1
|
Wilkinson T, Fletcher E, Ring A, Bradley C, Papoutsi E, Miserlis D, Smith RS, Bohannon WT, Pipinos II, Koutakis P. Deciphering the Role of Ferroptosis in the Pathogenesis of Peripheral Artery Disease Myopathy. BIOLOGY 2025; 14:537. [PMID: 40427725 DOI: 10.3390/biology14050537] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/20/2025] [Revised: 05/01/2025] [Accepted: 05/07/2025] [Indexed: 05/29/2025]
Abstract
This study investigates ferroptosis in the context of peripheral artery disease (PAD), a vascular disease characterized by atherosclerosis of the lower extremities. Muscle atrophy and increased oxidative stress are hallmarks of PAD and correlate with worse clinical outcomes. Given ferroptosis' association with oxidative stress, we explored its role in PAD myopathy by examining gene and protein markers related to metabolic pathways implicated in ferroptosis using both human PAD patients and cultured myotubes. Intermittent claudication (IC) PAD patients, critical limb ischemia (CLI) PAD patients, and non-PAD controls were recruited for this study. Calf muscle biopsies were analyzed for gene expression using qPCR, and protein levels were determined by Western blotting. Cultured myotubes treated with the ferroptosis inducer erastin provided an in vitro comparison. Results demonstrated upregulation of ferroptosis markers such as lipid peroxidation and PTGS2 gene expression in the muscle of CLI PAD patients compared to controls. Increased expression of ferroptosis-related genes HMOX1, ACSL4, ELAVL1, and Beclin-1 was also observed. Protein analysis showed trends consistent with gene expression in some ferroptosis markers. The increase in ferroptosis markers in CLI PAD patients, particularly in iron metabolism and autophagy pathways, suggests ferroptosis contributes to PAD myopathy.
Collapse
Affiliation(s)
| | - Emma Fletcher
- Department of Public Health, University of West Florida, Pensacola, FL 32514, USA
| | - Andrew Ring
- Department of Biology, Baylor University, Waco, TX 76798, USA
| | | | - Evlampia Papoutsi
- Department of Public Health, University of West Florida, Pensacola, FL 32514, USA
| | - Dimitrios Miserlis
- Department of Surgery and Perioperative Care, Dell Medical School, University of Texas, Austin, TX 78712, USA
| | - Robert S Smith
- Department of Surgery, Baylor Scott and White Medical Center, Temple, TX 76508, USA
| | - William T Bohannon
- Department of Surgery, Baylor Scott and White Medical Center, Temple, TX 76508, USA
| | - Iraklis I Pipinos
- Department of Surgery, University of Nebraska at Medical Center, Omaha, NE 68198, USA
| | - Panagiotis Koutakis
- Department of Biology, Baylor University, Waco, TX 76798, USA
- Department of Public Health, University of West Florida, Pensacola, FL 32514, USA
- Department of Surgery, University of Nebraska at Medical Center, Omaha, NE 68198, USA
| |
Collapse
|
2
|
Koleva P, He J, Dunsmore G, Bozorgmehr N, Lu J, Huynh M, Tollenaar S, Huang V, Walter J, Way SS, Elahi S. CD71 + erythroid cells promote intestinal symbiotic microbial communities in pregnancy and neonatal period. MICROBIOME 2024; 12:142. [PMID: 39080725 PMCID: PMC11290123 DOI: 10.1186/s40168-024-01859-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/02/2023] [Accepted: 06/15/2024] [Indexed: 08/02/2024]
Abstract
BACKGROUND The establishment of microbial communities in neonatal mammals plays a pivotal role in shaping their immune responses to infections and other immune-related conditions. This process is influenced by a combination of endogenous and exogenous factors. Previously, we reported that depletion of CD71 + erythroid cells (CECs) results in an inflammatory response to microbial communities in newborn mice. RESULTS Here, we systemically tested this hypothesis and observed that the small intestinal lamina propria of neonatal mice had the highest frequency of CECs during the early days of life. This high abundance of CECs was attributed to erythropoiesis niches within the small intestinal tissues. Notably, the removal of CECs from the intestinal tissues by the anti-CD71 antibody disrupted immune homeostasis. This disruption was evident by alteration in the expression of antimicrobial peptides (AMPs), toll-like receptors (TLRs), inflammatory cytokines/chemokines, and resulting in microbial dysbiosis. Intriguingly, these alterations in microbial communities persisted when tested 5 weeks post-treatment, with a more notable effect observed in female mice. This illustrates a sex-dependent association between CECs and neonatal microbiome modulation. Moreover, we extended our studies on pregnant mice, observing that modulating CECs substantially alters the frequency and diversity of their microbial communities. Finally, we found a significantly lower proportion of CECs in the cord blood of pre-term human newborns, suggesting a potential role in dysregulated immune responses to microbial communities in the gut. CONCLUSIONS Our findings provide novel insights into pivotal role of CECs in immune homeostasis and swift adaptation of microbial communities in newborns. Despite the complexity of the cellular biology of the gut, our findings shed light on the previously unappreciated role of CECs in the dialogue between the microbiota and immune system. These findings have significant implications for human health. Video Abstract.
Collapse
Affiliation(s)
- Petya Koleva
- School of Dentistry, Division of Foundational Sciences, Faculty of Medicine and Dentistry, Edmonton, Canada
| | - Jia He
- School of Dentistry, Division of Foundational Sciences, Faculty of Medicine and Dentistry, Edmonton, Canada
| | - Garett Dunsmore
- School of Dentistry, Division of Foundational Sciences, Faculty of Medicine and Dentistry, Edmonton, Canada
| | - Najmeh Bozorgmehr
- School of Dentistry, Division of Foundational Sciences, Faculty of Medicine and Dentistry, Edmonton, Canada
| | - Julia Lu
- School of Dentistry, Division of Foundational Sciences, Faculty of Medicine and Dentistry, Edmonton, Canada
| | - Maia Huynh
- School of Dentistry, Division of Foundational Sciences, Faculty of Medicine and Dentistry, Edmonton, Canada
| | - Stephanie Tollenaar
- Department of Agricultural, Food & Nutritional Sciences, Edmonton, University of Alberta, Edmonton, Canada
| | - Vivian Huang
- Division of Gastroenterology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Canada
- Division of Gastroenterology, Mount Sinai Hospital, Toronto, Canada
| | - Jens Walter
- Department of Agricultural, Food & Nutritional Sciences, Edmonton, University of Alberta, Edmonton, Canada
- School of Microbiology and Department of Medicine, APC Microbiome Ireland, University College Cork, National University of Ireland, Cork, Ireland
| | - Sing Sing Way
- Centre for Inflammation and Tolerance, Cincinnati Childrens Hospital, College of Medicine, University of Cincinnati, Cincinnati, OH, USA
| | - Shokrollah Elahi
- School of Dentistry, Division of Foundational Sciences, Faculty of Medicine and Dentistry, Edmonton, Canada.
- Li Ka Shing Institute of Virology, University of Alberta, Edmonton, Canada.
- Glycomics Institute of Alberta, University of Alberta, Edmonton, Canada.
- Women and Children's Health Research Institute, University of Alberta, Edmonton, Canada.
- Alberta Transplant Institute, Edmonton, AB, Canada.
- 7020G Katz Group Centre for Pharmacology and Health Research, 11361-87Th Ave NW, Edmonton, AB, T6G2E1, Canada.
| |
Collapse
|
3
|
Xie T, Yao L, Li X. Advance in Iron Metabolism, Oxidative Stress and Cellular Dysfunction in Experimental and Human Kidney Diseases. Antioxidants (Basel) 2024; 13:659. [PMID: 38929098 PMCID: PMC11200795 DOI: 10.3390/antiox13060659] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2024] [Revised: 05/22/2024] [Accepted: 05/24/2024] [Indexed: 06/28/2024] Open
Abstract
Kidney diseases pose a significant global health issue, frequently resulting in the gradual decline of renal function and eventually leading to end-stage renal failure. Abnormal iron metabolism and oxidative stress-mediated cellular dysfunction facilitates the advancement of kidney diseases. Iron homeostasis is strictly regulated in the body, and disturbance in this regulatory system results in abnormal iron accumulation or deficiency, both of which are associated with the pathogenesis of kidney diseases. Iron overload promotes the production of reactive oxygen species (ROS) through the Fenton reaction, resulting in oxidative damage to cellular molecules and impaired cellular function. Increased oxidative stress can also influence iron metabolism through upregulation of iron regulatory proteins and altering the expression and activity of key iron transport and storage proteins. This creates a harmful cycle in which abnormal iron metabolism and oxidative stress perpetuate each other, ultimately contributing to the advancement of kidney diseases. The crosstalk of iron metabolism and oxidative stress involves multiple signaling pathways, such as hypoxia-inducible factor (HIF) and nuclear factor erythroid 2-related factor 2 (Nrf2) pathways. This review delves into the functions and mechanisms of iron metabolism and oxidative stress, along with the intricate relationship between these two factors in the context of kidney diseases. Understanding the underlying mechanisms should help to identify potential therapeutic targets and develop novel and effective therapeutic strategies to combat the burden of kidney diseases.
Collapse
Affiliation(s)
- Tiancheng Xie
- Department of Internal Medicine, Mayo Clinic, Rochester, MN 55905, USA;
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN 55905, USA
| | - Li Yao
- Department of Nephrology, The First Hospital of China Medical University, Shenyang 110001, China;
| | - Xiaogang Li
- Department of Internal Medicine, Mayo Clinic, Rochester, MN 55905, USA;
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN 55905, USA
| |
Collapse
|
4
|
Huang W, Das NK, Radyk MD, Keeley T, Quiros M, Jain C, El-Derany MO, Swaminathan T, Dziechciarz S, Greenson JK, Nusrat A, Samuelson LC, Shah YM. Dietary Iron Is Necessary to Support Proliferative Regeneration after Intestinal Injury. J Nutr 2024; 154:1153-1164. [PMID: 38246358 PMCID: PMC11181351 DOI: 10.1016/j.tjnut.2024.01.013] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2023] [Revised: 12/04/2023] [Accepted: 01/04/2024] [Indexed: 01/23/2024] Open
Abstract
BACKGROUND Tissue repair and regeneration in the gastrointestinal system are crucial for maintaining homeostasis, with the process relying on intricate cellular interactions and affected by micro- and macro-nutrients. Iron, essential for various biological functions, plays a dual role in tissue healing by potentially causing oxidative damage and participating in anti-inflammatory mechanisms, underscoring its complex relationship with inflammation and tissue repair. OBJECTIVE The study aimed to elucidate the role of low dietary iron in gastrointestinal tissue repair. METHODS We utilized quantitative iron measurements to assess iron levels in inflamed regions of patients with ulcerative colitis and Crohn's disease. In addition, 3 mouse models of gastrointestinal injury/repair (dextran sulfate sodium-induced colitis, radiation injury, and wound biopsy) were used to assess the effects of low dietary iron on tissue repair. RESULTS We found that levels of iron in inflamed regions of both patients with ulcerative colitis and Crohn's disease are elevated. Similarly, during gastrointestinal repair, iron levels were found to be heightened, specifically in intestinal epithelial cells across the 3 injury/repair models. Mice on a low-iron diet showed compromised tissue repair with reduced proliferation. In standard diet, epithelial cells and the stem cell compartment maintain adequate iron stores. However, during a period of iron deficiency, epithelial cells exhaust their iron reserves, whereas the stem cell compartments maintain their iron pools. During injury, when the stem compartment is disrupted, low iron levels impair proliferation and compromise repair mechanisms. CONCLUSIONS Low dietary iron impairs intestinal repair through compromising the ability of epithelial cells to aid in intestinal proliferation.
Collapse
Affiliation(s)
- Wesley Huang
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI, United States; Department of Cellular and Molecular Biology, University of Michigan, Ann Arbor, MI, United States; Medical Scientist Training Program, University of Michigan, Ann Arbor, MI, United States
| | - Nupur K Das
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI, United States
| | - Megan D Radyk
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI, United States
| | - Theresa Keeley
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI, United States
| | - Miguel Quiros
- Department of Pathology, University of Michigan, Ann Arbor, MI, United States
| | - Chesta Jain
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI, United States
| | - Marwa O El-Derany
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI, United States; Department of Biochemistry, Faculty of Pharmacy, Ain Shams University, Cairo, Egypt
| | - Thaarini Swaminathan
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI, United States
| | - Sofia Dziechciarz
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI, United States
| | - Joel K Greenson
- Department of Pathology, University of Michigan, Ann Arbor, MI, United States
| | - Asma Nusrat
- Department of Pathology, University of Michigan, Ann Arbor, MI, United States
| | - Linda C Samuelson
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI, United States
| | - Yatrik M Shah
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI, United States; Department of Cellular and Molecular Biology, University of Michigan, Ann Arbor, MI, United States.
| |
Collapse
|
5
|
Zhang Y, Chen L, Xuan Y, Zhang L, Tian W, Zhu Y, Wang J, Wang X, Qiu J, Yu J, Tang M, He Z, Zhang H, Chen S, Shen Y, Wang S, Zhang R, Xu L, Ma X, Liao Y, Hu C. Iron overload in hypothalamic AgRP neurons contributes to obesity and related metabolic disorders. Cell Rep 2024; 43:113900. [PMID: 38460132 DOI: 10.1016/j.celrep.2024.113900] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Revised: 01/12/2024] [Accepted: 02/15/2024] [Indexed: 03/11/2024] Open
Abstract
Iron overload is closely associated with metabolic dysfunction. However, the role of iron in the hypothalamus remains unclear. Here, we find that hypothalamic iron levels are increased, particularly in agouti-related peptide (AgRP)-expressing neurons in high-fat-diet-fed mice. Using pharmacological or genetic approaches, we reduce iron overload in AgRP neurons by central deferoxamine administration or transferrin receptor 1 (Tfrc) deletion, ameliorating diet-induced obesity and related metabolic dysfunction. Conversely, Tfrc-mediated iron overload in AgRP neurons leads to overeating and adiposity. Mechanistically, the reduction of iron overload in AgRP neurons inhibits AgRP neuron activity; improves insulin and leptin sensitivity; and inhibits iron-induced oxidative stress, endoplasmic reticulum stress, nuclear factor κB signaling, and suppression of cytokine signaling 3 expression. These results highlight the critical role of hypothalamic iron in obesity development and suggest targets for treating obesity and related metabolic disorders.
Collapse
Affiliation(s)
- Yi Zhang
- Shanghai Diabetes Institute, Shanghai Key Laboratory of Diabetes Mellitus, Shanghai Clinical Centre for Diabetes, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China
| | - Liwei Chen
- Shanghai Diabetes Institute, Shanghai Key Laboratory of Diabetes Mellitus, Shanghai Clinical Centre for Diabetes, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China
| | - Ye Xuan
- Shanghai Diabetes Institute, Shanghai Key Laboratory of Diabetes Mellitus, Shanghai Clinical Centre for Diabetes, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China
| | - Lina Zhang
- Shanghai Diabetes Institute, Shanghai Key Laboratory of Diabetes Mellitus, Shanghai Clinical Centre for Diabetes, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China; School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Wen Tian
- Shanghai Diabetes Institute, Shanghai Key Laboratory of Diabetes Mellitus, Shanghai Clinical Centre for Diabetes, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China; Department of Endocrinology, Jinzhou Medical University, Jinzhou 121001, China
| | - Yangyang Zhu
- Shanghai Diabetes Institute, Shanghai Key Laboratory of Diabetes Mellitus, Shanghai Clinical Centre for Diabetes, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China; Institute for Metabolic Disease, Fengxian Central Hospital Affiliated to Southern Medical University, Shanghai 226001, China
| | - Jinghui Wang
- Shanghai Diabetes Institute, Shanghai Key Laboratory of Diabetes Mellitus, Shanghai Clinical Centre for Diabetes, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China; Department of Endocrinology, Xihua Xian People's Hospital, Zhoukou 466000, China
| | - Xinyu Wang
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Jin Qiu
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai 200241, China
| | - Jian Yu
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai 200241, China
| | - Mengyang Tang
- Shanghai Diabetes Institute, Shanghai Key Laboratory of Diabetes Mellitus, Shanghai Clinical Centre for Diabetes, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China; Institute for Metabolic Disease, Fengxian Central Hospital Affiliated to Southern Medical University, Shanghai 226001, China
| | - Zhen He
- Shanghai Diabetes Institute, Shanghai Key Laboratory of Diabetes Mellitus, Shanghai Clinical Centre for Diabetes, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China
| | - Hong Zhang
- Shanghai Diabetes Institute, Shanghai Key Laboratory of Diabetes Mellitus, Shanghai Clinical Centre for Diabetes, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China
| | - Si Chen
- Shanghai Diabetes Institute, Shanghai Key Laboratory of Diabetes Mellitus, Shanghai Clinical Centre for Diabetes, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China
| | - Yun Shen
- Shanghai Diabetes Institute, Shanghai Key Laboratory of Diabetes Mellitus, Shanghai Clinical Centre for Diabetes, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China
| | - Siyi Wang
- Department of Pathology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Rong Zhang
- Shanghai Diabetes Institute, Shanghai Key Laboratory of Diabetes Mellitus, Shanghai Clinical Centre for Diabetes, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China
| | - Lingyan Xu
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai 200241, China.
| | - Xinran Ma
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai 200241, China; Chongqing Institute for Brain and Intelligence, Guangyang Bay Laboratory, Chongqing 400064, China.
| | - Yunfei Liao
- Department of Endocrinology, Wuhan Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, China.
| | - Cheng Hu
- Shanghai Diabetes Institute, Shanghai Key Laboratory of Diabetes Mellitus, Shanghai Clinical Centre for Diabetes, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China; Institute for Metabolic Disease, Fengxian Central Hospital Affiliated to Southern Medical University, Shanghai 226001, China.
| |
Collapse
|
6
|
Galy B, Conrad M, Muckenthaler M. Mechanisms controlling cellular and systemic iron homeostasis. Nat Rev Mol Cell Biol 2024; 25:133-155. [PMID: 37783783 DOI: 10.1038/s41580-023-00648-1] [Citation(s) in RCA: 241] [Impact Index Per Article: 241.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/31/2023] [Indexed: 10/04/2023]
Abstract
In mammals, hundreds of proteins use iron in a multitude of cellular functions, including vital processes such as mitochondrial respiration, gene regulation and DNA synthesis or repair. Highly orchestrated regulatory systems control cellular and systemic iron fluxes ensuring sufficient iron delivery to target proteins is maintained, while limiting its potentially deleterious effects in iron-mediated oxidative cell damage and ferroptosis. In this Review, we discuss how cells acquire, traffick and export iron and how stored iron is mobilized for iron-sulfur cluster and haem biogenesis. Furthermore, we describe how these cellular processes are fine-tuned by the combination of various sensory and regulatory systems, such as the iron-regulatory protein (IRP)-iron-responsive element (IRE) network, the nuclear receptor co-activator 4 (NCOA4)-mediated ferritinophagy pathway, the prolyl hydroxylase domain (PHD)-hypoxia-inducible factor (HIF) axis or the nuclear factor erythroid 2-related factor 2 (NRF2) regulatory hub. We further describe how these pathways interact with systemic iron homeostasis control through the hepcidin-ferroportin axis to ensure appropriate iron fluxes. This knowledge is key for the identification of novel therapeutic opportunities to prevent diseases of cellular and/or systemic iron mismanagement.
Collapse
Affiliation(s)
- Bruno Galy
- German Cancer Research Center (DKFZ), Division of Virus-associated Carcinogenesis (F170), Heidelberg, Germany
| | - Marcus Conrad
- Helmholtz Zentrum München, Institute of Metabolism and Cell Death, Neuherberg, Germany
| | - Martina Muckenthaler
- Department of Paediatric Hematology, Oncology and Immunology, University of Heidelberg, Heidelberg, Germany.
- Molecular Medicine Partnership Unit, University of Heidelberg, Heidelberg, Germany.
- German Centre for Cardiovascular Research (DZHK), Partner site Heidelberg/Mannheim, Heidelberg, Germany.
- Translational Lung Research Center Heidelberg (TLRC), German Center for Lung Research (DZL), University of Heidelberg, Heidelberg, Germany.
| |
Collapse
|
7
|
I Kutyavin V, Korn LL, Medzhitov R. Nutrient-derived signals regulate eosinophil adaptation to the small intestine. Proc Natl Acad Sci U S A 2024; 121:e2316446121. [PMID: 38271336 PMCID: PMC10835075 DOI: 10.1073/pnas.2316446121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2023] [Accepted: 12/05/2023] [Indexed: 01/27/2024] Open
Abstract
Eosinophils are well recognized as effector cells of type 2 immunity, yet they also accumulate in many tissues under homeostatic conditions. However, the processes that govern homeostatic eosinophil accumulation and tissue-specific adaptation, and their functional significance, remain poorly defined. Here, we investigated how eosinophils adapt to the small intestine (SI) microenvironment and the local signals that regulate this process. We observed that eosinophils gradually migrate along the crypt-villus axis, giving rise to a villus-resident subpopulation with a distinct transcriptional signature. Retinoic acid signaling was specifically required for maintenance of this subpopulation, while IL-5 was largely dispensable outside of its canonical role in eosinophil production. Surprisingly, we found that a high-protein diet suppressed the accumulation of villus-resident eosinophils. Purified amino acids were sufficient for this effect, which was a consequence of accelerated eosinophil turnover within the tissue microenvironment and was not due to altered development in the bone marrow. Our study provides insight into the process of eosinophil adaptation to the SI, highlighting its reliance on nutrient-derived signals.
Collapse
Affiliation(s)
- Vassily I Kutyavin
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06510
| | - Lisa L Korn
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06510
- Department of Medicine (Rheumatology), Yale University School of Medicine, New Haven, CT 06510
| | - Ruslan Medzhitov
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06510
- HHMI, Yale University School of Medicine, New Haven, CT 06510
| |
Collapse
|
8
|
Xiong L, Helm EY, Dean JW, Sun N, Jimenez-Rondan FR, Zhou L. Nutrition impact on ILC3 maintenance and function centers on a cell-intrinsic CD71-iron axis. Nat Immunol 2023; 24:1671-1684. [PMID: 37709985 PMCID: PMC11256193 DOI: 10.1038/s41590-023-01612-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2022] [Accepted: 08/04/2023] [Indexed: 09/16/2023]
Abstract
Iron metabolism is pivotal for cell fitness in the mammalian host; however, its role in group 3 innate lymphoid cells (ILC3s) is unknown. Here we show that transferrin receptor CD71 (encoded by Tfrc)-mediated iron metabolism cell-intrinsically controls ILC3 proliferation and host protection against Citrobacter rodentium infection and metabolically affects mitochondrial respiration by switching of oxidative phosphorylation toward glycolysis. Iron deprivation or Tfrc ablation in ILC3s reduces the expression and/or activity of the aryl hydrocarbon receptor (Ahr), a key ILC3 regulator. Genetic ablation or activation of Ahr in ILC3s leads to CD71 upregulation or downregulation, respectively, suggesting Ahr-mediated suppression of CD71. Mechanistically, Ahr directly binds to the Tfrc promoter to inhibit transcription. Iron overload partially restores the defective ILC3 compartment in the small intestine of Ahr-deficient mice, consistent with the compensatory upregulation of CD71. These data collectively demonstrate an under-appreciated role of the Ahr-CD71-iron axis in the regulation of ILC3 maintenance and function.
Collapse
Affiliation(s)
- Lifeng Xiong
- Department of Infectious Diseases and Immunology, College of Veterinary Medicine, University of Florida, Gainesville, FL, USA
| | - Eric Y Helm
- Department of Infectious Diseases and Immunology, College of Veterinary Medicine, University of Florida, Gainesville, FL, USA
| | - Joseph W Dean
- Department of Infectious Diseases and Immunology, College of Veterinary Medicine, University of Florida, Gainesville, FL, USA
| | - Na Sun
- Department of Infectious Diseases and Immunology, College of Veterinary Medicine, University of Florida, Gainesville, FL, USA
| | - Felix R Jimenez-Rondan
- Center for Nutritional Sciences and Food Science and Human Nutrition Department, University of Florida, Gainesville, FL, USA
| | - Liang Zhou
- Department of Infectious Diseases and Immunology, College of Veterinary Medicine, University of Florida, Gainesville, FL, USA.
| |
Collapse
|
9
|
Xiong W, Jin L, Zhao Y, Wu Y, Dong J, Guo Z, Zhu M, Dai Y, Pan Y, Zhu X. Deletion of Transferrin Receptor 1 in Parvalbumin Interneurons Induces a Hereditary Spastic Paraplegia-Like Phenotype. J Neurosci 2023; 43:5092-5113. [PMID: 37308296 PMCID: PMC10325000 DOI: 10.1523/jneurosci.2277-22.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Revised: 06/01/2023] [Accepted: 06/05/2023] [Indexed: 06/14/2023] Open
Abstract
Hereditary spastic paraplegia (HSP) is a severe neurodegenerative movement disorder, the underlying pathophysiology of which remains poorly understood. Mounting evidence has suggested that iron homeostasis dysregulation can lead to motor function impairment. However, whether deficits in iron homeostasis are involved in the pathophysiology of HSP remains unknown. To address this knowledge gap, we focused on parvalbumin-positive (PV+) interneurons, a large category of inhibitory neurons in the central nervous system, which play a critical role in motor regulation. The PV+ interneuron-specific deletion of the gene encoding transferrin receptor 1 (TFR1), a key component of the neuronal iron uptake machinery, induced severe progressive motor deficits in both male and female mice. In addition, we observed skeletal muscle atrophy, axon degeneration in the spinal cord dorsal column, and alterations in the expression of HSP-related proteins in male mice with Tfr1 deletion in the PV+ interneurons. These phenotypes were highly consistent with the core clinical features of HSP cases. Furthermore, the effects on motor function induced by Tfr1 ablation in PV+ interneurons were mostly concentrated in the dorsal spinal cord; however, iron repletion partly rescued the motor defects and axon loss seen in both sexes of conditional Tfr1 mutant mice. Our study describes a new mouse model for mechanistic and therapeutic studies relating to HSP and provides novel insights into iron metabolism in spinal cord PV+ interneurons and its role in the regulation of motor functions.SIGNIFICANCE STATEMENT Iron is crucial for neuronal functioning. Mounting evidence suggests that iron homeostasis dysregulation can induce motor function deficits. Transferrin receptor 1 (TFR1) is thought to be the key component in neuronal iron uptake. We found that deletion of Tfr1 in parvalbumin-positive (PV+) interneurons in mice induced severe progressive motor deficits, skeletal muscle atrophy, axon degeneration in the spinal cord dorsal column, and alterations in the expression of hereditary spastic paraplegia (HSP)-related proteins. These phenotypes were highly consistent with the core clinical features of HSP cases and partly rescued by iron repletion. This study describes a new mouse model for the study of HSP and provides novel insights into iron metabolism in spinal cord PV+ interneurons.
Collapse
Affiliation(s)
- Wenchao Xiong
- School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Liqiang Jin
- School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Yulu Zhao
- School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Yu Wu
- School of Psychology, Shenzhen University, Shenzhen 518060, China
| | - Jinghua Dong
- School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Zhixin Guo
- School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Minzhen Zhu
- School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Yongfeng Dai
- School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Yida Pan
- School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Xinhong Zhu
- School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
- School of Psychology, Shenzhen University, Shenzhen 518060, China
- Research Center for Brain Health, Pazhou Lab, Guangzhou 510330, China
| |
Collapse
|
10
|
Denton SL, Mejia A, Nevarez LL, Soares MP, Fox BA, Bzik DJ, Gigley JP. Theft of Host Transferrin Receptor-1 by Toxoplasma gondii is required for infection. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.06.23.546322. [PMID: 39372795 PMCID: PMC11451604 DOI: 10.1101/2023.06.23.546322] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/08/2024]
Abstract
Nutrient acquisition by apicomplexan parasites is essential to drive their intracellular replication, yet the mechanisms that underpin essential nutrient acquisition are not defined. Using the apicomplexan model Toxoplasma gondii , we show that host cell proteins including the transferrin receptor 1, transferrin, ferritin heavy and light chains, and clathrin light chain are robustly taken up by tachyzoites. Tachyzoite acquisition of host cell protein was not related to host cell type or parasite virulence phenotypes. Bradyzoites possessed little capacity to acquire host cell proteins consistent with the cyst wall representing a barrier to host cell protein cargo. Increased trafficking of host cell transferrin receptor 1 and transferrin to endolysosomes boosted tachyzoite acquisition of host proteins and growth rate. Theft of host transferrin 1 and transferrin did not significantly affect iron levels in the tachyzoite. This study provides insight into essential functions associated with parasite theft of host iron sequestration and storage proteins.
Collapse
|
11
|
Cheli VT, Santiago González DA, Wan R, Rosenblum SL, Denaroso GE, Angeliu CG, Smith Z, Wang C, Paez PM. Transferrin Receptor Is Necessary for Proper Oligodendrocyte Iron Homeostasis and Development. J Neurosci 2023; 43:3614-3629. [PMID: 36977582 PMCID: PMC10198458 DOI: 10.1523/jneurosci.1383-22.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Revised: 03/15/2023] [Accepted: 03/21/2023] [Indexed: 03/30/2023] Open
Abstract
To test the hypothesis that the transferrin (Tf) cycle has unique importance for oligodendrocyte development and function, we disrupted the expression of the Tf receptor (Tfr) gene in oligodendrocyte progenitor cells (OPCs) on mice of either sex using the Cre/lox system. This ablation results in the elimination of iron incorporation via the Tf cycle but leaves other Tf functions intact. Mice lacking Tfr, specifically in NG2 or Sox10-positive OPCs, developed a hypomyelination phenotype. Both OPC differentiation and myelination were affected, and Tfr deletion resulted in impaired OPC iron absorption. Specifically, the brains of Tfr cKO animals presented a reduction in the quantity of myelinated axons, as well as fewer mature oligodendrocytes. In contrast, the ablation of Tfr in adult mice affected neither mature oligodendrocytes nor myelin synthesis. RNA-seq analysis performed in Tfr cKO OPCs revealed misregulated genes involved in OPC maturation, myelination, and mitochondrial activity. Tfr deletion in cortical OPCs also disrupted the activity of the mTORC1 signaling pathway, epigenetic mechanisms critical for gene transcription and the expression of structural mitochondrial genes. RNA-seq studies were additionally conducted in OPCs in which iron storage was disrupted by deleting the ferritin heavy chain. These OPCs display abnormal regulation of genes associated with iron transport, antioxidant activity, and mitochondrial activity. Thus, our results indicate that the Tf cycle is central for iron homeostasis in OPCs during postnatal development and suggest that both iron uptake via Tfr and iron storage in ferritin are critical for energy production, mitochondrial activity, and maturation of postnatal OPCs.SIGNIFICANCE STATEMENT By knocking-out transferrin receptor (Tfr) specifically in oligodendrocyte progenitor cells (OPCs), we have established that iron incorporation via the Tf cycle is key for OPC iron homeostasis and for the normal function of these cells during the postnatal development of the CNS. Moreover, RNA-seq analysis indicated that both Tfr iron uptake and ferritin iron storage are critical for proper OPC mitochondrial activity, energy production, and maturation.
Collapse
Affiliation(s)
- Veronica T Cheli
- Institute for Myelin and Glia Exploration, Department of Pharmacology and Toxicology, Jacobs School of Medicine and Biomedical Sciences, State University of New York, University at Buffalo, Buffalo, New York 14203
| | - Diara A Santiago González
- Institute for Myelin and Glia Exploration, Department of Pharmacology and Toxicology, Jacobs School of Medicine and Biomedical Sciences, State University of New York, University at Buffalo, Buffalo, New York 14203
| | - Rensheng Wan
- Institute for Myelin and Glia Exploration, Department of Pharmacology and Toxicology, Jacobs School of Medicine and Biomedical Sciences, State University of New York, University at Buffalo, Buffalo, New York 14203
| | - Shaina L Rosenblum
- Institute for Myelin and Glia Exploration, Department of Pharmacology and Toxicology, Jacobs School of Medicine and Biomedical Sciences, State University of New York, University at Buffalo, Buffalo, New York 14203
| | - Giancarlo E Denaroso
- Institute for Myelin and Glia Exploration, Department of Pharmacology and Toxicology, Jacobs School of Medicine and Biomedical Sciences, State University of New York, University at Buffalo, Buffalo, New York 14203
| | - Christina G Angeliu
- Institute for Myelin and Glia Exploration, Department of Pharmacology and Toxicology, Jacobs School of Medicine and Biomedical Sciences, State University of New York, University at Buffalo, Buffalo, New York 14203
| | - Zachary Smith
- Institute for Myelin and Glia Exploration, Department of Pharmacology and Toxicology, Jacobs School of Medicine and Biomedical Sciences, State University of New York, University at Buffalo, Buffalo, New York 14203
| | - Congying Wang
- Institute for Myelin and Glia Exploration, Department of Pharmacology and Toxicology, Jacobs School of Medicine and Biomedical Sciences, State University of New York, University at Buffalo, Buffalo, New York 14203
| | - Pablo M Paez
- Institute for Myelin and Glia Exploration, Department of Pharmacology and Toxicology, Jacobs School of Medicine and Biomedical Sciences, State University of New York, University at Buffalo, Buffalo, New York 14203
| |
Collapse
|
12
|
Yu J, Ruaux C, Griebsch C, Boland L, Wong N, Bennett P, Wasinger VC. Serum proteome of dogs with chronic enteropathy. J Vet Intern Med 2023; 37:925-935. [PMID: 37186013 DOI: 10.1111/jvim.16682] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Accepted: 02/24/2023] [Indexed: 05/17/2023] Open
Abstract
BACKGROUND Chronic enteropathy (CE) is common in dogs and can occur with multiple etiologies including food-responsive enteropathy (FRE) and idiopathic inflammatory bowel disease (IBD). HYPOTHESIS/OBJECTIVE To study the protein profile and pathway differences among dogs with FRE, IBD, and healthy controls using serum proteome analysis. ANIMALS Nine CE dogs with signs of gastrointestinal disease and histologically confirmed chronic inflammatory enteropathy and 16 healthy controls. METHODS A cross-sectional study with cases recruited from 2 veterinary hospitals between May 2019 and November 2020 was performed. Serum samples were analyzed using mass spectrometry-based proteomic techniques. RESULTS Proteomic profiles showed marked variation in relative protein abundances. Forty-five proteins were significantly (P ≤ .01) differentially expressed among the dogs with CE and controls with ≥2-fold change in abundance. The fold change of dogs with IBD normalized to controls was more pronounced for the majority of proteins than that seen in the dogs with FRE normalized to control dogs. Proteins involving reactive oxygen species, cytokine activation, acute phase response signaling, and lipid metabolism were altered in dogs with CE. CONCLUSIONS AND CLINICAL IMPORTANCE Cytokine alterations, acute phase response signaling, and lipid metabolism are likely involved in pathogenesis of CE. Although there are insufficient current data to justify the use of proteomic biomarkers for assessment of CE in dogs, our study identifies potential candidates.
Collapse
Affiliation(s)
- Jane Yu
- Sydney School of Veterinary Science, Faculty of Science, The University of Sydney, Sydney, New South Wales 2006, Australia
| | - Craig Ruaux
- Sydney School of Veterinary Science, Faculty of Science, The University of Sydney, Sydney, New South Wales 2006, Australia
| | - Christine Griebsch
- Sydney School of Veterinary Science, Faculty of Science, The University of Sydney, Sydney, New South Wales 2006, Australia
| | - Lara Boland
- Sydney School of Veterinary Science, Faculty of Science, The University of Sydney, Sydney, New South Wales 2006, Australia
| | - Nadia Wong
- McIvor Road Veterinary Centre, Bendigo, Victoria, Australia
| | - Peter Bennett
- Sydney School of Veterinary Science, Faculty of Science, The University of Sydney, Sydney, New South Wales 2006, Australia
| | - Valerie C Wasinger
- Bioanalytical Mass Spectrometry Facility, Mark Wainwright Analytical Centre, University of New South Wales, Sydney, New South Wales, Australia
| |
Collapse
|
13
|
Zhang X, Liu J, Wang H. The cGAS-STING-autophagy pathway: Novel perspectives in neurotoxicity induced by manganese exposure. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2022; 315:120412. [PMID: 36240967 DOI: 10.1016/j.envpol.2022.120412] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Revised: 08/28/2022] [Accepted: 10/07/2022] [Indexed: 06/16/2023]
Abstract
Chronic high-level heavy metal exposure increases the risk of developing different neurodegenerative diseases. Chronic excessive manganese (Mn) exposure is known to lead to neurodegenerative diseases. In addition, some evidence suggests that autophagy dysfunction plays an important role in the pathogenesis of various neurodegenerative diseases. Over the past decade, the DNA-sensing receptor cyclic GMP-AMP synthase (cGAS) and its downstream signal-efficient interferon gene stimulator (STING), as well as the molecular composition and regulatory mechanisms of this pathway have been well understood. The cGAS-STING pathway has emerged as a crucial mechanism to induce effective innate immune responses by inducing type I interferons in mammalian cells. Moreover, recent studies have found that Mn2+ is the second activator of the cGAS-STING pathway besides dsDNA, and inducing autophagy is a primitive function for the activation of the cGAS-STING pathway. However, overactivation of the immune response can lead to tissue damage. This review discusses the mechanism of neurotoxicity induced by Mn exposure from the cGAS-STING-autophagy pathway. Future work exploiting the cGAS-STING-autophagy pathway may provide a novel perspective for manganese neurotoxicity.
Collapse
Affiliation(s)
- Xin Zhang
- Department of Toxicology, School of Public Health, Lanzhou University, Lanzhou, Gansu, China
| | - Jingjing Liu
- Department of Toxicology, School of Public Health, Lanzhou University, Lanzhou, Gansu, China
| | - Hui Wang
- Department of Toxicology, School of Public Health, Lanzhou University, Lanzhou, Gansu, China.
| |
Collapse
|
14
|
The Impacts of Iron Overload and Ferroptosis on Intestinal Mucosal Homeostasis and Inflammation. Int J Mol Sci 2022; 23:ijms232214195. [PMID: 36430673 PMCID: PMC9697168 DOI: 10.3390/ijms232214195] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Revised: 11/12/2022] [Accepted: 11/15/2022] [Indexed: 11/18/2022] Open
Abstract
Intestinal homeostasis is maintained through the interplay of the intestinal mucosa, local and systemic immune factors, and the microbial content of the gut. Iron is a trace mineral in most organisms, including humans, which is essential for growth, systemic metabolism and immune response. Paradoxically, excessive iron intake and/or high iron status can be detrimental to iron metabolism in the intestine and lead to iron overload and ferroptosis-programmed cell death mediated by iron-dependent lipid peroxidation within cell membranes, which contributes to several intestinal diseases. In this review, we comprehensively review recent findings on the impacts of iron overload and ferroptosis on intestinal mucosal homeostasis and inflammation and then present the progress of iron overload and ferroptosis-targeting therapy in intestinal diseases. Understanding the involved mechanisms can provide a new understanding of intestinal disease pathogenesis and facilitate advanced preventive and therapeutic strategies for intestinal dysfunction and diseases.
Collapse
|
15
|
Andreani C, Bartolacci C, Scaglioni PP. Ferroptosis: A Specific Vulnerability of RAS-Driven Cancers? Front Oncol 2022; 12:923915. [PMID: 35912247 PMCID: PMC9337859 DOI: 10.3389/fonc.2022.923915] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2022] [Accepted: 05/03/2022] [Indexed: 12/11/2022] Open
Abstract
Ferroptosis has emerged as a new type of programmed cell death that can be harnessed for cancer therapy. The concept of ferroptosis was for the first time proposed in in the early 2000s, as an iron-dependent mode of regulated cell death caused by unrestricted lipid peroxidation (LPO) and subsequent plasma membrane rupture. Since the discovery and characterization of ferroptosis, a wealth of research has improved our understanding of the main pathways regulating this process, leading to both the repurposing and the development of small molecules. However, ferroptosis is still little understood and several aspects remain to be investigated. For instance, it is unclear whether specific oncogenes, cells of origin or tumor niches impose specific susceptibility/resistance to ferroptosis or if there are some ferroptosis-related genes that may be used as bona fide pan-cancer targetable dependencies. In this context, even though RAS-driven cancer cell lines seemed to be selectively sensitive to ferroptosis inducers, subsequent studies have questioned these results, indicating that in some cases mutant RAS is necessary, but not sufficient to induce ferroptosis. In this perspective, based on publicly available genomic screening data and the literature, we discuss the relationship between RAS-mutation and ferroptosis susceptibility in cancer.
Collapse
Affiliation(s)
| | | | - Pier Paolo Scaglioni
- Department of Internal Medicine, University of Cincinnati College of Medicine, Cincinnati, OH, United States
| |
Collapse
|
16
|
Das BK, Wang L, Fujiwara T, Zhou J, Aykin-Burns N, Krager KJ, Lan R, Mackintosh SG, Edmondson R, Jennings ML, Wang X, Feng JQ, Barrientos T, Gogoi J, Kannan A, Gao L, Xing W, Mohan S, Zhao H. Transferrin receptor 1-mediated iron uptake regulates bone mass in mice via osteoclast mitochondria and cytoskeleton. eLife 2022; 11:73539. [PMID: 35758636 PMCID: PMC9352353 DOI: 10.7554/elife.73539] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2021] [Accepted: 06/25/2022] [Indexed: 11/13/2022] Open
Abstract
Increased intracellular iron spurs mitochondrial biogenesis and respiration to satisfy high-energy demand during osteoclast differentiation and bone-resorbing activities. Transferrin receptor 1 (Tfr1) mediates cellular iron uptake through endocytosis of iron-loaded transferrin, and its expression increases during osteoclast differentiation. Nonetheless, the precise functions of Tfr1 and Tfr1-mediated iron uptake in osteoclast biology and skeletal homeostasis remain incompletely understood. To investigate the role of Tfr1 in osteoclast lineage cells in vivo and in vitro, we crossed Tfrc (encoding Tfr1)-floxed mice with Lyz2 (LysM)-Cre and Cathepsin K (Ctsk)-Cre mice to generate Tfrc conditional knockout mice in myeloid osteoclast precursors (Tfr1ΔLysM) or differentiated osteoclasts (Tfr1ΔCtsk), respectively. Skeletal phenotyping by µCT and histology unveiled a significant increase in trabecular bone mass with normal osteoclast number in long bones of 10-week-old young and 6-month-old adult female but not male Tfr1ΔLysM mice. Although high trabecular bone volume in long bones was observed in both male and female Tfr1ΔCtsk mice, this phenotype was more pronounced in female knockout mice. Consistent with this gender-dependent phenomena, estrogen deficiency induced by ovariectomy decreased trabecular bone mass in Tfr1ΔLysM mice. Mechanistically, disruption of Tfr1 expression attenuated mitochondrial metabolism and cytoskeletal organization in mature osteoclasts in vitro by attenuating mitochondrial respiration and activation of the Src-Rac1-WAVE regulatory complex axis, respectively, leading to decreased bone resorption with little impact on osteoclast differentiation. These results indicate that Tfr1-mediated iron uptake is specifically required for osteoclast function and is indispensable for bone remodeling in a gender-dependent manner.
Collapse
Affiliation(s)
- Bhaba K Das
- Long Beach VA Healthcare System, Southern California Institute for Research and Education, Long Beach, United States
| | - Lei Wang
- Department of Orthopedics, Anhui Medical University, Hefei, China
| | - Toshifumi Fujiwara
- Department of Orthopedic Surgery, Kyushu University Hospital, Fukuoka, Japan
| | - Jian Zhou
- Department of Orthopedics, Anhui Medical University, HeFei, China
| | - Nukhet Aykin-Burns
- Department of Pharmaceutical Sciences, University of Arkansas for Medical Sciences, Little Rock, United States
| | - Kimberly J Krager
- Department of Pharmaceutical Sciences, University of Arkansas for Medical Sciences, Little Rock, United States
| | - Renny Lan
- Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, United States
| | - Samuel G Mackintosh
- Department of Biochemistry and Molecular Biology, University of Arkansas for Medical Sciences, Little Rock, United States
| | - Ricky Edmondson
- Department of Biochemistry and Molecular Biology, University of Arkansas for Medical Sciences, Little Rock, United States
| | - Michael L Jennings
- Department of Physiology and Cell Biology, University of Arkansas for Medical Sciences, Little Rock, United States
| | - Xiaofang Wang
- Department of Biomedical Sciences, Texas A&M University, Dallas, United States
| | - Jian Q Feng
- Department of Biomedical Sciences, Texas A&M University, Dallas, United States
| | | | - Jyoti Gogoi
- Long Beach VA Healthcare System, Southern California Institute for Research and Education, Long Beach, United States
| | - Aarthi Kannan
- Long Beach VA Healthcare System, Southern California Institute for Research and Education, Long Beach, United States
| | - Ling Gao
- Long Beach VA Healthcare System, Southern California Institute for Research and Education, Long Beach, United States
| | - Weirong Xing
- Musculoskeletal Disease Center, VA Loma Linda Healthcare System, Loma Linda, United States
| | - Subburaman Mohan
- Musculoskeletal Disease Center, VA Loma Linda Healthcare System, Loma Linda, United States
| | - Haibo Zhao
- Long Beach VA Healthcare System, Southern California Institute for Research and Education, Long Beach, United States
| |
Collapse
|
17
|
Zhang N, Wang J, Bing T, Liu X, Shangguan D. Transferrin receptor-mediated internalization and intracellular fate of conjugates of a DNA aptamer. MOLECULAR THERAPY. NUCLEIC ACIDS 2022; 27:1249-1259. [PMID: 35282414 PMCID: PMC8899136 DOI: 10.1016/j.omtn.2022.02.006] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/18/2021] [Accepted: 02/10/2022] [Indexed: 02/06/2023]
Abstract
Aptamers have excellent specificity and affinity in targeting cell surface receptors, showing great potential in targeted delivery of drugs, siRNA, mRNA, and various nanomaterials with therapeutic function. A better insight of the receptor-mediated internalization process of aptameric conjugates could facilitate the design of new targeted drugs. In this paper, human transferrin receptor-targeted DNA aptamer (termed HG1-9)-fluorophore conjugates were synthesized to visualize the internalization, intracellular transport, and nano-environmental pH of aptameric conjugates. Unlike transferrin that showed high recycling rate and short duration time in cells, the synthetic aptameric conjugates continuously accumulated within cells at a relatively slower rate, besides recycling back to cell surface. After long incubation (≥2 h), only very small amounts of HG1-9 conjugates (approximately 5%) entered late endosomes or lysosomes, and more than 90% of internalized HG1-9 was retained in cellular vesicles (pH 6.0–6.8), escaping from degradation. And among the internalized HG1-9 conjugates, approximately 20% was dissociated from transferrin receptor. The lower recycling ratios of HG1-9 conjugates and their dissociation from receptors promote the accurate and efficient release of their loaded drugs. These results suggest that aptamer HG1-9 could be provided as a versatile tool for specific and effective delivery of diverse therapeutic payloads.
Collapse
Affiliation(s)
- Nan Zhang
- Beijing National Laboratory for Molecular Sciences, Key Laboratory of Analytical Chemistry for Living Biosystems, CAS Research/Education Center for Excellence in Molecular Sciences, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, China
| | - Junyan Wang
- Beijing National Laboratory for Molecular Sciences, Key Laboratory of Analytical Chemistry for Living Biosystems, CAS Research/Education Center for Excellence in Molecular Sciences, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, China
| | - Tao Bing
- Beijing National Laboratory for Molecular Sciences, Key Laboratory of Analytical Chemistry for Living Biosystems, CAS Research/Education Center for Excellence in Molecular Sciences, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, China.,School of Chemical Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Xiangjun Liu
- Beijing National Laboratory for Molecular Sciences, Key Laboratory of Analytical Chemistry for Living Biosystems, CAS Research/Education Center for Excellence in Molecular Sciences, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, China.,School of Chemical Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Dihua Shangguan
- Beijing National Laboratory for Molecular Sciences, Key Laboratory of Analytical Chemistry for Living Biosystems, CAS Research/Education Center for Excellence in Molecular Sciences, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, China.,School of Chemical Sciences, University of Chinese Academy of Sciences, Beijing 100049, China.,School of Molecular Medicine, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou 310024, China
| |
Collapse
|
18
|
Pearce SC, Suntornsaratoon P, Kishida K, Al‐Jawadi A, Guardia J, Nadler I, Flores J, Shiarella R, Auvinen M, Yu S, Gao N, Ferraris RP. Expression of SARS-CoV-2 entry factors, electrolyte, and mineral transporters in different mouse intestinal epithelial cell types. Physiol Rep 2021; 9:e15061. [PMID: 34755492 PMCID: PMC8578880 DOI: 10.14814/phy2.15061] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2021] [Revised: 09/03/2021] [Accepted: 09/13/2021] [Indexed: 11/24/2022] Open
Abstract
Angiotensin-converting enzyme 2 (ACE2) and transmembrane proteases (TMPRSS) are multifunctional proteins required for SARS-CoV-2 infection or for amino acid (AA) transport, and are abundantly expressed in mammalian small intestine, but the identity of the intestinal cell type(s) and sites of expression are unclear. Here we determined expression of SARS-CoV-2 entry factors in different cell types and then compared it to that of representative AA, electrolyte, and mineral transporters. We tested the hypothesis that SARS-CoV-2, AA, electrolyte, and mineral transporters are expressed heterogeneously in different intestinal cell types by making mouse enteroids enriched in enterocytes (ENT), goblet (GOB), Paneth (PAN), or stem (ISC) cells. Interestingly, the expression of ACE2 was apical and modestly greater in ENT, the same pattern observed for its associated AA transporters B0 AT1 and SIT1. TMPRSS2 and TMPRSS4 were more highly expressed in crypt-residing ISC. Expression of electrolyte transporters was dramatically heterogeneous. DRA, NBCe1, and NHE3 were greatest in ENT, while those of CFTR and NKCC1 that play important roles in secretory diarrhea, were mainly expressed in ISC and PAN that also displayed immunohistochemically abundant basolateral NKCC1. Intestinal iron transporters were generally expressed higher in ENT and GOB, while calcium transporters were expressed mainly in PAN. Heterogeneous expression of its entry factors suggests that the ability of SARS-CoV-2 to infect the intestine may vary with cell type. Parallel cell-type expression patterns of ACE2 with B0 AT1 and SIT1 provides further evidence of ACE2's multifunctional properties and importance in AA absorption.
Collapse
Affiliation(s)
- Sarah C. Pearce
- Department of Pharmacology, Physiology and NeurosciencesNew Jersey Medical SchoolRutgers UniversityNewarkNew JerseyUSA
- Present address:
USDA‐ARS National Laboratory for Agriculture and the Environment1015 N. University Blvd.AmesIowa50011USA
| | - Panan Suntornsaratoon
- Department of Pharmacology, Physiology and NeurosciencesNew Jersey Medical SchoolRutgers UniversityNewarkNew JerseyUSA
- Department of Physiology, Faculty of ScienceMahidol UniversityBangkokThailand
| | - Kunihiro Kishida
- Department of Pharmacology, Physiology and NeurosciencesNew Jersey Medical SchoolRutgers UniversityNewarkNew JerseyUSA
- Present address:
Department of Science and Technology on Food SafetyKindai UniversityWakayama649‐6493Japan
| | - Arwa Al‐Jawadi
- Department of Pharmacology, Physiology and NeurosciencesNew Jersey Medical SchoolRutgers UniversityNewarkNew JerseyUSA
- Present address:
Thermo Fisher Scientific5823 Newton DriveCarlsbadCalifornia92008USA
| | - Joshua Guardia
- Department of Pharmacology, Physiology and NeurosciencesNew Jersey Medical SchoolRutgers UniversityNewarkNew JerseyUSA
| | - Ian Nadler
- Department of Pharmacology, Physiology and NeurosciencesNew Jersey Medical SchoolRutgers UniversityNewarkNew JerseyUSA
| | - Juan Flores
- Department of Biological SciencesLife Science CenterRutgers UniversityNewarkNew JerseyUSA
| | - Reilly Shiarella
- Department of Pharmacology, Physiology and NeurosciencesNew Jersey Medical SchoolRutgers UniversityNewarkNew JerseyUSA
| | - Madelyn Auvinen
- Department of Pharmacology, Physiology and NeurosciencesNew Jersey Medical SchoolRutgers UniversityNewarkNew JerseyUSA
| | - Shiyan Yu
- Department of Biological SciencesLife Science CenterRutgers UniversityNewarkNew JerseyUSA
| | - Nan Gao
- Department of Biological SciencesLife Science CenterRutgers UniversityNewarkNew JerseyUSA
| | - Ronaldo P. Ferraris
- Department of Pharmacology, Physiology and NeurosciencesNew Jersey Medical SchoolRutgers UniversityNewarkNew JerseyUSA
| |
Collapse
|
19
|
Phoaubon S, Lertsuwan K, Teerapornpuntakit J, Charoenphandhu N. Hepcidin induces intestinal calcium uptake while suppressing iron uptake in Caco-2 cells. PLoS One 2021; 16:e0258433. [PMID: 34644351 PMCID: PMC8513844 DOI: 10.1371/journal.pone.0258433] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2021] [Accepted: 09/28/2021] [Indexed: 11/30/2022] Open
Abstract
Abnormal calcium absorption and iron overload from iron hyperabsorption can contribute to osteoporosis as found in several diseases, including hemochromatosis and thalassemia. Previous studies in thalassemic mice showed the positive effects of the iron uptake suppressor, hepcidin, on calcium transport. However, whether this effect could be replicated in other conditions is not known. Therefore, this study aimed to investigate the effects of hepcidin on iron and calcium uptake ability under physiological, iron uptake stimulation and calcium uptake suppression. To investigate the potential mechanism, effects of hepcidin on the expression of iron and calcium transporter and transport-associated protein in Caco-2 cells were also determined. Our results showed that intestinal cell iron uptake was significantly increased by ascorbic acid together with ferric ammonium citrate (FAC), but this phenomenon was suppressed by hepcidin. Interestingly, hepcidin significantly increased calcium uptake under physiological condition but not under iron uptake stimulation. While hepcidin significantly suppressed the expression of iron transporter, it had no effect on calcium transporter expression. This indicated that hepcidin-induced intestinal cell calcium uptake did not occur through the stimulation of calcium transporter expression. On the other hand, 1,25(OH)2D3 effectively induced intestinal cell calcium uptake, but it did not affect intestinal cell iron uptake or iron transporter expression. The 1,25(OH)2D3-induced intestinal cell calcium uptake was abolished by 12 mM CaCl2; however, hepcidin could not rescue intestinal cell calcium uptake suppression by CaCl2. Taken together, our results showed that hepcidin could effectively and concurrently induce intestinal cell calcium uptake while reducing intestinal cell iron uptake under physiological and iron uptake stimulation conditions, suggesting its therapeutic potential for inactive calcium absorption, particularly in thalassemic patients or patients who did not adequately respond to 1,25(OH)2D3.
Collapse
Affiliation(s)
- Supathra Phoaubon
- Doctor of Philosophy Program in Biochemistry (International Program), Faculty of Science, Mahidol University, Bangkok, Thailand
- Department of Biochemistry, Faculty of Science, Mahidol University, Bangkok, Thailand
| | - Kornkamon Lertsuwan
- Department of Biochemistry, Faculty of Science, Mahidol University, Bangkok, Thailand
- Center of Calcium and Bone Research (COCAB), Faculty of Science, Mahidol University, Bangkok, Thailand
- * E-mail:
| | - Jarinthorn Teerapornpuntakit
- Center of Calcium and Bone Research (COCAB), Faculty of Science, Mahidol University, Bangkok, Thailand
- Department of Physiology, Faculty of Medical Science, Naresuan University, Phitsanulok, Thailand
| | - Narattaphol Charoenphandhu
- Center of Calcium and Bone Research (COCAB), Faculty of Science, Mahidol University, Bangkok, Thailand
- Department of Physiology, Faculty of Science, Mahidol University, Bangkok, Thailand
- Institute of Molecular Biosciences, Mahidol University, Nakhon Pathom, Thailand
- The Academy of Science, The Royal Society of Thailand, Dusit, Bangkok, Thailand
| |
Collapse
|
20
|
Sinha S, Pereira-Reis J, Guerra A, Rivella S, Duarte D. The Role of Iron in Benign and Malignant Hematopoiesis. Antioxid Redox Signal 2021; 35:415-432. [PMID: 33231101 PMCID: PMC8328043 DOI: 10.1089/ars.2020.8155] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Revised: 10/26/2020] [Accepted: 11/20/2020] [Indexed: 12/21/2022]
Abstract
Significance: Iron is an essential element required for sustaining a normal healthy life. However, an excess amount of iron in the bloodstream and tissue generates toxic hydroxyl radicals through Fenton reactions. Henceforth, a balance in iron concentration is extremely important to maintain cellular homeostasis in both normal hematopoiesis and erythropoiesis. Iron deficiency or iron overload can impact hematopoiesis and is associated with many hematological diseases. Recent Advances: The mechanisms of action of key iron regulators such as erythroferrone and the discovery of new drugs, such as ACE-536/luspatercept, are of potential interest to treat hematological disorders, such as β-thalassemia. New therapies targeting inflammation-induced ineffective erythropoiesis are also in progress. Furthermore, emerging evidences support differential interactions between iron and its cellular antioxidant responses of hematopoietic and neighboring stromal cells. Both iron and its systemic regulator, such as hepcidin, play a significant role in regulating erythropoiesis. Critical Issues: Significant pre-clinical studies are on the way and new drugs targeting iron metabolism have been recently approved or are undergoing clinical trials to treat pathological conditions with impaired erythropoiesis such as myelodysplastic syndromes or β-thalassemia. Future Directions: Future studies should explore how iron regulates hematopoiesis in both benign and malignant conditions. Antioxid. Redox Signal. 35, 415-432.
Collapse
Affiliation(s)
- Sayantani Sinha
- Division of Hematology, Department of Pediatrics, The Children's Hospital of Philadelphia (CHOP), Philadelphia, Pennsylvania, USA
| | - Joana Pereira-Reis
- Instituto de Investigação e Inovação em Saúde (i3S), Universidade do Porto, Porto, Portugal
| | - Amaliris Guerra
- Division of Hematology, Department of Pediatrics, The Children's Hospital of Philadelphia (CHOP), Philadelphia, Pennsylvania, USA
| | - Stefano Rivella
- Division of Hematology, Department of Pediatrics, The Children's Hospital of Philadelphia (CHOP), Philadelphia, Pennsylvania, USA
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Cell and Molecular Biology Affinity Group (CAMB), University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Raymond G. Perelman Center for Cellular and Molecular Therapeutics, The Children's Hospital of Philadelphia (CHOP), Philadelphia, Pennsylvania, USA
- Penn Center for Musculoskeletal Disorders, The Children's Hospital of Philadelphia (CHOP), Philadelphia, Pennsylvania, USA
| | - Delfim Duarte
- Instituto de Investigação e Inovação em Saúde (i3S), Universidade do Porto, Porto, Portugal
- Department of Onco-Hematology, Instituto Português de Oncologia (IPO), Porto, Portugal
- Unit of Biochemistry, Department of Biomedicine, Faculdade de Medicina da Universidade do Porto (FMUP), Porto, Portugal
- Porto Comprehensive Cancer Center (P.CCC), Porto, Portugal
| |
Collapse
|
21
|
Zhang Z, Funcke JB, Zi Z, Zhao S, Straub LG, Zhu Y, Zhu Q, Crewe C, An YA, Chen S, Li N, Wang MY, Ghaben AL, Lee C, Gautron L, Engelking LJ, Raj P, Deng Y, Gordillo R, Kusminski CM, Scherer PE. Adipocyte iron levels impinge on a fat-gut crosstalk to regulate intestinal lipid absorption and mediate protection from obesity. Cell Metab 2021; 33:1624-1639.e9. [PMID: 34174197 PMCID: PMC8338877 DOI: 10.1016/j.cmet.2021.06.001] [Citation(s) in RCA: 67] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/21/2020] [Revised: 04/06/2021] [Accepted: 06/02/2021] [Indexed: 02/07/2023]
Abstract
Iron overload is positively associated with diabetes risk. However, the role of iron in adipose tissue remains incompletely understood. Here, we report that transferrin-receptor-1-mediated iron uptake is differentially required for distinct subtypes of adipocytes. Notably, adipocyte-specific transferrin receptor 1 deficiency substantially protects mice from high-fat-diet-induced metabolic disorders. Mechanistically, low cellular iron levels have a positive impact on the health of the white adipose tissue and can restrict lipid absorption from the intestine through modulation of vesicular transport in enterocytes following high-fat diet feeding. Specific reduction of adipocyte iron by AAV-mediated overexpression of the iron exporter Ferroportin1 in adult mice effectively mimics these protective effects. In summary, our studies highlight an important role of adipocyte iron in the maintenance of systemic metabolism through an adipocyte-enterocyte axis, offering an additional level of control over caloric influx into the system after feeding by regulating intestinal lipid absorption.
Collapse
Affiliation(s)
- Zhuzhen Zhang
- Touchstone Diabetes Center, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Jan-Bernd Funcke
- Touchstone Diabetes Center, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Zhenzhen Zi
- Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Shangang Zhao
- Touchstone Diabetes Center, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Leon G Straub
- Touchstone Diabetes Center, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Yi Zhu
- Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA
| | - Qingzhang Zhu
- Touchstone Diabetes Center, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Clair Crewe
- Touchstone Diabetes Center, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Yu A An
- Touchstone Diabetes Center, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Shiuhwei Chen
- Touchstone Diabetes Center, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Na Li
- Touchstone Diabetes Center, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - May-Yun Wang
- Touchstone Diabetes Center, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Alexandra L Ghaben
- Touchstone Diabetes Center, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Charlotte Lee
- Center for Hypothalamic Research, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Laurent Gautron
- Center for Hypothalamic Research, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Luke J Engelking
- Department of Molecular Genetics, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Prithvi Raj
- Department of Immunology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Yingfeng Deng
- Touchstone Diabetes Center, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Ruth Gordillo
- Touchstone Diabetes Center, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Christine M Kusminski
- Touchstone Diabetes Center, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Philipp E Scherer
- Touchstone Diabetes Center, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA; Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas, TX, USA.
| |
Collapse
|
22
|
Bevacqua E, Curcio M, Saletta F, Vittorio O, Cirillo G, Tucci P. Dextran-Curcumin Nanosystems Inhibit Cell Growth and Migration Regulating the Epithelial to Mesenchymal Transition in Prostate Cancer Cells. Int J Mol Sci 2021; 22:7013. [PMID: 34209825 PMCID: PMC8269310 DOI: 10.3390/ijms22137013] [Citation(s) in RCA: 7] [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: 05/20/2021] [Revised: 06/21/2021] [Accepted: 06/25/2021] [Indexed: 12/16/2022] Open
Abstract
Functional nanocarriers which are able to simultaneously vectorize drugs to the site of interest and exert their own cytotoxic activity represent a significant breakthrough in the search for effective anticancer strategies with fewer side effects than conventional chemotherapeutics. Here, we propose previously developed, self-assembling dextran-curcumin nanoparticles for the treatment of prostate cancer in combination therapy with Doxorubicin (DOXO). Biological effectiveness was investigated by evaluating the cell viability in either cancer and normal cells, reactive oxygen species (ROS) production, apoptotic effect, interference with the cell cycle, and the ability to inhibit cell migration and reverse the epithelial to mesenchymal transition (EMT). The results proved a significant enhancement of curcumin efficiency upon immobilization in nanoparticles: IC50 reduced by a half, induction of apoptotic effect, and improved ROS production (from 67 to 134%) at low concentrations. Nanoparticles guaranteed a pH-dependent DOXO release, with a more efficient release in acidic environments. Finally, a synergistic effect between nanoparticles and Doxorubicin was demonstrated, with the free curcumin showing additive activity. Although in vivo studies are required to support the findings of this study, these preliminary in vitro data can be considered a proof of principle for the design of an effective therapy for prostate cancer treatment.
Collapse
Affiliation(s)
- Emilia Bevacqua
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036 Rende, Italy; (E.B.); (M.C.); (G.C.)
| | - Manuela Curcio
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036 Rende, Italy; (E.B.); (M.C.); (G.C.)
| | - Federica Saletta
- Lowy Cancer Research Centre, Children’s Cancer Institute, University of New South Wales, High Street, Randwick, NSW 2052, Australia; (F.S.); (O.V.)
- School of Women’s and Children’s Health, University of New South Wales, Kensington, NSW 2052, Australia
| | - Orazio Vittorio
- Lowy Cancer Research Centre, Children’s Cancer Institute, University of New South Wales, High Street, Randwick, NSW 2052, Australia; (F.S.); (O.V.)
- School of Women’s and Children’s Health, University of New South Wales, Kensington, NSW 2052, Australia
- ARC Centre of Excellence for Convergent BioNano Science and Technology, Australian Centre for NanoMedicine, University of New South Wales, Kensington, NSW 2052, Australia
| | - Giuseppe Cirillo
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036 Rende, Italy; (E.B.); (M.C.); (G.C.)
| | - Paola Tucci
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036 Rende, Italy; (E.B.); (M.C.); (G.C.)
| |
Collapse
|
23
|
Corral VM, Schultz ER, Eisenstein RS, Connell GJ. Roquin is a major mediator of iron-regulated changes to transferrin receptor-1 mRNA stability. iScience 2021; 24:102360. [PMID: 33898949 PMCID: PMC8058555 DOI: 10.1016/j.isci.2021.102360] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Revised: 09/18/2020] [Accepted: 03/23/2021] [Indexed: 11/21/2022] Open
Abstract
Transferrin receptor-1 (TfR1) has essential iron transport and proposed signal transduction functions. Proper TfR1 regulation is a requirement for hematopoiesis, neurological development, and the homeostasis of tissues including the intestine and muscle, while dysregulation is associated with cancers and immunodeficiency. TfR1 mRNA degradation is highly regulated, but the identity of the degradation activity remains uncertain. Here, we show with gene knockouts and siRNA knockdowns that two Roquin paralogs are major mediators of iron-regulated changes to the steady-state TfR1 mRNA level within four different cell types (HAP1, HUVEC, L-M, and MEF). Roquin is demonstrated to destabilize the TfR1 mRNA, and its activity is fully dependent on three hairpin loops within the TfR1 mRNA 3′-UTR that are essential for iron-regulated instability. We further show in L-M cells that TfR1 mRNA degradation does not require ongoing translation, consistent with Roquin-mediated instability. We conclude that Roquin is a major effector of TfR1 mRNA abundance. Roquin is a major mediator of iron-regulated TfR1 mRNA instability Roquin-mediated instability requires three stem loops within the TfR1 3′-UTR Iron-regulated TfR1 mRNA instability can occur in the absence of Regnase-1
Collapse
Affiliation(s)
- Victor M Corral
- Department of Pharmacology, University of Minnesota, Minneapolis, MN 55455, USA
| | - Eric R Schultz
- Department of Pharmacology, University of Minnesota, Minneapolis, MN 55455, USA
| | - Richard S Eisenstein
- Department of Nutritional Sciences, University of Wisconsin, Madison, WI 53706, USA
| | - Gregory J Connell
- Department of Pharmacology, University of Minnesota, Minneapolis, MN 55455, USA
| |
Collapse
|
24
|
Chatterton DEW, Aagaard S, Hesselballe Hansen T, Nguyen DN, De Gobba C, Lametsch R, Sangild PT. Bioactive proteins in bovine colostrum and effects of heating, drying and irradiation. Food Funct 2021; 11:2309-2327. [PMID: 32108849 DOI: 10.1039/c9fo02998b] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Bovine colostrum (BC) contains bioactive proteins, such as immunoglobulin G (IgG), lactoferrin (LF) and lactoperoxidase (LP). BC was subjected to low-temperature, long-time pasteurization (LTLT, 63 °C, 30 min) or high-temperature, short-time pasteurization (HTST, 72 °C, 15 s) and spray-drying (SD), with or without γ-irradiation (GI, ∼14 kGy) to remove microbial contamination. Relative to unpasteurized liquid BC, SD plus GI increased protein denaturation by 6 and 11%, respectively, increasing to 19 and 27% after LTLT and to 48% after HTST, with no further effects after GI (all P < 0.05). LTLT, without or with GI, resulted in 15 or 29% denaturation of IgG, compared with non-pasteurized BC, and 34 or 58% for HTST treatment (all P < 0.05, except LTLT without GI). For IgG, only GI, not SD or LTLT, increased denaturation (30-38%, P < 0.05) but HTST increased denaturation to 40%, with further increases after GI (60%, P < 0.05). LTLT and HTST reduced LP levels (56 and 81% respectively) and LTLT reduced LF levels (21%), especially together with GI (47%, P < 0.05). Denaturation of BSA, β-LgA, β-LgB and α-La were similar to IgG. Methionine, a protective amino acid against free oxygen radicals, was oxidised by LTLT + GI (P < 0.05) while LTLT and HTST had no effect. Many anti-inflammatory proteins, including serpin anti-proteinases were highly sensitive to HTST and GI but preserved after LTLT pasteurization. LTLT, followed by SD is an optimal processing technique preserving bioactive proteins when powdered BC is used as a diet supplement for sensitive patients.
Collapse
Affiliation(s)
| | - Sasha Aagaard
- Department of Food Science, University of Copenhagen, DK-1958, Denmark. and Comparative Pediatrics and Nutrition, Department of Veterinary Clinical and Animal Sciences, Denmark
| | | | - Duc Ninh Nguyen
- Comparative Pediatrics and Nutrition, Department of Veterinary Clinical and Animal Sciences, Denmark
| | - Cristian De Gobba
- Department of Food Science, University of Copenhagen, DK-1958, Denmark.
| | - René Lametsch
- Department of Food Science, University of Copenhagen, DK-1958, Denmark.
| | - Per T Sangild
- Comparative Pediatrics and Nutrition, Department of Veterinary Clinical and Animal Sciences, Denmark
| |
Collapse
|
25
|
Mleczko‐Sanecka K, Silvestri L. Cell-type-specific insights into iron regulatory processes. Am J Hematol 2021; 96:110-127. [PMID: 32945012 DOI: 10.1002/ajh.26001] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Revised: 08/20/2020] [Accepted: 09/14/2020] [Indexed: 12/16/2022]
Abstract
Despite its essential role in many biological processes, iron is toxic when in excess due to its propensity to generate reactive oxygen species. To prevent diseases associated with iron deficiency or iron loading, iron homeostasis must be tightly controlled. Intracellular iron content is regulated by the Iron Regulatory Element-Iron Regulatory Protein (IRE-IRP) system, whereas systemic iron availability is adjusted to body iron needs chiefly by the hepcidin-ferroportin (FPN) axis. Here, we aimed to review advances in the field that shed light on cell-type-specific regulatory mechanisms that control or modify systemic and local iron balance, and how shifts in cellular iron levels may affect specialized cell functions.
Collapse
Affiliation(s)
| | - Laura Silvestri
- Regulation of Iron Metabolism Unit, Division of Genetics and Cell Biology IRCCS San Raffaele Scientific Institute Milan Italy
- Vita‐Salute San Raffaele University Milan Italy
| |
Collapse
|
26
|
Li Y, Cheng JX, Yang HH, Chen LP, Liu FJ, Wu Y, Fan M, Wu HT. Transferrin receptor 1 plays an important role in muscle development and denervation-induced muscular atrophy. Neural Regen Res 2021; 16:1308-1316. [PMID: 33318410 PMCID: PMC8284266 DOI: 10.4103/1673-5374.301024] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Previous studies demonstrate an accumulation of transferrin and transferrin receptor 1 (TfR1) in regenerating peripheral nerves. However, the expression and function of transferrin and TfR1 in the denervated skeletal muscle remain poorly understood. In this study, a mouse model of denervation was produced by complete tear of the left brachial plexus nerve. RNA-sequencing revealed that transferrin expression in the denervated skeletal muscle was upregulated, while TfR1 expression was downregulated. We also investigated the function of TfR1 during development and in adult skeletal muscles in mice with inducible deletion or loss of TfR1. The ablation of TfR1 in skeletal muscle in early development caused severe muscular atrophy and early death. In comparison, deletion of TfR1 in adult skeletal muscles did not affect survival or glucose metabolism, but caused skeletal muscle atrophy and motor functional impairment, similar to the muscular atrophy phenotype observed after denervation. These findings suggest that TfR1 plays an important role in muscle development and denervation-induced muscular atrophy. This study was approved by the Institutional Animal Care and Use Committee of Beijing Institute of Basic Medical Sciences, China (approval No. SYXK 2017-C023) on June 1, 2018.
Collapse
Affiliation(s)
- Ying Li
- Department of Neurobiology, Beijing Institute of Basic Medical Sciences, Beijing, China
| | - Juan-Xian Cheng
- Department of Neurobiology, Beijing Institute of Basic Medical Sciences, Beijing, China
| | - Hai-Hong Yang
- Department of Neurobiology, Beijing Institute of Basic Medical Sciences, Beijing; Department of Anesthesiology, the General Hospital of Western Theater Command, Chengdu, Sichuan Province, China
| | - Li-Ping Chen
- Department of Neurobiology, Beijing Institute of Basic Medical Sciences, Beijing, China
| | - Feng-Jiao Liu
- Department of Neurobiology, Beijing Institute of Basic Medical Sciences, Beijing, China
| | - Yan Wu
- Department of Neurobiology, Beijing Institute of Basic Medical Sciences, Beijing, China
| | - Ming Fan
- Department of Neurobiology, Beijing Institute of Basic Medical Sciences; Chinese Institute for Brain Research (CIBR), Beijing, China
| | - Hai-Tao Wu
- Department of Neurobiology, Beijing Institute of Basic Medical Sciences; Chinese Institute for Brain Research (CIBR), Beijing; Key Laboratory of Neuroregeneration, Coinnovation Center of Neuroregeneration, Nantong University, Nantong, Jiangsu Province, China
| |
Collapse
|
27
|
Jennifer B, Berg V, Modak M, Puck A, Seyerl-Jiresch M, Künig S, Zlabinger GJ, Steinberger P, Chou J, Geha RS, Öhler L, Yachie A, Choe H, Kraller M, Stockinger H, Stöckl J. Transferrin receptor 1 is a cellular receptor for human heme-albumin. Commun Biol 2020; 3:621. [PMID: 33110194 PMCID: PMC7591885 DOI: 10.1038/s42003-020-01294-5] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2019] [Accepted: 09/15/2020] [Indexed: 12/13/2022] Open
Abstract
Iron is essential for living cells. Uptake of iron-loaded transferrin by the transferrin receptor 1 (CD71, TFR) is a major but not sufficient mechanism and an alternative iron-loaded ligand for CD71 has been assumed. Here, we demonstrate that CD71 utilizes heme-albumin as cargo to transport iron into human cells. Binding and endocytosis of heme-albumin via CD71 was sufficient to promote proliferation of various cell types in the absence of transferrin. Growth and differentiation of cells induced by heme-albumin was dependent on heme-oxygenase 1 (HO-1) function and was accompanied with an increase of the intracellular labile iron pool (LIP). Import of heme-albumin via CD71 was further found to contribute to the efficacy of albumin-based drugs such as the chemotherapeutic Abraxane. Thus, heme-albumin/CD71 interaction is a novel route to transport nutrients or drugs into cells and adds to the emerging function of CD71 as a scavenger receptor. Brell, Berg et al find that iron enters cells not only through iron-transferrin uptake by the transferrin receptor (CD71) but also through uptake of heme-albumin by this receptor and that heme-albumin stimulates proliferation in a manner dependent on heme oxygenase 1. This study presents a new route for iron uptake in mammalian cells.
Collapse
Affiliation(s)
- Brell Jennifer
- Institute of Immunology, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, 1090, Vienna, Austria
| | - Verena Berg
- Institute of Immunology, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, 1090, Vienna, Austria
| | - Madhura Modak
- Institute of Immunology, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, 1090, Vienna, Austria
| | - Alexander Puck
- Institute of Immunology, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, 1090, Vienna, Austria
| | - Maria Seyerl-Jiresch
- Institute of Immunology, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, 1090, Vienna, Austria
| | - Sarojinidevi Künig
- Institute of Immunology, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, 1090, Vienna, Austria
| | - Gerhard J Zlabinger
- Institute of Immunology, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, 1090, Vienna, Austria
| | - Peter Steinberger
- Institute of Immunology, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, 1090, Vienna, Austria
| | - Janet Chou
- Division of Immunology, Boston Children´s Hospital, Boston, MA, 02115, USA
| | - Raif S Geha
- Division of Immunology, Boston Children´s Hospital, Boston, MA, 02115, USA
| | - Leopold Öhler
- Department of Internal Medicine, St. Josef Hospital, 1130, Vienna, Austria
| | - Akihiro Yachie
- Department of Pediatrics, School of Medicine, Institute of Medical, Pharmaceutical, and Health Sciences, Kanazawa University, Kanazawa, Japan
| | - Hyeryun Choe
- Department of Immunology and Microbiology, The Scripps Research Institute, Florida, CA, 92037, USA
| | - Markus Kraller
- Institute of Hygiene and Applied Immunology, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, 1090, Vienna, Austria
| | - Hannes Stockinger
- Institute of Hygiene and Applied Immunology, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, 1090, Vienna, Austria
| | - Johannes Stöckl
- Institute of Immunology, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, 1090, Vienna, Austria.
| |
Collapse
|
28
|
Chen X, Yu C, Kang R, Tang D. Iron Metabolism in Ferroptosis. Front Cell Dev Biol 2020; 8:590226. [PMID: 33117818 PMCID: PMC7575751 DOI: 10.3389/fcell.2020.590226] [Citation(s) in RCA: 572] [Impact Index Per Article: 114.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Accepted: 09/17/2020] [Indexed: 01/12/2023] Open
Abstract
Ferroptosis is a form of regulated cell death that is characterized by iron-dependent oxidative damage and subsequent plasma membrane ruptures and the release of damage-associated molecular patterns. Due to the role of iron in mediating the production of reactive oxygen species and enzyme activity in lipid peroxidation, ferroptosis is strictly controlled by regulators involved in many aspects of iron metabolism, such as iron uptake, storage, utilization, and efflux. Translational and transcriptional regulation of iron homeostasis provide an integrated network to determine the sensitivity of ferroptosis. Impaired ferroptosis is implicated in various iron-related pathological conditions or diseases, such as cancer, neurodegenerative diseases, and ischemia-reperfusion injury. Understanding the molecular mechanisms underlying the regulation of iron metabolism during ferroptosis may provide effective strategies for the treatment of ferroptosis-associated diseases. Indeed, iron chelators effectively prevent the occurrence of ferroptosis, which may provide new approaches for the treatment of iron-related disorders. In this review, we summarize recent advances in the theoretical modeling of iron-dependent ferroptosis, and highlight the therapeutic implications of iron chelators in diseases.
Collapse
Affiliation(s)
- Xin Chen
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Protein Modification and Degradation, The Third Affiliated Hospital, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, China.,Department of Surgery, UT Southwestern Medical Center, Dallas, TX, United States
| | - Chunhua Yu
- Department of Surgery, UT Southwestern Medical Center, Dallas, TX, United States
| | - Rui Kang
- Department of Surgery, UT Southwestern Medical Center, Dallas, TX, United States
| | - Daolin Tang
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Protein Modification and Degradation, The Third Affiliated Hospital, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, China.,Department of Surgery, UT Southwestern Medical Center, Dallas, TX, United States
| |
Collapse
|
29
|
Altamura S, Marques O, Colucci S, Mertens C, Alikhanyan K, Muckenthaler MU. Regulation of iron homeostasis: Lessons from mouse models. Mol Aspects Med 2020; 75:100872. [DOI: 10.1016/j.mam.2020.100872] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2020] [Revised: 06/28/2020] [Accepted: 07/02/2020] [Indexed: 12/13/2022]
|
30
|
Cheli VT, Correale J, Paez PM, Pasquini JM. Iron Metabolism in Oligodendrocytes and Astrocytes, Implications for Myelination and Remyelination. ASN Neuro 2020; 12:1759091420962681. [PMID: 32993319 PMCID: PMC7545512 DOI: 10.1177/1759091420962681] [Citation(s) in RCA: 97] [Impact Index Per Article: 19.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Iron is a key nutrient for normal central nervous system (CNS) development and function; thus, iron deficiency as well as iron excess may result in harmful effects in the CNS. Oligodendrocytes and astrocytes are crucial players in brain iron equilibrium. However, the mechanisms of iron uptake, storage, and efflux in oligodendrocytes and astrocytes during CNS development or under pathological situations such as demyelination are not completely understood. In the CNS, iron is directly required for myelin production as a cofactor for enzymes involved in ATP, cholesterol and lipid synthesis, and oligodendrocytes are the cells with the highest iron levels in the brain which is linked to their elevated metabolic needs associated with the process of myelination. Unlike oligodendrocytes, astrocytes do not have a high metabolic requirement for iron. However, these cells are in close contact with blood vessel and have a strong iron transport capacity. In several pathological situations, changes in iron homoeostasis result in altered cellular iron distribution and accumulation and oxidative stress. In inflammatory demyelinating diseases such as multiple sclerosis, reactive astrocytes accumulate iron and upregulate iron efflux and influx molecules, which suggest that they are outfitted to take up and safely recycle iron. In this review, we will discuss the participation of oligodendrocytes and astrocytes in CNS iron homeostasis. Understanding the molecular mechanisms of iron uptake, storage, and efflux in oligodendrocytes and astrocytes is necessary for planning effective strategies for iron management during CNS development as well as for the treatment of demyelinating diseases.
Collapse
Affiliation(s)
- Veronica T Cheli
- Department of Pharmacology and Toxicology, Jacobs School of Medicine and Biomedical Sciences, Hunter James Kelly Research Institute, The State University of New York, University at Buffalo, Buffalo, New York, United States
| | | | - Pablo M Paez
- Department of Pharmacology and Toxicology, Jacobs School of Medicine and Biomedical Sciences, Hunter James Kelly Research Institute, The State University of New York, University at Buffalo, Buffalo, New York, United States
| | - Juana M Pasquini
- CONICET, Instituto de Química y Fisicoquímica Biológicas, Universidad de Buenos Aires, Buenos Aires, Argentina
| |
Collapse
|
31
|
Wang S, He X, Wu Q, Jiang L, Chen L, Yu Y, Zhang P, Huang X, Wang J, Ju Z, Min J, Wang F. Transferrin receptor 1-mediated iron uptake plays an essential role in hematopoiesis. Haematologica 2020; 105:2071-2082. [PMID: 31601687 PMCID: PMC7395265 DOI: 10.3324/haematol.2019.224899] [Citation(s) in RCA: 61] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2019] [Accepted: 10/04/2019] [Indexed: 12/13/2022] Open
Abstract
Transferrin receptor 1 (Tfr1) mediates the endocytosis of diferric transferrin in order to transport iron, and Tfr1 has been suggested to play an important role in hematopoiesis. To study the role of Tfr1 in hematopoiesis, we generated hematopoietic stem cell (HSC) specific Tfr1 knockout mice. We found that Tfr1 conditional knockout mice reached full term but died within one week of birth. Further analyses revealed that Tfr1-deficient HSC had impaired development of all hematopoietic progenitors except thrombocytes and B lymphocytes. In addition, Tfr1-deficient cells had cellular iron deficiency, which blocked the proliferation and differentiation of hematopoietic precursor cells, attenuated the commitment of hematopoietic lineages, and reduced the regeneration potential of HSC. Notably, hemin rescued the colony-forming capacity of Tfr1-deficient HSC, whereas expressing a mutant Tfr1 that lacks the protein's iron-transporting capacity failed to rescue hematopoiesis. These findings provide direct evidence that Tfr1 is essential for hematopoiesis through binding diferric transferrin to supply iron to cells.
Collapse
Affiliation(s)
- Shufen Wang
- The First Affiliated Hospital, School of Public Health, Institute of Translational Medicine, Zhenjiang Provincial Key Laboratory of Pancreatic Disease, Zhejiang University School of Medicine, Hangzhou
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, China Agricultural University, Beijing
| | - Xuyan He
- The First Affiliated Hospital, School of Public Health, Institute of Translational Medicine, Zhenjiang Provincial Key Laboratory of Pancreatic Disease, Zhejiang University School of Medicine, Hangzhou
| | - Qian Wu
- The First Affiliated Hospital, School of Public Health, Institute of Translational Medicine, Zhenjiang Provincial Key Laboratory of Pancreatic Disease, Zhejiang University School of Medicine, Hangzhou
| | - Li Jiang
- The First Affiliated Hospital, School of Public Health, Institute of Translational Medicine, Zhenjiang Provincial Key Laboratory of Pancreatic Disease, Zhejiang University School of Medicine, Hangzhou
| | - Liyun Chen
- The First Affiliated Hospital, School of Public Health, Institute of Translational Medicine, Zhenjiang Provincial Key Laboratory of Pancreatic Disease, Zhejiang University School of Medicine, Hangzhou
| | - Yingying Yu
- The First Affiliated Hospital, School of Public Health, Institute of Translational Medicine, Zhenjiang Provincial Key Laboratory of Pancreatic Disease, Zhejiang University School of Medicine, Hangzhou
| | - Pan Zhang
- The First Affiliated Hospital, School of Public Health, Institute of Translational Medicine, Zhenjiang Provincial Key Laboratory of Pancreatic Disease, Zhejiang University School of Medicine, Hangzhou
| | - Xin Huang
- Department of Nutrition, Precision Nutrition Innovation Center, School of Public Health, Zhengzhou University, Zhengzhou
| | - Jia Wang
- Department of Nutrition, Precision Nutrition Innovation Center, School of Public Health, Zhengzhou University, Zhengzhou
| | - Zhenyu Ju
- Key Laboratory of Regenerative Medicine of Ministry of Education, Institute of Aging and Regenerative Medicine, Jinan University, Guangzhou, China
| | - Junxia Min
- The First Affiliated Hospital, School of Public Health, Institute of Translational Medicine, Zhenjiang Provincial Key Laboratory of Pancreatic Disease, Zhejiang University School of Medicine, Hangzhou
| | - Fudi Wang
- The First Affiliated Hospital, School of Public Health, Institute of Translational Medicine, Zhenjiang Provincial Key Laboratory of Pancreatic Disease, Zhejiang University School of Medicine, Hangzhou
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, China Agricultural University, Beijing
- Department of Nutrition, Precision Nutrition Innovation Center, School of Public Health, Zhengzhou University, Zhengzhou
| |
Collapse
|
32
|
Zhang S, Cao Y, Yang Q. Transferrin receptor 1 levels at the cell surface influence the susceptibility of newborn piglets to PEDV infection. PLoS Pathog 2020; 16:e1008682. [PMID: 32730327 PMCID: PMC7419007 DOI: 10.1371/journal.ppat.1008682] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Revised: 08/11/2020] [Accepted: 06/04/2020] [Indexed: 12/16/2022] Open
Abstract
Porcine epidemic diarrhea virus (PEDV) mainly infects the intestinal epithelial cells of newborn piglets causing acute, severe atrophic enteritis. The underlying mechanisms of PEDV infection and the reasons why newborn piglets are more susceptible than older pigs remain incompletely understood. Iron deficiency is common in newborn piglets. Here we found that high levels of transferrin receptor 1 (TfR1) distributed in the apical tissue of the intestinal villi of newborns, and intracellular iron levels influence the susceptibility of newborn piglets to PEDV. We show that iron deficiency induced by deferoxamine (DFO, an iron chelating agent) promotes PEDV infection while iron accumulation induced by ferric ammonium citrate (FAC, an iron supplement) impairs PEDV infection in vitro and in vivo. Besides, PEDV infection was inhibited by occluding TfR1 with antibodies or decreasing TfR1 expression. Additionally, PEDV infection was increased in PEDV-resistant Caco-2 and HEK 293T cells over-expressed porcine TfR1. Mechanistically, the PEDV S1 protein interacts with the extracellular region of TfR1 during PEDV entry, promotes TfR1 re-localization and clustering, then activates TfR1 tyrosine phosphorylation mediated by Src kinase, and heightens the internalization of TfR1, thereby promoting PEDV entry. Taken together, these data suggest that the higher expression of TfR1 in the apical tissue of the intestinal villi caused by iron deficiency, accounts for newborn piglets being acutely susceptible to PEDV. Newborn piglets are particularly susceptible to infection by PEDV, with 80–100% dying within days of infection. The reasons for newborns’ acute susceptibility to PEDV infection have not been elucidated clearly. The primarily target of PEDV is the porcine intestinal epithelial cells. Here, we show that the high expression of TfR1 in the apical tissue of intestinal villi in newborn piglets with iron deficiency is a reason for their susceptibility to PEDV. Further, we demonstrate that iron supplementation reduces PEDV infection. This study reveals that iron plays an important role in the susceptibility of newborn piglets to PEDV and provides insights into therapies for the prevention and treatment of PEDV infections.
Collapse
Affiliation(s)
- Shuai Zhang
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Weigang, Nanjing, Jiangsu, PR China
| | - Yanan Cao
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Weigang, Nanjing, Jiangsu, PR China
| | - Qian Yang
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Weigang, Nanjing, Jiangsu, PR China
| |
Collapse
|
33
|
Li J, Pan X, Pan G, Song Z, He Y, Zhang S, Ye X, Yang X, Xie E, Wang X, Mai X, Yin X, Tang B, Shu X, Chen P, Dai X, Tian Y, Yao L, Han M, Xu G, Zhang H, Sun J, Chen H, Wang F, Min J, Xie L. Transferrin Receptor 1 Regulates Thermogenic Capacity and Cell Fate in Brown/Beige Adipocytes. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2020; 7:1903366. [PMID: 32596110 PMCID: PMC7312276 DOI: 10.1002/advs.201903366] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/28/2019] [Revised: 02/24/2020] [Indexed: 05/02/2023]
Abstract
Iron homeostasis is essential for maintaining cellular function in a wide range of cell types. However, whether iron affects the thermogenic properties of adipocytes is currently unknown. Using integrative analyses of multi-omics data, transferrin receptor 1 (Tfr1) is identified as a candidate for regulating thermogenesis in beige adipocytes. Furthermore, it is shown that mice lacking Tfr1 specifically in adipocytes have impaired thermogenesis, increased insulin resistance, and low-grade inflammation accompanied by iron deficiency and mitochondrial dysfunction. Mechanistically, the cold treatment in beige adipocytes selectively stabilizes hypoxia-inducible factor 1-alpha (HIF1α), upregulating the Tfr1 gene, and thermogenic adipocyte-specific Hif1α deletion reduces thermogenic gene expression in beige fat without altering core body temperature. Notably, Tfr1 deficiency in interscapular brown adipose tissue (iBAT) leads to the transdifferentiation of brown preadipocytes into white adipocytes and muscle cells; in contrast, long-term exposure to a low-iron diet fails to phenocopy the transdifferentiation effect found in Tfr1-deficient mice. Moreover, mice lacking transmembrane serine protease 6 (Tmprss6) develop iron deficiency in both inguinal white adipose tissue (iWAT) and iBAT, and have impaired cold-induced beige adipocyte formation and brown fat thermogenesis. Taken together, these findings indicate that Tfr1 plays an essential role in thermogenic adipocytes via both iron-dependent and iron-independent mechanisms.
Collapse
Affiliation(s)
- Jin Li
- The First Affiliated HospitalInstitute of Translational MedicineSchool of Public HealthZhejiang University School of MedicineHangzhou310058China
- Beijing Advanced Innovation Center for Food Nutrition and Human HealthChina Agricultural UniversityBeijing100193China
- Department of NutritionPrecision Nutrition Innovation CenterSchool of Public HealthZhengzhou UniversityZhengzhou450001China
| | - Xiaohan Pan
- State Key Laboratory of Applied Microbiology Southern ChinaGuangdong Provincial Key Laboratory of Microbial Culture Collection and ApplicationGuangdong Open Laboratory of Applied MicrobiologyGuangdong Institute of MicrobiologyGuangdong Academy of SciencesZhujiang HospitalSouthern Medical UniversityGuangzhou510070China
| | - Guihua Pan
- State Key Laboratory of Applied Microbiology Southern ChinaGuangdong Provincial Key Laboratory of Microbial Culture Collection and ApplicationGuangdong Open Laboratory of Applied MicrobiologyGuangdong Institute of MicrobiologyGuangdong Academy of SciencesZhujiang HospitalSouthern Medical UniversityGuangzhou510070China
| | - Zijun Song
- The First Affiliated HospitalInstitute of Translational MedicineSchool of Public HealthZhejiang University School of MedicineHangzhou310058China
| | - Yao He
- The First Affiliated HospitalInstitute of Translational MedicineSchool of Public HealthZhejiang University School of MedicineHangzhou310058China
| | - Susu Zhang
- State Key Laboratory of Applied Microbiology Southern ChinaGuangdong Provincial Key Laboratory of Microbial Culture Collection and ApplicationGuangdong Open Laboratory of Applied MicrobiologyGuangdong Institute of MicrobiologyGuangdong Academy of SciencesZhujiang HospitalSouthern Medical UniversityGuangzhou510070China
| | - Xueru Ye
- Nanfang HospitalSouthern Medical UniversityGuangzhou510515China
| | - Xiang Yang
- The First Affiliated HospitalInstitute of Translational MedicineSchool of Public HealthZhejiang University School of MedicineHangzhou310058China
| | - Enjun Xie
- The First Affiliated HospitalInstitute of Translational MedicineSchool of Public HealthZhejiang University School of MedicineHangzhou310058China
| | - Xinhui Wang
- The First Affiliated HospitalInstitute of Translational MedicineSchool of Public HealthZhejiang University School of MedicineHangzhou310058China
| | - Xudong Mai
- State Key Laboratory of Applied Microbiology Southern ChinaGuangdong Provincial Key Laboratory of Microbial Culture Collection and ApplicationGuangdong Open Laboratory of Applied MicrobiologyGuangdong Institute of MicrobiologyGuangdong Academy of SciencesZhujiang HospitalSouthern Medical UniversityGuangzhou510070China
| | - Xiangju Yin
- The First Affiliated HospitalInstitute of Translational MedicineSchool of Public HealthZhejiang University School of MedicineHangzhou310058China
| | - Biyao Tang
- The First Affiliated HospitalInstitute of Translational MedicineSchool of Public HealthZhejiang University School of MedicineHangzhou310058China
| | - Xuan Shu
- The First Affiliated HospitalInstitute of Translational MedicineSchool of Public HealthZhejiang University School of MedicineHangzhou310058China
| | - Pengyu Chen
- The First Affiliated HospitalInstitute of Translational MedicineSchool of Public HealthZhejiang University School of MedicineHangzhou310058China
| | - Xiaoshuang Dai
- BGI Institute of Applied AgricultureBGI‐ShenzhenShenzhen518120China
| | - Ye Tian
- State Key Laboratory of Applied Microbiology Southern ChinaGuangdong Provincial Key Laboratory of Microbial Culture Collection and ApplicationGuangdong Open Laboratory of Applied MicrobiologyGuangdong Institute of MicrobiologyGuangdong Academy of SciencesZhujiang HospitalSouthern Medical UniversityGuangzhou510070China
| | - Liheng Yao
- State Key Laboratory of Applied Microbiology Southern ChinaGuangdong Provincial Key Laboratory of Microbial Culture Collection and ApplicationGuangdong Open Laboratory of Applied MicrobiologyGuangdong Institute of MicrobiologyGuangdong Academy of SciencesZhujiang HospitalSouthern Medical UniversityGuangzhou510070China
| | - Mulan Han
- State Key Laboratory of Applied Microbiology Southern ChinaGuangdong Provincial Key Laboratory of Microbial Culture Collection and ApplicationGuangdong Open Laboratory of Applied MicrobiologyGuangdong Institute of MicrobiologyGuangdong Academy of SciencesZhujiang HospitalSouthern Medical UniversityGuangzhou510070China
| | - Guohuan Xu
- State Key Laboratory of Applied Microbiology Southern ChinaGuangdong Provincial Key Laboratory of Microbial Culture Collection and ApplicationGuangdong Open Laboratory of Applied MicrobiologyGuangdong Institute of MicrobiologyGuangdong Academy of SciencesZhujiang HospitalSouthern Medical UniversityGuangzhou510070China
| | - Huijie Zhang
- Nanfang HospitalSouthern Medical UniversityGuangzhou510515China
| | - Jia Sun
- State Key Laboratory of Applied Microbiology Southern ChinaGuangdong Provincial Key Laboratory of Microbial Culture Collection and ApplicationGuangdong Open Laboratory of Applied MicrobiologyGuangdong Institute of MicrobiologyGuangdong Academy of SciencesZhujiang HospitalSouthern Medical UniversityGuangzhou510070China
| | - Hong Chen
- State Key Laboratory of Applied Microbiology Southern ChinaGuangdong Provincial Key Laboratory of Microbial Culture Collection and ApplicationGuangdong Open Laboratory of Applied MicrobiologyGuangdong Institute of MicrobiologyGuangdong Academy of SciencesZhujiang HospitalSouthern Medical UniversityGuangzhou510070China
| | - Fudi Wang
- The First Affiliated HospitalInstitute of Translational MedicineSchool of Public HealthZhejiang University School of MedicineHangzhou310058China
- Beijing Advanced Innovation Center for Food Nutrition and Human HealthChina Agricultural UniversityBeijing100193China
- Department of NutritionPrecision Nutrition Innovation CenterSchool of Public HealthZhengzhou UniversityZhengzhou450001China
| | - Junxia Min
- The First Affiliated HospitalInstitute of Translational MedicineSchool of Public HealthZhejiang University School of MedicineHangzhou310058China
| | - Liwei Xie
- State Key Laboratory of Applied Microbiology Southern ChinaGuangdong Provincial Key Laboratory of Microbial Culture Collection and ApplicationGuangdong Open Laboratory of Applied MicrobiologyGuangdong Institute of MicrobiologyGuangdong Academy of SciencesZhujiang HospitalSouthern Medical UniversityGuangzhou510070China
| |
Collapse
|
34
|
Camaschella C, Nai A, Silvestri L. Iron metabolism and iron disorders revisited in the hepcidin era. Haematologica 2020; 105:260-272. [PMID: 31949017 PMCID: PMC7012465 DOI: 10.3324/haematol.2019.232124] [Citation(s) in RCA: 402] [Impact Index Per Article: 80.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2019] [Accepted: 12/18/2019] [Indexed: 02/06/2023] Open
Abstract
Iron is biologically essential, but also potentially toxic; as such it is tightly controlled at cell and systemic levels to prevent both deficiency and overload. Iron regulatory proteins post-transcriptionally control genes encoding proteins that modulate iron uptake, recycling and storage and are themselves regulated by iron. The master regulator of systemic iron homeostasis is the liver peptide hepcidin, which controls serum iron through degradation of ferroportin in iron-absorptive enterocytes and iron-recycling macrophages. This review emphasizes the most recent findings in iron biology, deregulation of the hepcidin-ferroportin axis in iron disorders and how research results have an impact on clinical disorders. Insufficient hepcidin production is central to iron overload while hepcidin excess leads to iron restriction. Mutations of hemochro-matosis genes result in iron excess by downregulating the liver BMP-SMAD signaling pathway or by causing hepcidin-resistance. In iron-loading anemias, such as β-thalassemia, enhanced albeit ineffective ery-thropoiesis releases erythroferrone, which sequesters BMP receptor ligands, thereby inhibiting hepcidin. In iron-refractory, iron-deficiency ane-mia mutations of the hepcidin inhibitor TMPRSS6 upregulate the BMP-SMAD pathway. Interleukin-6 in acute and chronic inflammation increases hepcidin levels, causing iron-restricted erythropoiesis and ane-mia of inflammation in the presence of iron-replete macrophages. Our improved understanding of iron homeostasis and its regulation is having an impact on the established schedules of oral iron treatment and the choice of oral versus intravenous iron in the management of iron deficiency. Moreover it is leading to the development of targeted therapies for iron overload and inflammation, mainly centered on the manipulation of the hepcidin-ferroportin axis.
Collapse
Affiliation(s)
- Clara Camaschella
- Regulation of Iron Metabolism Unit, Division of Genetics and Cell Biology, San Raffaele Scientific Institute, Milan
| | - Antonella Nai
- Regulation of Iron Metabolism Unit, Division of Genetics and Cell Biology, San Raffaele Scientific Institute, Milan.,Vita Salute San Raffaele University, Milan, Italy
| | - Laura Silvestri
- Regulation of Iron Metabolism Unit, Division of Genetics and Cell Biology, San Raffaele Scientific Institute, Milan.,Vita Salute San Raffaele University, Milan, Italy
| |
Collapse
|
35
|
Abstract
Intestinal iron homeostasis is like the Zhong-Yong in traditional Chinese culture, which is a dynamic balance between Yin and Yang.
Collapse
Affiliation(s)
- Haoxuan Ding
- College of Animal Science
- Zhejiang University
- Key Laboratory of animal feed and nutrition of Zhejiang Province
- Hangzhou
- China
| | - Xiaonan Yu
- College of Animal Science
- Zhejiang University
- Key Laboratory of animal feed and nutrition of Zhejiang Province
- Hangzhou
- China
| | - Jie Feng
- College of Animal Science
- Zhejiang University
- Key Laboratory of animal feed and nutrition of Zhejiang Province
- Hangzhou
- China
| |
Collapse
|
36
|
Iron Metabolism in the Peripheral Nervous System: The Role of DMT1, Ferritin, and Transferrin Receptor in Schwann Cell Maturation and Myelination. J Neurosci 2019; 39:9940-9953. [PMID: 31676601 DOI: 10.1523/jneurosci.1409-19.2019] [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: 06/17/2019] [Revised: 10/09/2019] [Accepted: 10/29/2019] [Indexed: 12/24/2022] Open
Abstract
Iron is an essential cofactor for many cellular enzymes involved in myelin synthesis, and iron homeostasis unbalance is a central component of peripheral neuropathies. However, iron absorption and management in the PNS are poorly understood. To study iron metabolism in Schwann cells (SCs), we have created 3 inducible conditional KO mice in which three essential proteins implicated in iron uptake and storage, the divalent metal transporter 1 (DMT1), the ferritin heavy chain (Fth), and the transferrin receptor 1 (Tfr1), were postnatally ablated specifically in SCs. Deleting DMT1, Fth, or Tfr1 in vitro significantly reduce SC proliferation, maturation, and the myelination of DRG axons. This was accompanied by an important reduction in iron incorporation and storage. When these proteins were KO in vivo during the first postnatal week, the sciatic nerve of all 3 conditional KO animals displayed a significant reduction in the synthesis of myelin proteins and in the percentage of myelinated axons. Knocking out Fth produced the most severe phenotype, followed by DMT1 and, last, Tfr1. Importantly, DMT1 as well as Fth KO mice showed substantial motor coordination deficits. In contrast, deleting these proteins in mature myelinating SCs results in milder phenotypes characterized by small reductions in the percentage of myelinated axons and minor changes in the g-ratio of myelinated axons. These results indicate that DMT1, Fth, and Tfr1 are critical proteins for early postnatal iron uptake and storage in SCs and, as a consequence, for the normal myelination of the PNS.SIGNIFICANCE STATEMENT To determine the function of the divalent metal transporter 1, the transferrin receptor 1, and the ferritin heavy chain in Schwann cell (SC) maturation and myelination, we created 3 conditional KO mice in which these proteins were postnatally deleted in Sox10-positive SCs. We have established that these proteins are necessary for normal SC iron incorporation and storage, and, as a consequence, for an effective myelination of the PNS. Since iron is indispensable for SC maturation, understanding iron metabolism in SCs is an essential prerequisite for developing therapies for demyelinating diseases in the PNS.
Collapse
|
37
|
Abstract
Iron is an essential element that is indispensable for life. The delicate physiological body iron balance is maintained by both systemic and cellular regulatory mechanisms. The iron-regulatory hormone hepcidin assures maintenance of adequate systemic iron levels and is regulated by circulating and stored iron levels, inflammation and erythropoiesis. The kidney has an important role in preventing iron loss from the body by means of reabsorption. Cellular iron levels are dependent on iron import, storage, utilization and export, which are mainly regulated by the iron response element-iron regulatory protein (IRE-IRP) system. In the kidney, iron transport mechanisms independent of the IRE-IRP system have been identified, suggesting additional mechanisms for iron handling in this organ. Yet, knowledge gaps on renal iron handling remain in terms of redundancy in transport mechanisms, the roles of the different tubular segments and related regulatory processes. Disturbances in cellular and systemic iron balance are recognized as causes and consequences of kidney injury. Consequently, iron metabolism has become a focus for novel therapeutic interventions for acute kidney injury and chronic kidney disease, which has fuelled interest in the molecular mechanisms of renal iron handling and renal injury, as well as the complex dynamics between systemic and local cellular iron regulation.
Collapse
|
38
|
Schwartz AJ, Converso-Baran K, Michele DE, Shah YM. A genetic mouse model of severe iron deficiency anemia reveals tissue-specific transcriptional stress responses and cardiac remodeling. J Biol Chem 2019; 294:14991-15002. [PMID: 31416832 DOI: 10.1074/jbc.ra119.009578] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2019] [Revised: 08/13/2019] [Indexed: 01/24/2023] Open
Abstract
Iron is a micronutrient fundamental for life. Iron homeostasis in mammals requires sustained postnatal intestinal iron absorption that maintains intracellular iron concentrations for central and systemic metabolism as well as for erythropoiesis and oxygen transport. More than 1 billion people worldwide suffer from iron deficiency anemia (IDA), a state of systemic iron insufficiency that limits the production of red blood cells and leads to tissue hypoxia and intracellular iron stress. Despite this tremendous public health concern, very few genetic models of IDA are available to study its progression. Here we developed and characterized a novel genetic mouse model of IDA. We found that tamoxifen-inducible deletion of the mammalian iron exporter ferroportin exclusively in intestinal epithelial cells leads to loss of intestinal iron absorption. Ferroportin ablation yielded a robust phenotype of progressive IDA that develops in as little as 3 months following disruption of intestinal iron absorption. We noted that, at end-stage IDA, tissue-specific transcriptional stress responses occur in which the heart shows little to no hypoxic and iron stress compared with other peripheral organs. However, morphometric and echocardiographic analysis revealed massive cardiac hypertrophy and chamber dilation, albeit with increased cardiac output at very low basal heart rates. We propose that our intestine-specific ferroportin knockout mouse model of end-stage IDA could be used in future studies to investigate IDA progression and cell-specific responses to hypoxic and iron stress.
Collapse
Affiliation(s)
- Andrew J Schwartz
- Department of Molecular & Integrative Physiology, and Internal Medicine, University of Michigan, Ann Arbor, Michigan 48109
| | - Kimber Converso-Baran
- Department of Molecular & Integrative Physiology, and Internal Medicine, University of Michigan, Ann Arbor, Michigan 48109
| | - Daniel E Michele
- Department of Molecular & Integrative Physiology, and Internal Medicine, University of Michigan, Ann Arbor, Michigan 48109.,Division of Cardiovascular Medicine, University of Michigan, Ann Arbor, Michigan 48109
| | - Yatrik M Shah
- Department of Molecular & Integrative Physiology, and Internal Medicine, University of Michigan, Ann Arbor, Michigan 48109 .,Division of Gastroenterology, University of Michigan, Ann Arbor, Michigan 48109
| |
Collapse
|
39
|
Germot A, Maftah A. POFUT1 and PLAGL2 gene pair linked by a bidirectional promoter: the two in one of tumour progression in colorectal cancer? EBioMedicine 2019; 46:25-26. [PMID: 31378697 PMCID: PMC6711883 DOI: 10.1016/j.ebiom.2019.07.065] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2019] [Accepted: 07/25/2019] [Indexed: 12/21/2022] Open
Affiliation(s)
- Agnès Germot
- Limoges University, PEIRENE, EA 7500, Glycosylation and Cell Differentiation, F-87060 Limoges, France
| | - Abderrahman Maftah
- Limoges University, PEIRENE, EA 7500, Glycosylation and Cell Differentiation, F-87060 Limoges, France.
| |
Collapse
|
40
|
Paterek A, Mackiewicz U, Mączewski M. Iron and the heart: A paradigm shift from systemic to cardiomyocyte abnormalities. J Cell Physiol 2019; 234:21613-21629. [DOI: 10.1002/jcp.28820] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2018] [Revised: 04/16/2019] [Accepted: 04/17/2019] [Indexed: 12/13/2022]
Affiliation(s)
- Aleksandra Paterek
- Department of Clinical Physiology Centre of Postgraduate Medical Education Warsaw Poland
| | - Urszula Mackiewicz
- Department of Clinical Physiology Centre of Postgraduate Medical Education Warsaw Poland
| | - Michał Mączewski
- Department of Clinical Physiology Centre of Postgraduate Medical Education Warsaw Poland
| |
Collapse
|
41
|
Recalcati S, Correnti M, Gammella E, Raggi C, Invernizzi P, Cairo G. Iron Metabolism in Liver Cancer Stem Cells. Front Oncol 2019; 9:149. [PMID: 30941302 PMCID: PMC6433741 DOI: 10.3389/fonc.2019.00149] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2018] [Accepted: 02/22/2019] [Indexed: 12/15/2022] Open
Abstract
Cancer stem cells (CSC) which have been identified in several tumors, including liver cancer, represent a particular subpopulation of tumor cells characterized by properties similar to those of adult stem cells. Importantly, CSC are resistant to standard therapies, thereby leading to metastatic dissemination and tumor relapse. Given the increasing evidence that iron homeostasis is deregulated in cancer, here we describe the iron homeostasis alterations in cancer cells, particularly in liver CSC. We also discuss two paradoxically opposite iron manipulation-strategies for tumor therapy based either on iron chelation or iron overload-mediated oxidant production leading to ferroptosis. A better understanding of iron metabolism modifications occurring in hepatic tumors and particularly in liver CSC cells may offer new therapeutic options for this cancer, which is characterized by increasing incidence and unfavorable prognosis.
Collapse
Affiliation(s)
- Stefania Recalcati
- Department of Biomedical Sciences for Health, University of Milan, Milan, Italy
| | | | - Elena Gammella
- Department of Biomedical Sciences for Health, University of Milan, Milan, Italy
| | - Chiara Raggi
- Humanitas Clinical and Research Center, IRCCS, Rozzano, Italy.,Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Pietro Invernizzi
- Division of Gastroenterology, Department of Medicine and Surgery, Center for Autoimmune Liver Diseases, San Gerardo Hospital, University of Milano-Bicocca, Monza, Italy
| | - Gaetano Cairo
- Department of Biomedical Sciences for Health, University of Milan, Milan, Italy
| |
Collapse
|
42
|
Kawabata H. Transferrin and transferrin receptors update. Free Radic Biol Med 2019; 133:46-54. [PMID: 29969719 DOI: 10.1016/j.freeradbiomed.2018.06.037] [Citation(s) in RCA: 409] [Impact Index Per Article: 68.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/13/2018] [Revised: 06/29/2018] [Accepted: 06/29/2018] [Indexed: 12/20/2022]
Abstract
In vertebrates, transferrin (Tf) safely delivers iron through circulation to cells. Tf-bound iron is incorporated through Tf receptor (TfR) 1-mediated endocytosis. TfR1 can mediate cellular uptake of both Tf and H-ferritin, an iron storage protein. New World arenaviruses, which cause hemorrhagic fever, and Plasmodium vivax use TfR1 for entry into host cells. Human TfR2, another receptor for Tf, is predominantly expressed in hepatocytes and erythroid precursors, and holo-Tf dramatically upregulates its expression. TfR2 forms a complex with hemochromatosis protein, HFE, and serves as a component of the iron sensing machinery in hepatocytes. Defects in TfR2 cause systemic iron overload, hemochromatosis, through down-regulation of hepcidin. In erythroid cells, TfR2 forms a complex with the erythropoietin receptor and regulates erythropoiesis. TfR2 facilitates iron transport from lysosomes to mitochondria in erythroblasts and dopaminergic neurons. Administration of apo-Tf, which scavenges free iron, has been explored for various clinical conditions including atransferrinemia, iron overload, and tissue ischemia. Apo-Tf has also been shown to ameliorate anemia in animal models of β-thalassemia. In this review, I provide an update and summary on our knowledge of mammalian Tf and its receptors.
Collapse
Affiliation(s)
- Hiroshi Kawabata
- Department of Hematology and Immunology, Kanazawa Medical University, 1-1 Daigaku, Uchinada-machi, Ishikawa-ken 920-0293, Japan.
| |
Collapse
|
43
|
Sangkhae V, Nemeth E. Placental iron transport: The mechanism and regulatory circuits. Free Radic Biol Med 2019; 133:254-261. [PMID: 29981833 PMCID: PMC7059975 DOI: 10.1016/j.freeradbiomed.2018.07.001] [Citation(s) in RCA: 71] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/15/2018] [Revised: 06/30/2018] [Accepted: 07/03/2018] [Indexed: 02/06/2023]
Abstract
As the interface between the fetal and maternal circulation, the placenta facilitates both nutrient and waste exchange for the developing fetus. Iron is essential for healthy pregnancy, and transport of iron across the placenta is required for fetal growth and development. Perturbation of this transfer can lead to adverse pregnancy outcomes. Despite its importance, our understanding of how a large amount of iron is transported across placental membranes, how this process is regulated, and which iron transporter proteins function in different placental cells remains rudimentary. Mechanistic studies in mouse models, including placenta-specific deletion or overexpression of iron-related proteins will be essential to make progress. This review summarizes our current understanding about iron transport across the syncytiotrophoblast under physiological conditions and identifies areas for further investigation.
Collapse
Affiliation(s)
- Veena Sangkhae
- Center for Iron Disorders, Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, 10833 LeConte Ave, CHS 37-131, Los Angeles, CA 90095, USA.
| | - Elizabeta Nemeth
- Center for Iron Disorders, Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, 10833 LeConte Ave, CHS 37-131, Los Angeles, CA 90095, USA.
| |
Collapse
|
44
|
Amos-Kroohs RM, Usach V, Piñero G, Vorhees CV, Vivot RM, Soto PA, Williams MT, Setton-Avruj P. Metal bashing: iron deficiency and manganese overexposure impact on peripheral nerves. JOURNAL OF TOXICOLOGY AND ENVIRONMENTAL HEALTH. PART A 2019; 82:99-112. [PMID: 30652531 PMCID: PMC6397089 DOI: 10.1080/15287394.2019.1566105] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
Iron (Fe) deficiency (FeD) and manganese (Mn) overexposure (MnOE) may result in several neurological alterations in the nervous system. Iron deficiency produces unique neurological deficits due to its elemental role in central nervous system (CNS) development and myelination, which might persist after normalization of Fe in the diet. Conversely, MnOE is associated with diverse neurocognitive deficits. Despite these well-known neurotoxic effects on the CNS, the influence of FeD and MnOE on the peripheral nervous system (PNS) remains poorly understood. The aim of the present investigation was to examine the effects of developmental FeD and MnOE or their combination on the sciatic nerve of young and adult rats. The parameters measured included divalent metal transporter 1 (DMT1), transferrin receptor (TfR), myelin basic protein (MBP) and peripheral myelin protein 22 (PMP22) expression, as well as Fe levels in the nerve. Our results showed that FeD produced a significant reduction in MBP and PMP22 content at P29, which persisted at P60 after Fe-sufficient diet replenishment regardless of Mn exposure levels. At P60 MnOE significantly increased sciatic nerve Fe content and DMT1 expression. However, the combination of FeD and MnOE produced no marked motor skill impairment. Evidence indicates that FeD appears to hinder developmental peripheral myelination, while MnOE may directly alter Fe homeostasis. Further studies are required to elucidate the interplay between these pathological conditions.
Collapse
Affiliation(s)
- Robyn M. Amos-Kroohs
- University of North Carolina at Chapel Hill, Nutrition Research Institute, Kannapolis, NC 28081
| | - Vanina Usach
- Departamento de Química Biológica, Facultad de Farmacia y Bíoquímica, Universidad de Buenos Aires. Instituto de Química y Físicoquímica Biológica (IQUIFIB), UBA-CONICET, Buenos Aires. Argentina
| | - Gonzalo Piñero
- Departamento de Química Biológica, Facultad de Farmacia y Bíoquímica, Universidad de Buenos Aires. Instituto de Química y Físicoquímica Biológica (IQUIFIB), UBA-CONICET, Buenos Aires. Argentina
| | - Charles V. Vorhees
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati OH 45229
- Cincinnati Children’s Research Foundation, Div. of Neurology, Cincinnati OH 45229
| | - Rocío Martinez Vivot
- Departamento de Química Biológica, Facultad de Farmacia y Bíoquímica, Universidad de Buenos Aires. Instituto de Química y Físicoquímica Biológica (IQUIFIB), UBA-CONICET, Buenos Aires. Argentina
| | - Paula A. Soto
- Departamento de Química Biológica, Facultad de Farmacia y Bíoquímica, Universidad de Buenos Aires. Instituto de Química y Físicoquímica Biológica (IQUIFIB), UBA-CONICET, Buenos Aires. Argentina
| | - Michael T. Williams
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati OH 45229
- Cincinnati Children’s Research Foundation, Div. of Neurology, Cincinnati OH 45229
| | - Patricia Setton-Avruj
- Departamento de Química Biológica, Facultad de Farmacia y Bíoquímica, Universidad de Buenos Aires. Instituto de Química y Físicoquímica Biológica (IQUIFIB), UBA-CONICET, Buenos Aires. Argentina
| |
Collapse
|
45
|
Transferrin receptor 1 controls systemic iron homeostasis by fine-tuning hepcidin expression to hepatocellular iron load. Blood 2018; 133:344-355. [PMID: 30538134 DOI: 10.1182/blood-2018-05-850404] [Citation(s) in RCA: 65] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2018] [Accepted: 12/04/2018] [Indexed: 02/06/2023] Open
Abstract
Transferrin receptor 1 (Tfr1) mediates uptake of circulating transferrin-bound iron to developing erythroid cells and other cell types. Its critical physiological function is highlighted by the embryonic lethal phenotype of Tfr1-knockout (Tfrc-/-) mice and the pathologies of several tissue-specific knockouts. We generated TfrcAlb-Cre mice bearing hepatocyte-specific ablation of Tfr1 to explore implications in hepatocellular and systemic iron homeostasis. TfrcAlb-Cre mice are viable and do not display any apparent liver pathology. Nevertheless, their liver iron content (LIC) is lower compared with that of control Tfrcfl/fl littermates as a result of the reduced capacity of Tfr1-deficient hepatocytes to internalize iron from transferrin. Even though liver Hamp messenger RNA (mRNA) and serum hepcidin levels do not differ between TfrcAlb-Cre and Tfrcfl/fl mice, Hamp/LIC and hepcidin/LIC ratios are significantly higher in the former. Importantly, this is accompanied by modest hypoferremia and microcytosis, and it predisposes TfrcAlb-Cre mice to iron-deficiency anemia. TfrcAlb-Cre mice appropriately regulate Hamp expression following dietary iron manipulations or holo-transferrin injection. Holo-transferrin also triggers proper induction of Hamp mRNA, ferritin, and Tfr2 in primary TfrcAlb-Cre hepatocytes. We further show that these cells can acquire 59Fe from 59Fe-transferrin, presumably via Tfr2. We conclude that Tfr1 is redundant for basal hepatocellular iron supply but essential for fine-tuning hepcidin responses according to the iron load of hepatocytes. Our data are consistent with an inhibitory function of Tfr1 on iron signaling to hepcidin via its interaction with Hfe. Moreover, they highlight hepatocellular Tfr1 as a link between cellular and systemic iron-regulatory pathways.
Collapse
|
46
|
Zhang S, Hu W, Yuan L, Yang Q. Transferrin receptor 1 is a supplementary receptor that assists transmissible gastroenteritis virus entry into porcine intestinal epithelium. Cell Commun Signal 2018; 16:69. [PMID: 30342530 PMCID: PMC6196004 DOI: 10.1186/s12964-018-0283-5] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2018] [Accepted: 10/11/2018] [Indexed: 12/15/2022] Open
Abstract
Background Transmissible gastroenteritis virus (TGEV), the etiologic agent of transmissible gastroenteritis, infects swine of all ages causing vomiting and diarrhea, in newborn piglets the mortality rate is near 100%. Intestinal epithelial cells are the primary target cells of TGEV. Transferrin receptor 1 (TfR1), which is highly expressed in piglets with anemia, may play a role in TGEV infection. However, the underlying mechanism of TGEV invasion remains largely unknown. Results Our study investigated the possibility that TfR1 can serve as a receptor for TGEV infection and enables the invasion and replication of TGEV. We observed that TGEV infection promoted TfR1 internalization, clustering, and co-localization with TfR1 early in infection, while TfR1 expression was significantly down-regulated as TGEV infection proceeded. TGEV infection and replication were inhibited by occluding TfR1 with antibodies or by decreasing TfR1 expression. TGEV infection increased in TGEV-susceptible ST or IPEC-J2 cell lines and TGEV-resistant Caco-2 cells when porcine TfR1 was over-expressed. Finally, we found that the TGEV S1 protein interacts with the extracellular region of TfR1, and that pre-incubating TGEV with a protein fragment containing the extracellular region of TfR1 blocked viral infection. Conclusions Our results support the hypothesis that TfR1 is an additional receptor for TGEV and assists TGEV invasion and replication. Electronic supplementary material The online version of this article (10.1186/s12964-018-0283-5) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Shuai Zhang
- College of Veterinary Medicine, Nanjing Agricultural University, Wei gang 1, Nanjing, Jiangsu, 210095, People's Republic of China
| | - Weiwei Hu
- College of Veterinary Medicine, Nanjing Agricultural University, Wei gang 1, Nanjing, Jiangsu, 210095, People's Republic of China
| | - Lvfeng Yuan
- College of Veterinary Medicine, Nanjing Agricultural University, Wei gang 1, Nanjing, Jiangsu, 210095, People's Republic of China
| | - Qian Yang
- College of Veterinary Medicine, Nanjing Agricultural University, Wei gang 1, Nanjing, Jiangsu, 210095, People's Republic of China.
| |
Collapse
|
47
|
Secreted IgD Amplifies Humoral T Helper 2 Cell Responses by Binding Basophils via Galectin-9 and CD44. Immunity 2018; 49:709-724.e8. [PMID: 30291028 DOI: 10.1016/j.immuni.2018.08.013] [Citation(s) in RCA: 64] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2018] [Revised: 08/05/2018] [Accepted: 08/09/2018] [Indexed: 01/24/2023]
Abstract
B cells thwart antigenic aggressions by releasing immunoglobulin M (IgM), IgG, IgA, and IgE, which deploy well-understood effector functions. In contrast, the role of secreted IgD remains mysterious. We found that some B cells generated IgD-secreting plasma cells following early exposure to external soluble antigens such as food proteins. Secreted IgD targeted basophils by interacting with the CD44-binding protein galectin-9. When engaged by antigen, basophil-bound IgD increased basophil secretion of interleukin-4 (IL-4), IL-5, and IL-13, which facilitated the generation of T follicular helper type 2 cells expressing IL-4. These germinal center T cells enhanced IgG1 and IgE but not IgG2a and IgG2b responses to the antigen initially recognized by basophil-bound IgD. In addition, IgD ligation by antigen attenuated allergic basophil degranulation induced by IgE co-ligation. Thus, IgD may link B cells with basophils to optimize humoral T helper type 2-mediated immunity against common environmental soluble antigens.
Collapse
|
48
|
Camaschella C, Pagani A. Advances in understanding iron metabolism and its crosstalk with erythropoiesis. Br J Haematol 2018; 182:481-494. [PMID: 29938779 DOI: 10.1111/bjh.15403] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Recent years have witnessed impressive advances in our understanding of iron metabolism. A number of studies of iron disorders and of their animal models have provided landmark insights into the mechanisms of iron trafficking, distribution and homeostatic regulation, the latter essential to prevent both iron deficiency and iron excess. Our perception of iron metabolism has been completely changed by an improved definition of cellular and systemic iron homeostasis, of the molecular pathogenesis of iron disorders, the fine tuning of the iron hormone hepcidin by activators and inhibitors and the dissection of the components of the hepcidin regulatory pathway. Important for haematology, the crosstalk of erythropoiesis, the most important iron consumer, and the hepcidin pathway has been at least partially clarified. Novel potential biomarkers are available and novel therapeutic targets for iron-related disorders have been tested in murine models. These preclinical studies provided proofs of principle and are laying the ground for clinical trials. Understanding iron control in tissues other than erythropoiesis remains a challenge for the future.
Collapse
Affiliation(s)
- Clara Camaschella
- Division of Genetics and Cell Biology, San Raffaele Scientific Institute and Vita Salute University, Milano, Italy
| | - Alessia Pagani
- Division of Genetics and Cell Biology, San Raffaele Scientific Institute and Vita Salute University, Milano, Italy
| |
Collapse
|
49
|
Wessling-Resnick M. Crossing the Iron Gate: Why and How Transferrin Receptors Mediate Viral Entry. Annu Rev Nutr 2018; 38:431-458. [PMID: 29852086 DOI: 10.1146/annurev-nutr-082117-051749] [Citation(s) in RCA: 100] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Because both the host and pathogen require iron, the innate immune response carefully orchestrates control over iron metabolism to limit its availability during times of infection. Nutritional iron deficiency can impair host immunity, while iron overload can cause oxidative stress to propagate harmful viral mutations. An emerging enigma is that many viruses use the primary gatekeeper of iron metabolism, the transferrin receptor, as a means to enter cells. Why and how this iron gate is a viral target for infection are the focus of this review.
Collapse
Affiliation(s)
- Marianne Wessling-Resnick
- Department of Genetics and Complex Diseases, Harvard T.H. Chan School of Public Health, Boston, Massachusetts 02115, USA;
| |
Collapse
|
50
|
Gammella E, Buratti P, Cairo G, Recalcati S. The transferrin receptor: the cellular iron gate. Metallomics 2018; 9:1367-1375. [PMID: 28671201 DOI: 10.1039/c7mt00143f] [Citation(s) in RCA: 191] [Impact Index Per Article: 27.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
The transferrin receptor (TfR1), which mediates cellular iron uptake through clathrin-dependent endocytosis of iron-loaded transferrin, plays a key role in iron homeostasis. Since the number of TfR1 molecules at the cell surface is the rate-limiting step for iron entry into cells and is essential to prevent iron overload, TfR1 expression is precisely controlled at multiple levels. In this review, we have discussed the latest advances in the molecular regulation of TfR1 expression and we have considered current understanding of TfR1 function beyond its canonical role in providing iron for erythroid precursors and rapidly proliferating cells.
Collapse
Affiliation(s)
- Elena Gammella
- Department of Biomedical Sciences for Health, University of Milan, Via Mangiagalli 31, 20133 Milano, Italy.
| | | | | | | |
Collapse
|