Malformatifs», Hôpital d'Enfants; Centre Hospitalier Universitaire de Dijon; Dijon France
- Fédération Hospitalo-Universitaire Médecine Translationnelle et Anomalies du Développement (FHU TRANSLAD); Centre Hospitalier Universitaire de Dijon et Université de Bourgogne-Franche Comté; Dijon France
- Génétique des Anomalies du Développement; Université de Bourgogne; Dijon France
| | - A.L. Beaudet
- Department of Molecular and Human Genetics; Baylor College of Medicine; Houston Texas
| | - L. Faivre
- Centre de Génétique et Centre de référence «Anomalies du Développement et Syndromes Malformatifs», Hôpital d'Enfants; Centre Hospitalier Universitaire de Dijon; Dijon France
- Fédération Hospitalo-Universitaire Médecine Translationnelle et Anomalies du Développement (FHU TRANSLAD); Centre Hospitalier Universitaire de Dijon et Université de Bourgogne-Franche Comté; Dijon France
- Génétique des Anomalies du Développement; Université de Bourgogne; Dijon France
| |
Collapse
|
99
|
Patnaik MM, Tefferi A. Refractory anemia with ring sideroblasts (RARS) and RARS with thrombocytosis (RARS-T): 2017 update on diagnosis, risk-stratification, and management. Am J Hematol 2017; 92:297-310. [PMID: 28188970 DOI: 10.1002/ajh.24637] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2016] [Revised: 12/23/2016] [Accepted: 12/23/2016] [Indexed: 02/06/2023]
Abstract
DISEASE OVERVIEW Ring sideroblasts (RS) are erythroid precursors with abnormal perinuclear mitochondrial iron accumulation. Two myeloid neoplasms defined by the presence of RS, include refractory anemia with ring sideroblasts (RARS), now classified under myelodysplastic syndromes with RS (MDS-RS) and RARS with thrombocytosis (RARS-T); now called myelodysplastic/myeloproliferative neoplasm with RS and thrombocytosis (MDS/MPN-RS-T). DIAGNOSIS MDS-RS is a lower risk MDS, with single or multilineage dysplasia (SLD/MLD), <5% bone marrow (BM) blasts and ≥15% BM RS (≥5% in the presence of SF3B1 mutations). MDS/MPN-RS-T, now a formal entity in the MDS/MPN overlap syndromes, has diagnostic features of MDS-RS-SLD, along with a platelet count ≥ 450 × 10(9)/L and large atypical megakaryocytes (similar to BCR-ABL1 negative MPN). MUTATIONS AND KARYOTYPE Mutations in SF3B1 are seen in ≥80% of patients with MDS-RS-SLD and MDS/MPN-RS-T, and strongly correlate with the presence of BM RS; MDS/MPN-RS-T patients also demonstrate JAK2V617F, ASXL1, DNMT3A, SETBP1, and TET2 mutations; with ASXL1/SETBP1 mutations adversely impacting survival. Cytogenetic abnormalities are uncommon in both diseases. RISK STRATIFICATION Most patients with MDS-RS-SLD are stratified into lower risk groups by the revised-International Prognostic Scoring System (R-IPSS). Disease outcome in MDS/MPN-RS-T is better than that of MDS-RS-SLD, but worse than that of essential thrombocythemia. Both diseases have a low risk of leukemic TREATMENT: Anemia and iron overload are complications seen in both and are managed similar to lower risk MDS and MPN. Aspirin therapy is reasonable in MDS/MPN-RS-T, especially in the presence of JAK2V617F, but the value of platelet-lowering drugs is uncertain.
Collapse
Affiliation(s)
- Mrinal M. Patnaik
- Division of Hematology, Department of Internal MedicineMayo ClinicRochester Minnesota
| | - Ayalew Tefferi
- Division of Hematology, Department of Internal MedicineMayo ClinicRochester Minnesota
| |
Collapse
|
100
|
A novel fluorescence probe based on triphenylamine Schiff base for bioimaging and responding to pH and Fe 3+. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2017; 72:551-557. [DOI: 10.1016/j.msec.2016.11.108] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/05/2016] [Revised: 10/25/2016] [Accepted: 11/23/2016] [Indexed: 11/22/2022]
|