1
|
Liang X, Tian S, Zhang H, Sun S, Zhang P, Li J, Li Y, Zhang Y, Liu Z. Efferocytosis: A new star of atherosclerotic plaques reversal. Int Immunopharmacol 2025; 146:113904. [PMID: 39724733 DOI: 10.1016/j.intimp.2024.113904] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Revised: 12/16/2024] [Accepted: 12/16/2024] [Indexed: 12/28/2024]
Abstract
Efferocytosis is considered the key to eliminate apoptotic cells (ACs) under physiological and pathological conditions in vivo, mainly through different types of macrophages to achieve this process. Especially, tissue-resident macrophages (TRMs) are very significant for inflammation regression and maintenance of homeostasis in vivo. Abnormal efferocytosis will lead to the accumulation of ACs and the release of a variety of pro-inflammatory factors, which mediates the occurrence of many inflammatory diseases, including atherosclerosis (AS). AS is a chronic inflammatory vascular disease with the participation of the immune system. Defective efferocytosis will accelerate the progress of AS to a certain extent. Therefore, it is of great significance to understand the mechanism of efferocytosis and realize the prevention and treatment of AS through efferocytosis. In this review, we will briefly describe the specific process of efferocytosis, deeply discuss the possible molecular mechanism of impaired efferocytosis promoting the development of AS, and summarize the ways to prevent and treat AS through efferocytosis intervention therapy.
Collapse
Affiliation(s)
- Xiangyu Liang
- College of Pharmaceutical Sciences, Key Laboratory of Pharmaceutical Quality Control of Hebei Province, Hebei University, Baoding, China; State Key Laboratory of New Pharmaceutical Preparations and Excipients, Baoding, China.
| | - Shuoqi Tian
- College of Pharmaceutical Sciences, Key Laboratory of Pharmaceutical Quality Control of Hebei Province, Hebei University, Baoding, China; State Key Laboratory of New Pharmaceutical Preparations and Excipients, Baoding, China.
| | - Han Zhang
- College of Pharmaceutical Sciences, Key Laboratory of Pharmaceutical Quality Control of Hebei Province, Hebei University, Baoding, China; State Key Laboratory of New Pharmaceutical Preparations and Excipients, Baoding, China.
| | - Shusen Sun
- College of Pharmaceutical Sciences, Key Laboratory of Pharmaceutical Quality Control of Hebei Province, Hebei University, Baoding, China; State Key Laboratory of New Pharmaceutical Preparations and Excipients, Baoding, China.
| | - Peixiang Zhang
- College of Pharmaceutical Sciences, Key Laboratory of Pharmaceutical Quality Control of Hebei Province, Hebei University, Baoding, China; State Key Laboratory of New Pharmaceutical Preparations and Excipients, Baoding, China.
| | - Jiameng Li
- College of Pharmaceutical Sciences, Key Laboratory of Pharmaceutical Quality Control of Hebei Province, Hebei University, Baoding, China; State Key Laboratory of New Pharmaceutical Preparations and Excipients, Baoding, China.
| | - Yong Li
- Beijing Yongkang Nian Health Technology Co., Ltd., Beijing, China.
| | - Yanfen Zhang
- Technology Transfer Center, Hebei University, Baoding, China.
| | - Zhongcheng Liu
- College of Pharmaceutical Sciences, Key Laboratory of Pharmaceutical Quality Control of Hebei Province, Hebei University, Baoding, China; State Key Laboratory of New Pharmaceutical Preparations and Excipients, Baoding, China.
| |
Collapse
|
2
|
Li Y, Zhou Q, Zhang K, Meng X. Iron Overload and Abdominal Aortic Aneurysm. Rev Cardiovasc Med 2024; 25:361. [PMID: 39484115 PMCID: PMC11522754 DOI: 10.31083/j.rcm2510361] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2024] [Revised: 04/15/2024] [Accepted: 04/19/2024] [Indexed: 11/03/2024] Open
Abstract
Abdominal aortic aneurysm (AAA) is a chronic vascular degenerative disease characterized by progressive segmental dilation of the abdominal aorta. The rupture of an AAA represents a leading cause of death in cardiovascular diseases. Despite numerous experimental and clinical studies examining potential drug targets and therapies, currently there are no pharmaceutical treatment to prevent AAA growth and rupture. Iron is an essential element in almost all living organisms and has important biological functions. Epidemiological studies have indicated that both iron deficiency and overload are associated with adverse clinical outcomes, particularly an increased risk of cardiovascular events. Recent evidence indicates that iron overload is involved in the pathogenesis of abdominal aortic aneurysms. In this review, we provide an overview of the role of iron overload in AAA progression and explore its potential pathological mechanisms. Although the exact molecular mechanisms of iron overload in the development of AAA remain to be elucidated, the inhibition of iron deposition may offer a promising strategy for preventing these aneurysms.
Collapse
Affiliation(s)
- Yunyi Li
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Department of Cardiology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, 250012 Jinan, Shandong, China
| | - Quan Zhou
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Department of Cardiology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, 250012 Jinan, Shandong, China
| | - Kai Zhang
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Department of Cardiology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, 250012 Jinan, Shandong, China
| | - Xiao Meng
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Department of Cardiology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, 250012 Jinan, Shandong, China
| |
Collapse
|
3
|
Mantovani A, Busti F, Borella N, Scoccia E, Pecoraro B, Sani E, Morandin R, Csermely A, Piasentin D, Grespan E, Castagna A, Bilson J, Byrne CD, Valenti L, Girelli D, Targher G. Elevated plasma hepcidin concentrations are associated with an increased risk of mortality and nonfatal cardiovascular events in patients with type 2 diabetes: a prospective study. Cardiovasc Diabetol 2024; 23:305. [PMID: 39154180 PMCID: PMC11330614 DOI: 10.1186/s12933-024-02377-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/27/2024] [Accepted: 07/25/2024] [Indexed: 08/19/2024] Open
Abstract
BACKGROUND The effect of plasma hepcidin concentrations on the long-term risk of developing adverse cardiovascular outcomes in people with type 2 diabetes mellitus (T2DM) is unclear. METHODS We followed for a median of 55.6 months 213 outpatients with established T2DM (45.5% women, mean age 69 ± 10 years; BMI 28.7 ± 4.7 kg/m2; median diabetes duration 11 years). Baseline plasma ferritin and hepcidin concentrations were measured with an electrochemiluminescence immunoassay and mass spectrometry-based assay, respectively. The primary study outcome was a composite of all-cause mortality or incident nonfatal cardiovascular events (inclusive of myocardial infarction, permanent atrial fibrillation, ischemic stroke, or new hospitalization for heart failure). RESULTS 42 patients developed the primary composite outcome over a median follow-up of 55.6 months. After stratifying patients by baseline hepcidin tertiles [1st tertile: median hepcidin 1.04 (IQR 0.50-1.95) nmol/L, 2nd tertile: 3.81 (IQR 3.01-4-42) nmol/L and 3rd tertile: 7.72 (IQR 6.37-10.4) nmol/L], the risk of developing the primary composite outcome in patients in the 3rd tertile was double that of patients in the 1st and 2nd tertile combined (unadjusted hazard ratio [HR] 2.32, 95%CI 1.27-4.26; p = 0.007). This risk was not attenuated after adjustment for age, sex, adiposity measures, smoking, hypertension, statin use, antiplatelet medication use, plasma hs-C-reactive protein and ferritin concentrations (adjusted HR 2.53, 95%CI 1.27-5.03; p = 0.008). CONCLUSIONS In outpatients with T2DM, higher baseline hepcidin concentrations were strongly associated with an increased long-term risk of overall mortality or nonfatal cardiovascular events, even after adjustment for established cardiovascular risk factors, plasma ferritin concentrations, medication use, and other potential confounders.
Collapse
Affiliation(s)
- Alessandro Mantovani
- Department of Medicine, Section of Endocrinology, Diabetes and Metabolism, University of Verona and Azienda Ospedaliera Universitaria Integrata of Verona, Verona, Italy
| | - Fabiana Busti
- Department of Medicine, Section of Internal Medicine, EuroBloodNet Center, University of Verona and Azienda Ospedaliera Universitaria Integrata of Verona, Verona, Italy
| | - Nicolò Borella
- Department of Medicine, Section of Endocrinology, Diabetes and Metabolism, University of Verona and Azienda Ospedaliera Universitaria Integrata of Verona, Verona, Italy
| | - Enrico Scoccia
- Department of Medicine, Section of Endocrinology, Diabetes and Metabolism, University of Verona and Azienda Ospedaliera Universitaria Integrata of Verona, Verona, Italy
| | - Barbara Pecoraro
- Department of Medicine, Section of Endocrinology, Diabetes and Metabolism, University of Verona and Azienda Ospedaliera Universitaria Integrata of Verona, Verona, Italy
| | - Elena Sani
- Department of Medicine, Section of Endocrinology, Diabetes and Metabolism, University of Verona and Azienda Ospedaliera Universitaria Integrata of Verona, Verona, Italy
| | - Riccardo Morandin
- Department of Medicine, Section of Endocrinology, Diabetes and Metabolism, University of Verona and Azienda Ospedaliera Universitaria Integrata of Verona, Verona, Italy
| | - Alessandro Csermely
- Department of Medicine, Section of Endocrinology, Diabetes and Metabolism, University of Verona and Azienda Ospedaliera Universitaria Integrata of Verona, Verona, Italy
| | - Daniele Piasentin
- Department of Medicine, Section of Endocrinology, Diabetes and Metabolism, University of Verona and Azienda Ospedaliera Universitaria Integrata of Verona, Verona, Italy
| | - Elisabetta Grespan
- Department of Medicine, Section of Endocrinology, Diabetes and Metabolism, University of Verona and Azienda Ospedaliera Universitaria Integrata of Verona, Verona, Italy
| | - Annalisa Castagna
- Department of Medicine, Section of Internal Medicine, EuroBloodNet Center, University of Verona and Azienda Ospedaliera Universitaria Integrata of Verona, Verona, Italy
| | - Josh Bilson
- School of Human Development and Health, Faculty of Medicine, University of Southampton, Southampton, UK
- National Institute for Health and Care Research Southampton Biomedical Research Centre, University Hospital Southampton, National Health Service Foundation Trust, Southampton, UK
| | - Christopher D Byrne
- School of Human Development and Health, Faculty of Medicine, University of Southampton, Southampton, UK
- National Institute for Health and Care Research Southampton Biomedical Research Centre, University Hospital Southampton, National Health Service Foundation Trust, Southampton, UK
| | - Luca Valenti
- Department of Transfusion Medicine, Precision Medicine Lab, Biological Resource Center, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milano, Italy
- Department of Pathophysiology and Transplantation, Università degli Studi di Milano, Milano, Italy
| | - Domenico Girelli
- Department of Medicine, Section of Internal Medicine, EuroBloodNet Center, University of Verona and Azienda Ospedaliera Universitaria Integrata of Verona, Verona, Italy
| | - Giovanni Targher
- Department of Medicine, University of Verona, Verona, Italy.
- Metabolic Diseases Research Unit, IRCCS Sacro Cuore - Don Calabria Hospital, Viale L. Rizzardi, 4, 37024, Negrar di Valpolicella, Italy.
| |
Collapse
|
4
|
Zhou QY, Ren C, Li JY, Wang L, Duan Y, Yao RQ, Tian YP, Yao YM. The crosstalk between mitochondrial quality control and metal-dependent cell death. Cell Death Dis 2024; 15:299. [PMID: 38678018 PMCID: PMC11055915 DOI: 10.1038/s41419-024-06691-w] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2023] [Revised: 04/16/2024] [Accepted: 04/17/2024] [Indexed: 04/29/2024]
Abstract
Mitochondria are the centers of energy and material metabolism, and they also serve as the storage and dispatch hubs of metal ions. Damage to mitochondrial structure and function can cause abnormal levels and distribution of metal ions, leading to cell dysfunction and even death. For a long time, mitochondrial quality control pathways such as mitochondrial dynamics and mitophagy have been considered to inhibit metal-induced cell death. However, with the discovery of new metal-dependent cell death including ferroptosis and cuproptosis, increasing evidence shows that there is a complex relationship between mitochondrial quality control and metal-dependent cell death. This article reviews the latest research results and mechanisms of crosstalk between mitochondrial quality control and metal-dependent cell death in recent years, as well as their involvement in neurodegenerative diseases, tumors and other diseases, in order to provide new ideas for the research and treatment of related diseases.
Collapse
Affiliation(s)
- Qi-Yuan Zhou
- Department of Emergency, the Second Hospital of Hebei Medical University, Shijiazhuang, 050000, China
| | - Chao Ren
- Department of Pulmonary and Critical Care Medicine, Beijing Chaoyang Hospital, Capital Medical University, Beijing, 100020, China
| | - Jing-Yan Li
- Department of Emergency, the Second Hospital of Hebei Medical University, Shijiazhuang, 050000, China
| | - Lu Wang
- Department of Critical Care Medicine, the First Medical Center of Chinese PLA General Hospital, Beijing, 100853, China
| | - Yu Duan
- Department of Critical Care Medicine, Affiliated Chenzhou Hospital (the First People's Hospital of Chenzhou), Southern Medical University, Chenzhou, 423000, China
| | - Ren-Qi Yao
- Department of General Surgery, the First Medical Center of Chinese PLA General Hospital, Beijing, 100853, China.
- Medical Innovation Research Division, Translational Medicine Research Center and the Fourth Medical Center of Chinese PLA General Hospital, Beijing, 100853, China.
| | - Ying-Ping Tian
- Department of Emergency, the Second Hospital of Hebei Medical University, Shijiazhuang, 050000, China.
| | - Yong-Ming Yao
- Medical Innovation Research Division, Translational Medicine Research Center and the Fourth Medical Center of Chinese PLA General Hospital, Beijing, 100853, China.
| |
Collapse
|
5
|
Yuan XM, Sultana N, Ghosh-Laskar M, Li W. Elevated Hepcidin Expression in Human Carotid Atheroma: Sex-Specific Differences and Associations with Plaque Vulnerability. Int J Mol Sci 2024; 25:1706. [PMID: 38338987 PMCID: PMC10855936 DOI: 10.3390/ijms25031706] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2023] [Revised: 01/19/2024] [Accepted: 01/27/2024] [Indexed: 02/12/2024] Open
Abstract
Hepcidin is upregulated by increased body iron stores and inflammatory cytokines. It is associated with cardiovascular events, arterial stiffness, and increased iron accumulation in human atheroma with hemorrhage. However, it is unknown whether the expression of hepcidin in human carotid plaques is related to plaque severity and whether hepcidin expression differs between men and women. Carotid samples from 58 patients (38 males and 20 females) were immunostained with hepcidin, macrophages, ferritin, and transferrin receptor. Immunocytochemistry of hepcidin was performed on THP-1 macrophages exposed to iron or 7betahydroxycholesterol. Hepcidin expression significantly increases with the progression of human atherosclerotic plaques. Plaques of male patients have significantly higher levels of hepcidin. Expressions of hepcidin are significantly correlated with the accumulation of CD68-positive macrophages and transferrin receptor 1 (TfR1) and apoptosis. In vitro, hepcidin is significantly increased in macrophages exposed to iron and moderately increased following 7-oxysterol treatment. In the cultured cells, suppression of hepcidin protected against macrophage cell death, lysosomal membrane permeabilization, and oxidative stress. Hepcidin may play a crucial role in the development and progression of atherosclerosis. The differential expression of hepcidin in male and female patients and its significant correlations with plaque severity, highlight the potential of hepcidin as a biomarker for risk stratification and therapeutic targeting in atherosclerosis.
Collapse
Affiliation(s)
- Xi-Ming Yuan
- Occupational and Environmental Medicine, Department of Health, Medicine and Caring Sciences, Linköping University, 581 85 Linköping, Sweden;
| | - Nargis Sultana
- Laboratory Medicine, Linköping University Hospital, 581 85 Linköping, Sweden;
| | | | - Wei Li
- Obstetrics and Gynaecology in Linköping, Department of Biomedical and Clinical Sciences, Linköping University, 581 85 Linköping, Sweden
| |
Collapse
|
6
|
Han Z, Hu H, Yin M, Lin Y, Yan Y, Han P, Liu B, Jing B. HOXA1 participates in VSMC-to-macrophage-like cell transformation via regulation of NF-κB p65 and KLF4: a potential mechanism of atherosclerosis pathogenesis. Mol Med 2023; 29:104. [PMID: 37528397 PMCID: PMC10394793 DOI: 10.1186/s10020-023-00685-8] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2022] [Accepted: 06/12/2023] [Indexed: 08/03/2023] Open
Abstract
BACKGROUND Macrophage-like transformation of vascular smooth muscle cells (VSMCs) is a risk factor of atherosclerosis (AS) progression. Transcription factor homeobox A1 (HOXA1) plays functional roles in differentiation and development. This study aims to explore the role of HOXA1 in VSMC transformation, thereby providing evidence for the potential mechanism of AS pathogenesis. METHODS High fat diet (HFD)-fed apolipoprotein E knockout (ApoE-/-) mice were applied as an in vivo model to imitate AS, while 1-palmitoyl-2-(5-oxovaleroyl)-sn-glycero-3-phosphocholine (POV-PC)-treated VSMCs were applied as an in vitro model. Recombinant adeno-associated-virus-1 (AAV-1) vectors that express short-hairpin RNAs targeting HOXA1, herein referred as AAV1-shHOXA1, were generated for the loss-of-function experiments throughout the study. RESULTS In the aortic root of AS mice, lipid deposition was severer and HOXA1 expression was higher than the wide-type mice fed with normal diet or HFD. Silencing of HOXA1 inhibited the AS-induced weight gain, inflammatory response, serum and liver lipid metabolism disorder and atherosclerotic plaque formation. Besides, lesions from AS mice with HOXA1 knockdown showed less trans-differentiation of VSMCs to macrophage-like cells, along with a suppression of krüppel-like factor 4 (KLF4) and nuclear factor (NF)-κB RelA (p65) expression. In vitro experiments consistently confirmed that HOXA1 knockdown suppressed lipid accumulation, VSMC-to-macrophage phenotypic switch and inflammation in POV-PC-treated VSMCs. Mechanism investigations further illustrated that HOXA1 transcriptionally activated RelA and KLF4 to participate in the pathological manifestations of VSMCs. CONCLUSIONS HOXA1 participates in AS progression by regulating VSMCs plasticity via regulation of NF-κB p65 and KLF4. HOXA1 has the potential to be a biomarker or therapeutic target for AS.
Collapse
Affiliation(s)
- Zhiyang Han
- Department of Vascular Surgery, The First Affiliated Hospital of Harbin Medical University, No. 23, Youzheng Street, Harbin, 150001, Heilongjiang, China
| | - Haidi Hu
- Department of General and Vascular Surgery, Shengjing Hospital of China Medical University, Shenyang, 110001, Liaoning, China
| | - MingZhu Yin
- Department of Dermatology, Xiangya Hospital Central South University, Changsha, 410008, Hunan, China
- Human Engineering Research Center of Skin Health and Disease, Changsha, 410008, Hunan, China
| | - Yu Lin
- Department of Vascular Surgery, The First Affiliated Hospital of Harbin Medical University, No. 23, Youzheng Street, Harbin, 150001, Heilongjiang, China
| | - Yan Yan
- Department of Vascular Surgery, The First Affiliated Hospital of Harbin Medical University, No. 23, Youzheng Street, Harbin, 150001, Heilongjiang, China
| | - Peng Han
- Department of Vascular Surgery, The First Affiliated Hospital of Harbin Medical University, No. 23, Youzheng Street, Harbin, 150001, Heilongjiang, China
| | - Bing Liu
- Department of Vascular Surgery, The First Affiliated Hospital of Harbin Medical University, No. 23, Youzheng Street, Harbin, 150001, Heilongjiang, China
| | - Bao Jing
- Department of Vascular Surgery, The First Affiliated Hospital of Harbin Medical University, No. 23, Youzheng Street, Harbin, 150001, Heilongjiang, China.
| |
Collapse
|
7
|
Xie L, Fang B, Zhang C. The role of ferroptosis in metabolic diseases. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2023; 1870:119480. [PMID: 37127193 DOI: 10.1016/j.bbamcr.2023.119480] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/08/2022] [Revised: 04/20/2023] [Accepted: 04/20/2023] [Indexed: 05/03/2023]
Abstract
The annual incidence of metabolic diseases such as diabetes, non-alcoholic fatty liver disease (NAFLD), osteoporosis, and atherosclerosis (AS) is increasing, resulting in a heavy burden on human health and the social economy. Ferroptosis is a novel form of programmed cell death driven by iron-dependent lipid peroxidation, which was discovered in recent years. Emerging evidence has suggested that ferroptosis contributes to the development of metabolic diseases. Here, we summarize the mechanisms and molecular signaling pathways involved in ferroptosis. Then we discuss the role of ferroptosis in metabolic diseases. Finally, we analyze the potential of targeting ferroptosis as a promising therapeutic approach for metabolic diseases.
Collapse
Affiliation(s)
- Ling Xie
- Department of Nephrology, Union Hospital, Tongji Medical College of Huazhong University of Science and Technology, Wuhan 430072, Hubei, China
| | - Bin Fang
- Department of Nephrology, Union Hospital, Tongji Medical College of Huazhong University of Science and Technology, Wuhan 430072, Hubei, China
| | - Chun Zhang
- Department of Nephrology, Union Hospital, Tongji Medical College of Huazhong University of Science and Technology, Wuhan 430072, Hubei, China.
| |
Collapse
|
8
|
Hossainey MRH, Yaparla A, Uzzaman Z, Moore T, Grayfer L. A comparison of amphibian (Xenopus laevis) tadpole and adult frog macrophages. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2023; 141:104647. [PMID: 36707039 DOI: 10.1016/j.dci.2023.104647] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/19/2022] [Revised: 12/30/2022] [Accepted: 01/23/2023] [Indexed: 06/18/2023]
Abstract
The amphibian declines are compounded by emerging pathogens that often preferentially target distinct amphibian developmental stages. While amphibian immune responses remain relatively unexplored, macrophage (Mφ)-lineage cells are believed to be important to both amphibian host defenses and to their pathogen infection strategies. As such, a greater understanding of tadpole and adult amphibian Mφ functionality is warranted. Mφ biology is interdependent of interleukin-34 (IL-34) and colony-stimulating factor-1 (CSF-1) cytokines and we previously showed that CSF-1- and IL-34-derived Mφs of the Xenopus laevis frog are morphologically, transcriptionally, and functionally distinct. Presently, we directly compared the cytology and transcriptomes of X. laevis tadpole and frog CSF-1- and IL-34-Mφs. Our results indicate that tadpole and frog CSF-1-Mφs possess greater non-specific esterase activity, typically associated with Mφ-lineage cells. By contrast, both tadpole and frog IL-34-Mφs have greater specific esterase activity, which is typically attributed to granulocyte-lineage cells. Our comparisons of tadpole CSF-1-Mφ transcriptomes with those of tadpole IL-34-Mφs indicate that the two tadpole populations possess significantly different transcriptional profiles of immune and non-immune genes. The frog CSF-1-Mφ gene expression profiles are likewise significantly disparate from those of frog IL-34-Mφs. Compared to their respective tadpole Mφ subtypes, frog CSF-1- and IL-34-Mφs exhibited greater expression of genes associated with antigen presentation. Conversely, compared to their frog Mφ counterparts, tadpole CSF-1- and IL-34-Mφs possessed greater levels of select Fc-like receptor genes. Presumably, these cytological and transcriptional differences manifest in distinct biological roles for these respective tadpole and frog Mφ subtypes.
Collapse
Affiliation(s)
| | - Amulya Yaparla
- Department of Biological Sciences, George Washington University, Washington, DC, 20052, USA
| | - Zarafsha Uzzaman
- Department of Biological Sciences, George Washington University, Washington, DC, 20052, USA
| | - Tyler Moore
- Department of Biological Sciences, George Washington University, Washington, DC, 20052, USA
| | - Leon Grayfer
- Department of Biological Sciences, George Washington University, Washington, DC, 20052, USA.
| |
Collapse
|
9
|
Rafati Rahimzadeh M, Rafati Rahimzadeh M, Kazemi S, Moghadamnia AR, Ghaemi Amiri M, Moghadamnia AA. Iron; Benefits or threatens (with emphasis on mechanism and treatment of its poisoning). Hum Exp Toxicol 2023; 42:9603271231192361. [PMID: 37526177 DOI: 10.1177/09603271231192361] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/02/2023]
Abstract
Iron is a necessary biological element and one of the richest in the human body, but it can cause changes in cell function and activity control. Iron is involved in a wide range of oxidation - reduction activities. Whenever iron exceeds the cellular metabolic needs, its excess causes changes in the products of cellular respiration, such as superoxide, hydrogen peroxide and hydroxyl. The formation of these compounds causes cellular toxicity. Lack of control over reactive oxygen species causes damages to DNA, proteins, and lipids. Conversely, superoxide, hydrogen peroxide and hydroxyl are reactive oxygen species, using antioxidants, restoring DNA function, and controlling iron stores lead to natural conditions. Iron poisoning causes clinical manifestations in the gastrointestinal tract, liver, heart, kidneys, and hematopoietic system. When serum iron is elevated, serum iron concentrations, total iron-binding capacity (TIBC) and ferritin will also increase. Supportive care is provided by whole bowel irrigation (WBI), esophagogastroduodenoscopy is required to evaluate mucosal injury and remove undissolved iron tablets. The use of chelator agents such as deferoxamine mesylate, deferasirox, deferiprone, deferitrin are very effective in removing excess iron. Of course, the combined treatment of these chelators plays an important role in increasing iron excretion, and reducing side effects.
Collapse
Affiliation(s)
| | | | - Sohrab Kazemi
- Cellular and Molecular Biology Research Center, Health Research Institute, Babol University of Medical Sciences, Babol, Iran
| | | | - Maryam Ghaemi Amiri
- Faculty of Education Development Center (EDC), Babol University of Medical Sciences, Babol, Iran
| | - Ali Akbar Moghadamnia
- Cellular and Molecular Biology Research Center, Health Research Institute, Babol University of Medical Sciences, Babol, Iran
| |
Collapse
|
10
|
Bukhari SNA, Yogesh R. An Overview of Tetramethylpyrazine (Ligustrazine) and its Derivatives as
Potent Anti-Alzheimer’s Disease Agents. LETT DRUG DES DISCOV 2022. [DOI: 10.2174/1570180819666220405232333] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Abstract:
Tetramethylpyrazine (TMP), or ligustrazine, is an alkaloid isolated from the Chinese herb
Ligusticum wallichii. It is known for its broad-spectrum medicinal properties against several diseases, and
various studies have shown that it can modulate diverse biological targets and signaling pathways to produce
neuroprotective effects, especially against Alzheimer’s disease (AD). This has attracted significant
research attention evaluating TMP as a potent multitarget anti-AD agent. This review compiles the results
of studies assessing the neuroprotective mechanisms exerted by TMP as well as its derivatives prepared
using a multi-target-directed ligand strategy to explore its multitarget modulating properties. The present
review also highlights the work done on the design, synthesis, structure-activity relationships, and mechanisms
of some potent TMP derivatives that have shown promising anti-AD activities. These derivatives
were designed, synthesized, and evaluated to develop anti-AD molecules with enhanced biological and
pharmacokinetic activities compared to TMP. This review article paves the way for the exploration and
development of TMP and TMP derivatives as an effective treatment for AD.
Collapse
Affiliation(s)
- Syed Nasir Abbas Bukhari
- Department of Pharmaceutical Chemistry, College of Pharmacy, Jouf University, Sakaka, Aljouf 2014, Saudi Arabia
| | - Ruchika Yogesh
- 22 A3, DS Tower 1, Sukhumvit Soi 33, Khlong Tan Nuea, Wattana, Bangkok 10110, Thailand
| |
Collapse
|
11
|
Naito Y, Tsujino T, Masuyama T, Ishihara M. Crosstalk between Iron and Arteriosclerosis. J Atheroscler Thromb 2022; 29:308-314. [PMID: 34421089 PMCID: PMC8894107 DOI: 10.5551/jat.rv17060] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2021] [Accepted: 07/25/2021] [Indexed: 11/11/2022] Open
Abstract
Iron is an important element for life; however, intracellular labile iron overload can lead to the generation of reactive oxygen species and cellular damage. Although iron is mainly utilized for heme synthesis and is incorporated into hemoglobin, body iron status is often implicated in the pathogenesis of cardiovascular diseases. In a cell, iron is used for basic processes such as cell growth, maintenance, and repair. Thus, iron is considered to be involved in the pathogenesis of arteriosclerosis. In fact, clinical and experimental studies have shown an association between iron and arteriosclerosis. These data suggest the crosstalk between iron and arteriosclerosis. However, iron metabolism in arteriosclerosis is often complicated, and the systemic and cellular mechanisms of iron homeostasis in arteriosclerosis remain completely unsolved. Thus, in this review, we aimed to examine the role of iron in arteriosclerosis.
Collapse
Affiliation(s)
- Yoshiro Naito
- Department of Cardiovascular and Renal Medicine, Hyogo College of Medicine, Nishinomiya, Japan
| | - Takeshi Tsujino
- Department of Cardiovascular and Renal Medicine, Hyogo College of Medicine, Nishinomiya, Japan
- Division of Pharmaceutical Therapeutics, Department of Pharmacy, School of Pharmacy, Hyogo University of Health Sciences, Kobe, Japan
| | - Tohru Masuyama
- Department of Cardiovascular and Renal Medicine, Hyogo College of Medicine, Nishinomiya, Japan
- Japan Community Health Care Organization, Hoshigaoka Medical Center, Hirakata, Japan
| | - Masaharu Ishihara
- Department of Cardiovascular and Renal Medicine, Hyogo College of Medicine, Nishinomiya, Japan
| |
Collapse
|
12
|
Jin H, Chen P, Zhang S, Wu P, Yu X. Iron Metabolism Markers and Lower Extremity Arterial Disease in People with Type 2 Diabetes. Diabetes Metab Syndr Obes 2022; 15:3103-3110. [PMID: 36237965 PMCID: PMC9553230 DOI: 10.2147/dmso.s380803] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Accepted: 09/16/2022] [Indexed: 11/23/2022] Open
Abstract
OBJECTIVE To determine the levels of serum iron, ferritin, total iron-binding capacity, and hepcidin in patients with type 2 diabetes mellitus (T2DM), and to elucidate the relationship of these biomarkers with lower extremity arterial disease (LEAD). METHODS Three hundred fifteen patients with T2DM were selected for the study and divided into non-LEAD (n = 119) and LEAD groups (n=196) based on the ankle-brachial index (ABI) results. Demographic data and clinical test results were collected from all patients. Serum iron, ferritin, total iron-binding capacity, and hepcidin levels were measured, and the transferrin saturation was calculated. RESULTS Hepcidin levels were substantially higher in the LEAD group (19.17 ± 8.66 ng/mL) than the non-LEAD group (15.44±7.55 ng/mL, P < 0.001), and there was a negative correlation between the ABI and serum lecithin level (r = -0.349, P < 0.001). There were no other correlations with the other iron metabolism indicators. The results of dichotomous logistic regression with LEAD as the dependent variable revealed that smoking history (OR = 4.442, P = 0.008), hypertension history (OR = 3.721, P = 0.006), cardiovascular disease history (OR = 11.126, P < 0.001), diabetes duration (OR = 1.305, P < 0.001), age (OR = 1.056, P = 0.021), hs-CRP level (OR = 1.376, P = 0.002), HbA1c concentration (OR = 1.394, P = 0.001), and hepcidin level (OR = 1.097, P = 0.003) were independent risk factors for LEAD in T2DM patients. CONCLUSION Serum hepcidin levels were elevated in the LEAD group compared with the non-LEAD group, and elevated hepcidin levels were associated with the development of LEAD in T2DM patients, suggesting that hepcidin may be involved in the occurrence and development of LEAD in T2DM patients.
Collapse
Affiliation(s)
- Hua Jin
- Department of Endocrinology and Metabolism, Fengxian Central Hospital, Shanghai, 201404, People’s Republic of China
| | - Peihong Chen
- Department of Endocrinology and Metabolism, Fengxian Central Hospital, Shanghai, 201404, People’s Republic of China
| | - Shan Zhang
- Department of Endocrinology and Metabolism, Fengxian Central Hospital, Shanghai, 201404, People’s Republic of China
| | - Ping Wu
- Department of Endocrinology and Metabolism, Fengxian Central Hospital, Shanghai, 201404, People’s Republic of China
| | - Xuemei Yu
- Department of Endocrinology and Metabolism, Fengxian Central Hospital, Shanghai, 201404, People’s Republic of China
- Correspondence: Xuemei Yu, Department of Endocrinology and Metabolism, Fengxian Central Hospital, No. 6600, Nanfeng Road, Nanqiaoxincheng, Fengxian District, Shanghai, 201404, People’s Republic of China, Tel +86 21-57413468, Email
| |
Collapse
|
13
|
Turpin C, Catan A, Meilhac O, Bourdon E, Canonne-Hergaux F, Rondeau P. Erythrocytes: Central Actors in Multiple Scenes of Atherosclerosis. Int J Mol Sci 2021; 22:ijms22115843. [PMID: 34072544 PMCID: PMC8198892 DOI: 10.3390/ijms22115843] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2021] [Revised: 05/26/2021] [Accepted: 05/26/2021] [Indexed: 12/16/2022] Open
Abstract
The development and progression of atherosclerosis (ATH) involves lipid accumulation, oxidative stress and both vascular and blood cell dysfunction. Erythrocytes, the main circulating cells in the body, exert determinant roles in the gas transport between tissues. Erythrocytes have long been considered as simple bystanders in cardiovascular diseases, including ATH. This review highlights recent knowledge concerning the role of erythrocytes being more than just passive gas carriers, as potent contributors to atherosclerotic plaque progression. Erythrocyte physiology and ATH pathology is first described. Then, a specific chapter delineates the numerous links between erythrocytes and atherogenesis. In particular, we discuss the impact of extravasated erythrocytes in plaque iron homeostasis with potential pathological consequences. Hyperglycaemia is recognised as a significant aggravating contributor to the development of ATH. Then, a special focus is made on glycoxidative modifications of erythrocytes and their role in ATH. This chapter includes recent data proposing glycoxidised erythrocytes as putative contributors to enhanced atherothrombosis in diabetic patients.
Collapse
Affiliation(s)
- Chloé Turpin
- Diabète Athérothrombose Thérapies Réunion Océan Indien (DéTROI), INSERM, UMR 1188, Université de La Réunion, 97400 Saint Denis, France; (C.T.); (A.C.); (O.M.); (E.B.)
| | - Aurélie Catan
- Diabète Athérothrombose Thérapies Réunion Océan Indien (DéTROI), INSERM, UMR 1188, Université de La Réunion, 97400 Saint Denis, France; (C.T.); (A.C.); (O.M.); (E.B.)
| | - Olivier Meilhac
- Diabète Athérothrombose Thérapies Réunion Océan Indien (DéTROI), INSERM, UMR 1188, Université de La Réunion, 97400 Saint Denis, France; (C.T.); (A.C.); (O.M.); (E.B.)
- Centre Hospitalier Universitaire de La Réunion, 97400 Saint Denis, France
| | - Emmanuel Bourdon
- Diabète Athérothrombose Thérapies Réunion Océan Indien (DéTROI), INSERM, UMR 1188, Université de La Réunion, 97400 Saint Denis, France; (C.T.); (A.C.); (O.M.); (E.B.)
| | | | - Philippe Rondeau
- Diabète Athérothrombose Thérapies Réunion Océan Indien (DéTROI), INSERM, UMR 1188, Université de La Réunion, 97400 Saint Denis, France; (C.T.); (A.C.); (O.M.); (E.B.)
- Correspondence: ; Tel.: +262(0)-2-62-93-88-43; Fax: +262-(0)-2-62-93-88-01
| |
Collapse
|
14
|
Xia Y, Li Y, Wu X, Zhang Q, Chen S, Ma X, Yu M. Ironing Out the Details: How Iron Orchestrates Macrophage Polarization. Front Immunol 2021; 12:669566. [PMID: 34054839 PMCID: PMC8149954 DOI: 10.3389/fimmu.2021.669566] [Citation(s) in RCA: 59] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Accepted: 04/26/2021] [Indexed: 12/12/2022] Open
Abstract
Iron fine-tunes innate immune responses, including macrophage inflammation. In this review, we summarize the current understanding about the iron in dictating macrophage polarization. Mechanistically, iron orchestrates macrophage polarization through several aspects, including cellular signaling, cellular metabolism, and epigenetic regulation. Therefore, iron modulates the development and progression of multiple macrophage-associated diseases, such as cancer, atherosclerosis, and liver diseases. Collectively, this review highlights the crucial role of iron for macrophage polarization, and indicates the potential application of iron supplementation as an adjuvant therapy in different inflammatory disorders relative to the balance of macrophage polarization.
Collapse
Affiliation(s)
- Yaoyao Xia
- State Key Laboratory of Livestock and Poultry Breeding, Key Laboratory of Animal Nutrition and Feed Science in South China, Ministry of Agriculture and Rural Affairs, Guangdong Provincial Key Laboratory of Animal Breeding and Nutrition, Guangdong Engineering Technology Research Center of Animal Meat Quality and Safety Control and Evaluation, Institute of Animal Science, Guangdong Academy of Agricultural Sciences, Guangzhou, China.,College of Animal Science, South China Agricultural University, Guangzhou, China
| | - Yikun Li
- College of Animal Science, South China Agricultural University, Guangzhou, China
| | - Xiaoyan Wu
- College of Animal Science, South China Agricultural University, Guangzhou, China
| | - Qingzhuo Zhang
- College of Animal Science, South China Agricultural University, Guangzhou, China
| | - Siyuan Chen
- College of Animal Science, South China Agricultural University, Guangzhou, China
| | - Xianyong Ma
- State Key Laboratory of Livestock and Poultry Breeding, Key Laboratory of Animal Nutrition and Feed Science in South China, Ministry of Agriculture and Rural Affairs, Guangdong Provincial Key Laboratory of Animal Breeding and Nutrition, Guangdong Engineering Technology Research Center of Animal Meat Quality and Safety Control and Evaluation, Institute of Animal Science, Guangdong Academy of Agricultural Sciences, Guangzhou, China.,Maoming Branch, Guangdong Laboratory for Lingnan Modern Agriculture, Guangzhou, China
| | - Miao Yu
- State Key Laboratory of Livestock and Poultry Breeding, Key Laboratory of Animal Nutrition and Feed Science in South China, Ministry of Agriculture and Rural Affairs, Guangdong Provincial Key Laboratory of Animal Breeding and Nutrition, Guangdong Engineering Technology Research Center of Animal Meat Quality and Safety Control and Evaluation, Institute of Animal Science, Guangdong Academy of Agricultural Sciences, Guangzhou, China.,Maoming Branch, Guangdong Laboratory for Lingnan Modern Agriculture, Guangzhou, China
| |
Collapse
|
15
|
Afsar RE, Kanbay M, Ibis A, Afsar B. In-depth review: is hepcidin a marker for the heart and the kidney? Mol Cell Biochem 2021; 476:3365-3381. [PMID: 33942218 DOI: 10.1007/s11010-021-04168-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2021] [Accepted: 04/21/2021] [Indexed: 12/15/2022]
Abstract
Iron is an essential trace element involved in oxidation-reduction reactions, oxygen transport and storage, and energy metabolism. Iron in excess can be toxic for cells, since iron produces reactive oxygen species and is important for survival of pathogenic microbes. There is a fine-tuning in the regulation of serum iron levels, determined by intestinal absorption, macrophage iron recycling, and mobilization of hepatocyte stores versus iron utilization, primarily by erythroid cells in the bone marrow. Hepcidin is the major regulatory hormone of systemic iron homeostasis and is upregulated during inflammation. Hepcidin metabolism is altered in chronic kidney disease. Ferroportin is an iron export protein and mediates iron release into the circulation from duodenal enterocytes, splenic reticuloendothelial macrophages, and hepatocytes. Systemic iron homeostasis is controlled by the hepcidin-ferroportin axis at the sites of iron entry into the circulation. Hepcidin binds to ferroportin, induces its internalization and intracellular degradation, and thus inhibits iron absorption from enterocytes, and iron release from macrophages and hepatocytes. Recent data suggest that hepcidin, by slowing or preventing the mobilization of iron from macrophages, may promote atherosclerosis and may be associated with increased cardiovascular disease risk. This article reviews the current data regarding the molecular and cellular pathways of systemic and autocrine hepcidin production and seeks the answer to the question whether changes in hepcidin translate into clinical outcomes of all-cause and cardiovascular mortality, and cardiovascular and renal end-points.
Collapse
Affiliation(s)
- Rengin Elsurer Afsar
- Department of Nephrology, Faculty of Medicine, Suleyman Demirel University, Isparta, Turkey.
| | - Mehmet Kanbay
- Department of Nephrology, Faculty of Medicine, Koc University, Istanbul, Turkey
| | - Avsin Ibis
- Department of Nephrology, Afyon Kocatepe Devlet Hastanesi, Afyon, Turkey
| | - Baris Afsar
- Department of Nephrology, Faculty of Medicine, Suleyman Demirel University, Isparta, Turkey
| |
Collapse
|
16
|
Kowara M, Cudnoch-Jedrzejewska A. Different Approaches in Therapy Aiming to Stabilize an Unstable Atherosclerotic Plaque. Int J Mol Sci 2021; 22:ijms22094354. [PMID: 33919446 PMCID: PMC8122261 DOI: 10.3390/ijms22094354] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2021] [Revised: 04/09/2021] [Accepted: 04/14/2021] [Indexed: 12/22/2022] Open
Abstract
Atherosclerotic plaque vulnerability is a vital clinical problem as vulnerable plaques tend to rupture, which results in atherosclerosis complications—myocardial infarctions and subsequent cardiovascular deaths. Therefore, methods aiming to stabilize such plaques are in great demand. In this brief review, the idea of atherosclerotic plaque stabilization and five main approaches—towards the regulation of metabolism, macrophages and cellular death, inflammation, reactive oxygen species, and extracellular matrix remodeling have been presented. Moreover, apart from classical approaches (targeted at the general mechanisms of plaque destabilization), there are also alternative approaches targeted either at certain plaques which have just become vulnerable or targeted at the minimization of the consequences of atherosclerotic plaque erosion or rupture. These alternative approaches have also been briefly mentioned in this review.
Collapse
|
17
|
Saadi S, Ghazali HM, Saari N, Abdulkarim SM. The structural reconformation of peptides in enhancing functional and therapeutic properties: Insights into their solid state crystallizations. Biophys Chem 2021; 273:106565. [PMID: 33780688 DOI: 10.1016/j.bpc.2021.106565] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2020] [Revised: 02/09/2021] [Accepted: 02/19/2021] [Indexed: 01/05/2023]
Abstract
Therapeutic peptides derived proteins with alpha-reconformation states like antibody shape have shown potential effects in combating terrible diseases linked with earlier signs of angiogensis, mutagenesis and transgenesis. Alpha reconformation in material design refers to the folding of the peptide chains and their transitions under reversible chemical bonds of disulfide chemical bridges and further non-covalence lesions. Thus, the rational design of signal peptides into alpha-helix is intended in increasing the defending effects of peptides into cores like adjuvant antibiotic and/or vaccines. Thereby, the signal peptides are able in displaying multiple eradicating regions by changing crystal-depositions and deviation angles. These types of molecular structures could have multiple advantages in tracing disease syndromes and impurities by increasing the host defense against the fates of pathogens and viruses, eventually leading to the loss in signaling by increasing peptide susceptibility levels to folding and unfolding and therefore, formation of transgenic peptide models. Alpha reconformation peptides is aimed in triggering as well as other regulatory functions such as remodulating metabolic chain disorders of lipolysis and glucolysis by increasing the insulin and leptin resistance for best lipid storages and lipoprotein density distributions.
Collapse
Affiliation(s)
- Sami Saadi
- Institut de la Nutrition, de l'Alimentation et des Technologies Agro-alimentaires INATAA 25017, Université Frères Mentouri, Constantine 1, Algeria; Department of Food Science, Faculty of Food Science and Technology, Universiti Putra Malaysia, 43400 Serdang, Selangor, Malaysia.
| | - Hasanah Mohd Ghazali
- Department of Food Science, Faculty of Food Science and Technology, Universiti Putra Malaysia, 43400 Serdang, Selangor, Malaysia
| | - Nazamid Saari
- Department of Food Science, Faculty of Food Science and Technology, Universiti Putra Malaysia, 43400 Serdang, Selangor, Malaysia
| | - Sabo Mohammed Abdulkarim
- Department of Food Science, Faculty of Food Science and Technology, Universiti Putra Malaysia, 43400 Serdang, Selangor, Malaysia
| |
Collapse
|
18
|
Assessment of Concentrations of Heavy Metals in Postmyocardial Infarction Patients and Patients Free from Cardiovascular Event. Cardiol Res Pract 2021; 2021:9546358. [PMID: 33604084 PMCID: PMC7868144 DOI: 10.1155/2021/9546358] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/19/2020] [Revised: 12/17/2020] [Accepted: 01/19/2021] [Indexed: 12/12/2022] Open
Abstract
Cardiovascular diseases (CVDs) constitute the first cause of death among the population of developing and developed countries. Atherosclerosis, which is a disorder with multifactorial etiopathogenesis, underlies most CVDs. The available literature includes ample research studies on the influence of classic cardiovascular (CV) risk factors. However, environmental exposure to heavy metals, among other substances, is still an unappreciated risk factor of CVDs. This study aimed to assess the concentration of some heavy metals (copper (Cu), zinc (Zn), manganese (Mn), cobalt (Co), and iron (Fe)) in the blood serum of postmyocardial infarction (post-MI) patients and patients free from myocardial infarction (MI) as well as estimate the relationship between the occurrence of MI and increased concentration of heavy metals. The concentration of heavy metals (Cu, Zn, Mn, Co, and Fe) was assessed using the inductively coupled plasma mass spectrometry technique in a group of 146 respondents divided into two groups: post-MI group (study group (SG), n = 74) and group without cardiovascular event (CVE) having a low CV risk (control group (CG), n = 72). The concentration of the analyzed heavy metals was higher in SG. All the heavy metals showed a significant diagnostic value (p < 0.001). The highest value of area under the curve (AUC) was observed for manganese (Mn) (0.955; 95% confidence interval (CI) = 0.922–0.988), while the lowest value was found for zinc (Zn) (0.691; 95% CI = 0.599–0.782). In one-dimensional models, high concentrations of each of the analyzed heavy metals significantly increased the chances of having MI from 7-fold (Cu) to 128-fold (Mn). All the models containing a particular metal showed a significant and high discrimination value for MI occurrence (AUC 0.72–0.92). Higher concentrations of Cu, Zn, Mn, Co, and Fe were found to considerably increase the chances of having MI. Considering the increasingly higher environmental exposure to heavy metals in recent times, their concentrations can be distinguished as a potential risk factor of CVDs.
Collapse
|
19
|
Wan Q, Yang M, Liu Z, Wu J. Ambient fine particulate matter aggravates atherosclerosis in apolipoprotein E knockout mice by iron overload via the hepcidin-ferroportin axis. Life Sci 2021; 264:118715. [PMID: 33160991 DOI: 10.1016/j.lfs.2020.118715] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Revised: 10/26/2020] [Accepted: 11/02/2020] [Indexed: 01/10/2023]
Abstract
AIMS Exposure to fine particulate matter (PM2.5) is correlated to atherosclerosis, but the mechanism remains largely undefined. Iron overload is a significant contributor to atherosclerosis, and iron homeostasis is highly regulated by the hepcidin-ferroportin (FPN) axis. Here we aimed to investigate the association between iron overload and PM2.5-induced atherosclerotic mice. MAIN METHODS Apolipoprotein E knockout (ApoE-/-) mice were randomly assigned to filtered air (FA group) or PM2.5 (PM2.5 group) for 3-month inhalation. Daily PM2.5 mass concentrations, serum levels of ferritin, iron, pro-atherosclerotic cytokines and lipid profiles, atherosclerotic lesion areas, hepcidin, FPN and iron depositions in atherosclerotic lesions, hepcidin, FPN mRNA and protein expressions in the aorta were detected, respectively. KEY FINDINGS The daily average concentration of atmospheric PM2.5 was 68.2 ± 21.8 μg/m3. Serum levels of ferritin, iron, VEGF, MCP-1, IL-6, TNF-α, TC and LDL-C, atherosclerotic lesion areas, hepcidin and iron depositions in atherosclerotic lesions, hepcidin mRNA and protein expressions in the PM2.5 group were observably higher than those in the FA group. Nevertheless, FPN deposition in atherosclerotic lesions, FPN mRNA and protein expressions in the aorta of the PM2.5 group were markedly lower than those of the FA group. SIGNIFICANCE PM2.5 inhalation could exacerbate the formation and development of atherosclerosis in ApoE-/- mice, the potential mechanisms may be partly associated with iron overload via the hepcidin-FPN axis, as well as iron-triggered systemic inflammation and hyperlipidemia.
Collapse
Affiliation(s)
- Qiang Wan
- Clinical Medical College, Jiangxi University of Traditional Chinese Medicine, Nanchang 330006, China; Jiangxi University of Traditional Chinese Medicine, Nanchang 330004, China.
| | - Ming Yang
- Jiangxi University of Traditional Chinese Medicine, Nanchang 330004, China
| | - Zhongyong Liu
- Clinical Medical College, Jiangxi University of Traditional Chinese Medicine, Nanchang 330006, China
| | - Jianguang Wu
- Clinical Medical College, Jiangxi University of Traditional Chinese Medicine, Nanchang 330006, China
| |
Collapse
|
20
|
Crea F. Dyslipidaemias and cardiovascular diseases: beyond cholesterol and atherosclerotic plaques. Eur Heart J 2020; 41:3865-3869. [PMID: 33175144 DOI: 10.1093/eurheartj/ehaa889] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Affiliation(s)
- Filippo Crea
- Department of Cardiovascular Medicine, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy.,Department of Cardiovascular and Pulmonary Sciences, Catholic University of the Sacred Heart, Rome, Italy
| |
Collapse
|
21
|
Demetz E, Tymoszuk P, Hilbe R, Volani C, Haschka D, Heim C, Auer K, Lener D, Zeiger LB, Pfeifhofer-Obermair C, Boehm A, Obermair GJ, Ablinger C, Coassin S, Lamina C, Kager J, Petzer V, Asshoff M, Schroll A, Nairz M, Dichtl S, Seifert M, von Raffay L, Fischer C, Barros-Pinkelnig M, Brigo N, Valente de Souza L, Sopper S, Hirsch J, Graber M, Gollmann-Tepeköylü C, Holfeld J, Halper J, Macheiner S, Gostner J, Vogel GF, Pechlaner R, Moser P, Imboden M, Marques-Vidal P, Probst-Hensch NM, Meiselbach H, Strauch K, Peters A, Paulweber B, Willeit J, Kiechl S, Kronenberg F, Theurl I, Tancevski I, Weiss G. The haemochromatosis gene Hfe and Kupffer cells control LDL cholesterol homeostasis and impact on atherosclerosis development. Eur Heart J 2020; 41:3949-3959. [PMID: 32227235 DOI: 10.1093/eurheartj/ehaa140] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/21/2019] [Revised: 10/16/2019] [Accepted: 02/18/2020] [Indexed: 12/12/2022] Open
Abstract
AIMS Imbalances of iron metabolism have been linked to the development of atherosclerosis. However, subjects with hereditary haemochromatosis have a lower prevalence of cardiovascular disease. The aim of our study was to understand the underlying mechanisms by combining data from genome-wide association study analyses in humans, CRISPR/Cas9 genome editing, and loss-of-function studies in mice. METHODS AND RESULTS Our analysis of the Global Lipids Genetics Consortium (GLGC) dataset revealed that single nucleotide polymorphisms (SNPs) in the haemochromatosis gene HFE associate with reduced low-density lipoprotein cholesterol (LDL-C) in human plasma. The LDL-C lowering effect could be phenocopied in dyslipidaemic ApoE-/- mice lacking Hfe, which translated into reduced atherosclerosis burden. Mechanistically, we identified HFE as a negative regulator of LDL receptor expression in hepatocytes. Moreover, we uncovered liver-resident Kupffer cells (KCs) as central players in cholesterol homeostasis as they were found to acquire and transfer LDL-derived cholesterol to hepatocytes in an Abca1-dependent fashion, which is controlled by iron availability. CONCLUSION Our results disentangle novel regulatory interactions between iron metabolism, KC biology and cholesterol homeostasis which are promising targets for treating dyslipidaemia but also provide a mechanistic explanation for reduced cardiovascular morbidity in subjects with haemochromatosis.
Collapse
Affiliation(s)
- Egon Demetz
- Department of Internal Medicine II, Medical University of Innsbruck, Anichstraße 35, 6020 Innsbruck, Austria
| | - Piotr Tymoszuk
- Department of Internal Medicine II, Medical University of Innsbruck, Anichstraße 35, 6020 Innsbruck, Austria
| | - Richard Hilbe
- Department of Internal Medicine II, Medical University of Innsbruck, Anichstraße 35, 6020 Innsbruck, Austria
| | - Chiara Volani
- Department of Internal Medicine II, Medical University of Innsbruck, Anichstraße 35, 6020 Innsbruck, Austria
| | - David Haschka
- Department of Internal Medicine II, Medical University of Innsbruck, Anichstraße 35, 6020 Innsbruck, Austria
| | - Christiane Heim
- Department of Internal Medicine II, Medical University of Innsbruck, Anichstraße 35, 6020 Innsbruck, Austria
| | - Kristina Auer
- Department of Internal Medicine II, Medical University of Innsbruck, Anichstraße 35, 6020 Innsbruck, Austria
| | - Daniela Lener
- Department of Internal Medicine III, Medical University of Innsbruck, Anichstraße 35, 6020 Innsbruck, Austria
| | - Lucas B Zeiger
- Department of Internal Medicine II, Medical University of Innsbruck, Anichstraße 35, 6020 Innsbruck, Austria
| | - Christa Pfeifhofer-Obermair
- Department of Internal Medicine II, Medical University of Innsbruck, Anichstraße 35, 6020 Innsbruck, Austria
| | - Anna Boehm
- Department of Internal Medicine II, Medical University of Innsbruck, Anichstraße 35, 6020 Innsbruck, Austria
| | - Gerald J Obermair
- Department of Physiology and Medical Physics, Medical University of Innsbruck, Fritz-Pregl-Straße 3, 6020 Innsbruck, Austria
- Division of Physiology, Karl Landsteiner University of Health Sciences, Dr.-Karl-Dorrek-Straße 30, 3500 Krems, Austria
| | - Cornelia Ablinger
- Department of Physiology and Medical Physics, Medical University of Innsbruck, Fritz-Pregl-Straße 3, 6020 Innsbruck, Austria
| | - Stefan Coassin
- Department of Genetics and Pharmacology, Institute of Genetic Epidemiology, Medical University of Innsbruck, Schöpfstraße 41, 6020 Innsbruck, Austria
| | - Claudia Lamina
- Department of Genetics and Pharmacology, Institute of Genetic Epidemiology, Medical University of Innsbruck, Schöpfstraße 41, 6020 Innsbruck, Austria
| | - Juliane Kager
- Department of Internal Medicine II, Medical University of Innsbruck, Anichstraße 35, 6020 Innsbruck, Austria
| | - Verena Petzer
- Department of Internal Medicine II, Medical University of Innsbruck, Anichstraße 35, 6020 Innsbruck, Austria
| | - Malte Asshoff
- Department of Internal Medicine II, Medical University of Innsbruck, Anichstraße 35, 6020 Innsbruck, Austria
| | - Andrea Schroll
- Department of Internal Medicine II, Medical University of Innsbruck, Anichstraße 35, 6020 Innsbruck, Austria
| | - Manfred Nairz
- Department of Internal Medicine II, Medical University of Innsbruck, Anichstraße 35, 6020 Innsbruck, Austria
| | - Stefanie Dichtl
- Department of Internal Medicine II, Medical University of Innsbruck, Anichstraße 35, 6020 Innsbruck, Austria
| | - Markus Seifert
- Department of Internal Medicine II, Medical University of Innsbruck, Anichstraße 35, 6020 Innsbruck, Austria
- Christian Doppler Laboratory for Iron Metabolism and Anemia Research, Medical University of Innsbruck, Innsbruck, Austria
| | - Laura von Raffay
- Department of Internal Medicine II, Medical University of Innsbruck, Anichstraße 35, 6020 Innsbruck, Austria
| | - Christine Fischer
- Department of Internal Medicine II, Medical University of Innsbruck, Anichstraße 35, 6020 Innsbruck, Austria
| | - Marina Barros-Pinkelnig
- Department of Internal Medicine II, Medical University of Innsbruck, Anichstraße 35, 6020 Innsbruck, Austria
| | - Natascha Brigo
- Department of Internal Medicine II, Medical University of Innsbruck, Anichstraße 35, 6020 Innsbruck, Austria
| | - Lara Valente de Souza
- Department of Internal Medicine II, Medical University of Innsbruck, Anichstraße 35, 6020 Innsbruck, Austria
- Christian Doppler Laboratory for Iron Metabolism and Anemia Research, Medical University of Innsbruck, Innsbruck, Austria
| | - Sieghart Sopper
- Department of Internal Medicine V, Medical University of Innsbruck, Anichstraße 35, 6020 Innsbruck, Austria
| | - Jakob Hirsch
- Department of Cardiac Surgery, Medical University of Innsbruck, Anichstraße 35, 6020 Innsbruck, Austria
| | - Michael Graber
- Department of Cardiac Surgery, Medical University of Innsbruck, Anichstraße 35, 6020 Innsbruck, Austria
| | - Can Gollmann-Tepeköylü
- Department of Cardiac Surgery, Medical University of Innsbruck, Anichstraße 35, 6020 Innsbruck, Austria
| | - Johannes Holfeld
- Department of Cardiac Surgery, Medical University of Innsbruck, Anichstraße 35, 6020 Innsbruck, Austria
| | - Julia Halper
- Department of Internal Medicine II, Medical University of Innsbruck, Anichstraße 35, 6020 Innsbruck, Austria
| | - Sophie Macheiner
- Department of Internal Medicine I, Medical University of Innsbruck, Anichstraße 35, 6020 Innsbruck, Austria
| | - Johanna Gostner
- Division of Medical Biochemistry, Medical University of Innsbruck, Innrain 80/IV, 6020 Innsbruck, Austria
| | - Georg F Vogel
- Department of Pediatrics I, Medical University of Innsbruck, Anichstraße 35, 6020 Innsbruck, Austria
| | - Raimund Pechlaner
- Department of Neurology, Medical University of Innsbruck, Anichstraße 35, 6020 Innsbruck, Austria
| | - Patrizia Moser
- Department of Pathology, Innsbruck University Hospital, Anichstraße 35, 6020 Innsbruck, Austria
| | - Medea Imboden
- Swiss Tropical and Public Health Institute, Socinstraße 57, 4051 Basel, Switzerland
- Department of Public Health, University of Basel, Bernoullistraße 28, 4056 Basel, Switzerland
| | - Pedro Marques-Vidal
- Department of Internal Medicine, Lausanne University Hospital, Rue du Bugnon 46, 1011 Lausanne, Switzerland
| | - Nicole M Probst-Hensch
- Swiss Tropical and Public Health Institute, Socinstraße 57, 4051 Basel, Switzerland
- Department of Public Health, University of Basel, Bernoullistraße 28, 4056 Basel, Switzerland
| | - Heike Meiselbach
- Department of Nephrology and Hypertension, University Hospital Erlangen, Maximiliansplatz 2, 91054 Erlangen, Germany
| | - Konstantin Strauch
- Institute of Genetic Epidemiology, Helmholtz Zentrum München-German Research Center for Environmental Health, Ingolstädter Landstraße 1, 85764 Neuherberg, Germany
- Institute of Medical Informatics, Biometry and Epidemiology, Ludwig-Maximilians-Universität, Marchioninistraße 15, 81377 Munich, Germany
| | - Annette Peters
- Institute of Epidemiology II, Helmholtz Zentrum München-German Research Center for Environmental Health, Ingolstädter Landstraße 1, 85764 Neuherberg, Germany
- German Center for Diabetes Research, Ingolstädter Landstraße 1, 85764 Neuherberg, Germany
- German Center for Cardiovascular Research, Lazarettstraße 36, 80636 Munich, Germany
| | - Bernhard Paulweber
- First Department of Medicine, Paracelsus Medical University Salzburg, Strubergasse 21, 5020 Salzburg, Austria
| | - Johann Willeit
- Department of Neurology, Medical University of Innsbruck, Anichstraße 35, 6020 Innsbruck, Austria
| | - Stefan Kiechl
- Department of Neurology, Medical University of Innsbruck, Anichstraße 35, 6020 Innsbruck, Austria
| | - Florian Kronenberg
- Department of Genetics and Pharmacology, Institute of Genetic Epidemiology, Medical University of Innsbruck, Schöpfstraße 41, 6020 Innsbruck, Austria
| | - Igor Theurl
- Department of Internal Medicine II, Medical University of Innsbruck, Anichstraße 35, 6020 Innsbruck, Austria
| | - Ivan Tancevski
- Department of Internal Medicine II, Medical University of Innsbruck, Anichstraße 35, 6020 Innsbruck, Austria
| | - Guenter Weiss
- Department of Internal Medicine II, Medical University of Innsbruck, Anichstraße 35, 6020 Innsbruck, Austria
- Christian Doppler Laboratory for Iron Metabolism and Anemia Research, Medical University of Innsbruck, Innsbruck, Austria
| |
Collapse
|
22
|
Hepcidin and ferritin levels in restless legs syndrome: a case-control study. Sci Rep 2020; 10:11914. [PMID: 32681031 PMCID: PMC7367854 DOI: 10.1038/s41598-020-68851-0] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2020] [Accepted: 06/24/2020] [Indexed: 01/11/2023] Open
Abstract
The association between restless legs syndrome (RLS) and iron homeostasis remains unclear. We compared serum hepcidin and ferritin levels in patients with RLS and controls, and assessed their relationships with RLS phenotype, drug intake, and history of augmentation syndrome. 102 drug-free RLS patients (age 58.9 [24.5–77.2], 63 females) and 73 controls (age 56.8 [23.46–76.6], 45 females) underwent a polysomnography recording. Hepcidin levels were quantified by ELISA. 34 RLS patients had a second assessment after starting dopaminergic drugs. Ferritin level was low (< 50 µg/l) in 14.7% of patients and 25% of controls, with no between-group differences in the mean values. Hepcidin levels were higher in patients even after adjustment for confounding factors, and excluding participants with low ferritin levels. Ferritin and hepcidin levels were comparable before and after treatment, and between patients with (n = 17) and without history of augmentation. Ferritin and hepcidin levels correlated with age, body mass index, and periodic leg movements. Higher hepcidin levels were associated with older age, older age at RLS onset, less daytime sleepiness and familial RLS. In conclusion, serum hepcidin levels but not ferritin were higher in RLS patients regardless of treatment and history of augmentation. Serum hepcidin may be a more relevant biomarker of RLS than ferritin.
Collapse
|
23
|
Weidmann H, Bannasch JH, Waldeyer C, Shrivastava A, Appelbaum S, Ojeda-Echevarria FM, Schnabel R, Lackner KJ, Blankenberg S, Zeller T, Karakas M. Iron Metabolism Contributes to Prognosis in Coronary Artery Disease: Prognostic Value of the Soluble Transferrin Receptor Within the AtheroGene Study. J Am Heart Assoc 2020; 9:e015480. [PMID: 32321351 PMCID: PMC7428563 DOI: 10.1161/jaha.119.015480] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Background Coronary heart disease is a leading cause of mortality worldwide. Iron deficiency, a frequent comorbidity of coronary heart disease, causes an increased expression of transferrin receptor and soluble transferrin receptor levels (sTfR) levels, while iron repletion returns sTfR levels to the normal physiological range. Recently, sTfR levels were proposed as a potential new marker of iron metabolism in cardiovascular diseases. Therefore, we aimed to evaluate the prognostic value of circulating sTfR levels in a large cohort of patients with coronary heart disease. Methods and Results The disease cohort comprised 3423 subjects who had angiographically documented coronary heart disease and who participated in the AtheroGene study. Serum levels of sTfR were determined at baseline using an automated immunoassay (Roche Cobas Integra 400). Two main outcomes were considered: a combined end point of myocardial infarction and cardiovascular death and cardiovascular death alone. During a median follow‐up of 4.0 years, 10.3% of the patients experienced an end point. In Cox regression analyses for sTfR levels, the hazard ratio (HR) for future cardiovascular death and/or myocardial infarction was 1.27 (95% CI, 1.11–1.44, P<0.001) after adjustment for sex and age. This association remained significant (HR, 1.23; 95% CI, 1.03–1.46, P=0.02) after additional adjustment for body mass index, smoking status, hypertension, diabetes mellitus, dyslipidemia, C‐reactive protein, and surrogates of cardiac function, size of myocardial necrosis (hs‐Tnl), and hemoglobin levels. Conclusions In this large cohort study, sTfR levels were strongly associated with future myocardial infarction and cardiovascular death. This implicates a role for sTfR in secondary cardiovascular risk prediction.
Collapse
Affiliation(s)
- Henri Weidmann
- Department of General and Interventional Cardiology University Heart Center Hamburg Hamburg Germany.,German Center for Cardiovascular Research (DZHK), Partner Site Hamburg, Lübeck, Kiel Hamburg Germany
| | - Johannes H Bannasch
- Department of General and Interventional Cardiology University Heart Center Hamburg Hamburg Germany
| | - Christoph Waldeyer
- Department of General and Interventional Cardiology University Heart Center Hamburg Hamburg Germany.,German Center for Cardiovascular Research (DZHK), Partner Site Hamburg, Lübeck, Kiel Hamburg Germany
| | - Apurva Shrivastava
- Department of General and Interventional Cardiology University Heart Center Hamburg Hamburg Germany.,German Center for Cardiovascular Research (DZHK), Partner Site Hamburg, Lübeck, Kiel Hamburg Germany
| | - Sebastian Appelbaum
- Department of General and Interventional Cardiology University Heart Center Hamburg Hamburg Germany
| | | | - Renate Schnabel
- Department of General and Interventional Cardiology University Heart Center Hamburg Hamburg Germany.,German Center for Cardiovascular Research (DZHK), Partner Site Hamburg, Lübeck, Kiel Hamburg Germany
| | - Karl J Lackner
- Department of Laboratory Medicine University Medical Center Johannes Gutenberg University Mainz Mainz Germany.,German Center for Cardiovascular Research (DZHK), Partner Site Rhein-Main Mainz Germany
| | - Stefan Blankenberg
- Department of General and Interventional Cardiology University Heart Center Hamburg Hamburg Germany.,German Center for Cardiovascular Research (DZHK), Partner Site Hamburg, Lübeck, Kiel Hamburg Germany
| | - Tanja Zeller
- Department of General and Interventional Cardiology University Heart Center Hamburg Hamburg Germany.,German Center for Cardiovascular Research (DZHK), Partner Site Hamburg, Lübeck, Kiel Hamburg Germany
| | - Mahir Karakas
- Department of General and Interventional Cardiology University Heart Center Hamburg Hamburg Germany.,German Center for Cardiovascular Research (DZHK), Partner Site Hamburg, Lübeck, Kiel Hamburg Germany
| |
Collapse
|
24
|
Cui J, Guo X, Li Q, Song N, Xie J. Hepcidin-to-Ferritin Ratio Is Decreased in Astrocytes With Extracellular Alpha-Synuclein and Iron Exposure. Front Cell Neurosci 2020; 14:47. [PMID: 32210768 PMCID: PMC7075942 DOI: 10.3389/fncel.2020.00047] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2019] [Accepted: 02/20/2020] [Indexed: 12/21/2022] Open
Abstract
Astrocytes are the most abundant glial cells in the central nervous system (CNS). As indispensable elements of the neurovascular unit, they are involved in the inflammatory response and disease-associated processes. Alpha-synuclein (α-syn) is released into the extracellular space by neurons and can be internalized by adjacent astrocytes, which activates glial cells to induce neuroinflammation. We were interested in whether astrocyte-mediated neuroinflammation is modulated by intracellular iron status and extracellular α-syn. Our results showed that recombinant α-syn (1 μg/ml and 5 μg/ml) treatment for 24 h did not affect the expression of the iron transporters divalent metal transporter 1 (DMT1) and ferroportin 1 (FPN1), nor those of iron regulatory protein (IRP) 1 or IRP2. Several proinflammatory cytokines, including tumor necrosis factor-α (TNF-α), interleukin (IL)-1β, and IL-6 exhibited up-regulated mRNA levels in 5 μg/ml α-syn-treated astrocytes. TNF-α release was increased, indicating that inflammatory responses were triggered in these cells. Pretreatment with the iron-overload reagent ferric ammonium citrate (FAC, 100 μmol/L) for 24 h had no effects on mRNA levels and release of proinflammatory cytokines. Inflammatory responses triggered by α-syn were not affected by iron overload. The iron chelator desferrioxamine (DFO, 100 μmol/L) exerted suppressive effects on TNF-α mRNA levels, although no change was observed for TNF-α release. Hepcidin mRNA levels were down-regulated significantly in astrocytes co-treated with FAC and α-syn, although independent treatment with either FAC or α-syn did not alter hepcidin levels. In contrast, hepcidin mRNA levels were up-regulated in DFO and α-syn co-treated cells. As expected, ferritin protein levels were up-regulated or down-regulated with FAC or DFO treatment, respectively. Following the up-regulation of ferritin mediated by α-syn, hepcidin-to-ferritin levels were indicative of modulatory effects in α-syn-treated astrocytes with altered iron status. Therefore, we propose that the hepcidin-to-ferritin ratio is indicative of a detrimental response in primary cultured astrocytes experiencing iron and extracellular α-syn.
Collapse
Affiliation(s)
- Juntao Cui
- Institute of Brain Science and Disease, Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders, Shandong Provincial Collaborative Innovation Center for Neurodegenerative Disorders, Qingdao University, Qingdao, China
| | - Xinli Guo
- Institute of Brain Science and Disease, Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders, Shandong Provincial Collaborative Innovation Center for Neurodegenerative Disorders, Qingdao University, Qingdao, China
| | - Qijun Li
- Institute of Brain Science and Disease, Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders, Shandong Provincial Collaborative Innovation Center for Neurodegenerative Disorders, Qingdao University, Qingdao, China
| | - Ning Song
- Institute of Brain Science and Disease, Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders, Shandong Provincial Collaborative Innovation Center for Neurodegenerative Disorders, Qingdao University, Qingdao, China
| | - Junxia Xie
- Institute of Brain Science and Disease, Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders, Shandong Provincial Collaborative Innovation Center for Neurodegenerative Disorders, Qingdao University, Qingdao, China
| |
Collapse
|
25
|
Preoperative biomarker evaluation for the prediction of cardiovascular events after major vascular surgery. J Vasc Surg 2019; 70:1564-1575. [PMID: 31653377 DOI: 10.1016/j.jvs.2019.02.041] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2018] [Accepted: 02/12/2019] [Indexed: 02/05/2023]
Abstract
OBJECTIVE The cause of perioperative myocardial infarction (PMI) is postulated to involve hemodynamic stress or coronary plaque destabilization. We aimed to evaluate perioperative factors in patients with peripheral artery disease (PAD) undergoing major vascular surgery to determine the likely mechanisms and predictors of PMI. METHODS This was a prospective cohort study of 133 patients undergoing major vascular surgery including open abdominal aortic aneurysm (AAA) repair (n = 40) and major suprainguinal or infrainguinal arterial bypasses (non-AAA; n = 93). Preoperative assessment with history, physical examination, and peripheral artery tonometry was performed in addition to plasma sampling of biomarkers associated with inflammation and coronary plaque instability. The primary outcome was occurrence of a 30-day cardiovascular event (CVE; composite of PMI [troponin I elevation >99th percentile reference of ≥0.1 μg/L], stroke, or death). RESULTS Of 133 patients, 36 patients (27%) developed a 30-day CVE after vascular surgery, and all were PMI. Patients with 30-day CVE were older (75 ± 8 years vs 69 ± 10 years, mean ± standard deviation; P = .001), had higher prevalence of hypertension (94% vs 79%; P = .01) and preoperative beta-blocker therapy (50% vs 29%; P = .02), and had longer duration of surgery (5.1 ± 1.8 hours vs 4.0 ± 1.1 hours; P < .0001). Significant elevations in cystatin C, N-terminal pro-B-type natriuretic peptide (NT-proBNP), troponin I, high-sensitivity troponin T, matrix metalloproteinase 3, and osteoprotegerin occurred in those who developed 30-day CVE (all P < .05). Multivariate binary logistic regression identified AAA surgery and log-transformed NT-proBNP to be independent preoperative predictors of 30-day CVE (area under the receiver operating characteristic curve = 0.81). CONCLUSIONS In patients with peripheral artery disease undergoing major vascular surgery, the likely mechanism of PMI appears to be the hemodynamic stress related to the type and duration of surgery. NT-proBNP was a useful independent predictor of CVE and thus may serve as an important biomarker of cardiovascular fitness for surgery.
Collapse
|
26
|
Xiao L, Luo G, Guo X, Jiang C, Zeng H, Zhou F, Li Y, Yu J, Yao P. Macrophage iron retention aggravates atherosclerosis: Evidence for the role of autocrine formation of hepcidin in plaque macrophages. Biochim Biophys Acta Mol Cell Biol Lipids 2019; 1865:158531. [PMID: 31666189 DOI: 10.1016/j.bbalip.2019.158531] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2019] [Revised: 09/10/2019] [Accepted: 09/13/2019] [Indexed: 12/18/2022]
Abstract
Iron accumulation has been frequently found in atherosclerotic lesions, especially in macrophages/foam cells, but the exact mechanisms by which hepcidin induces iron retention in plaque macrophages and its roles in atherogenesis remain unknown. Double immunofluorescence staining showed colocalization of hepcidin-positive macrophages with ox-LDL, TLR4, p-p65 and ferritin light chain (ferritin-L) both in human and murine atherosclerotic lesions. RAW264.7 macrophages incubated with ox-LDL showed elevated expression of TLR4, p-p65, hepcidin, ferritin-L/H, CYP27A1, CD36, PPARγ, liver X receptor α (LXRα), and ATP binding cassette transporter A1/G1 (ABCA1/G1), as well as increased intracellular labile iron pool level and lipid accumulation. Ox-LDL-induced iron retention and lipid accumulation were aggravated by lipopolysaccharide but blocked by TAK-242, an antagonist of TLR4. Moreover, macrophage TLR4/NF-κB pathway activation and foaming triggered by ox-LDL was enhanced by ferric ammonium citrate or exogenous hepcidin but attenuated by hepcidin silencing or the use of iron chelator. Meanwhile, the addition of hepcidin stimulated CD36-mediated Dil-labeled-ox-LDL uptake and inhibited the LXRα-ABCA1/G1 pathway-dependent cholesterol efflux in macrophages, which was significantly reversed by 27-hydroxycholesterol but further exacerbated by cyclosporin A, a selective inhibitor of CYP27A1. Our study provided the evidence that iron trapped in atherosclerosis plaque macrophages contributes to cholesterol disequilibrium-initiated foam cell formation, which is provoked by the unique but largely unknown autocrine formation of hepcidin in plaque macrophages via activating the TLR4/NF-κB pathway when exposed to ox-LDL. Such findings, considering the intricate vicious cycle between macrophage hepcidin autocrine-triggered iron retention and cholesterol disequilibrium, may shed new light on the "iron hypothesis" of atherosclerosis.
Collapse
Affiliation(s)
- Lin Xiao
- Department of Nutrition and Food Hygiene, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; Hubei Key Laboratory of Food Nutrition and Safety, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; Ministry of Education Key Laboratory of Environment, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Gang Luo
- Department of Nutrition and Food Hygiene, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; Hubei Key Laboratory of Food Nutrition and Safety, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; Ministry of Education Key Laboratory of Environment, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Xiaoping Guo
- Department of Nutrition and Food Hygiene, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; Hubei Key Laboratory of Food Nutrition and Safety, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; Ministry of Education Key Laboratory of Environment, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Chunjie Jiang
- Department of Nutrition and Food Hygiene, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; Hubei Key Laboratory of Food Nutrition and Safety, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; Ministry of Education Key Laboratory of Environment, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Hongmei Zeng
- Department of Nutrition and Food Hygiene, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; Hubei Key Laboratory of Food Nutrition and Safety, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; Ministry of Education Key Laboratory of Environment, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Feng Zhou
- Department of Nutrition and Food Hygiene, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; Hubei Key Laboratory of Food Nutrition and Safety, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; Ministry of Education Key Laboratory of Environment, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Yanyan Li
- Shenzhen Center for Chronic Disease Control, 2021 Buxin Road, Shenzhen 518020, China
| | - Jiasheng Yu
- Department of Neurosurgery, Tongji Hospital, Tongji Medical College, HuaZhong University of Science and Technology, Wuhan 430030, China
| | - Ping Yao
- Department of Nutrition and Food Hygiene, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; Hubei Key Laboratory of Food Nutrition and Safety, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; Ministry of Education Key Laboratory of Environment, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China.
| |
Collapse
|
27
|
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
|
28
|
Hu X, Cai X, Ma R, Fu W, Zhang C, Du X. Iron‐load exacerbates the severity of atherosclerosis via inducing inflammation and enhancing the glycolysis in macrophages. J Cell Physiol 2019; 234:18792-18800. [PMID: 30927265 DOI: 10.1002/jcp.28518] [Citation(s) in RCA: 63] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2018] [Revised: 02/22/2019] [Accepted: 03/05/2019] [Indexed: 12/12/2022]
Affiliation(s)
- Xiaorong Hu
- Department of Cardiology Zhongnan Hospital of Wuhan University Wuhan Hubei People's Republic of China
| | - Xinyong Cai
- Department of Cardiology Jiangxi Provincial People's Hospital Affiliated to Nanchang University Nanchang Jiangxi People's Republic of China
| | - Ruisong Ma
- Department of Cardiology Zhongnan Hospital of Wuhan University Wuhan Hubei People's Republic of China
| | - Wenwen Fu
- Department of Cardiology Renmin Hospital of Wuhan University, Cardiovascular Research Institute of Wuhan University Wuhan Hubei People's Republic of China
| | - Changjiang Zhang
- Department of Cardiology Renmin Hospital of Wuhan University, Cardiovascular Research Institute of Wuhan University Wuhan Hubei People's Republic of China
| | - Xianjin Du
- Department of Emergency Renmin Hospital of Wuhan University Wuhan Hubei People's Republic of China
| |
Collapse
|
29
|
Zhu Y, He B, Xiao Y, Chen Y. Iron metabolism and its association with dyslipidemia risk in children and adolescents: a cross-sectional study. Lipids Health Dis 2019; 18:50. [PMID: 30755213 PMCID: PMC6371579 DOI: 10.1186/s12944-019-0985-8] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2018] [Accepted: 01/23/2019] [Indexed: 11/24/2022] Open
Abstract
Background Information on the association between iron metabolism and dyslipidaemia in children is limited. Thus, this study aims to evaluate the iron metabolic status of children with different body mass index (BMI) and to examine the association between iron metabolism and dyslipidaemia risk. Method In total, 1866 children and adolescents aged 7–18 were enrolled in this study, including 912 boys and 954 girls. In this cross-sectional study, parameters for anthropometry, lipids and iron metabolism including transferrin, soluble transferrin receptor (sTfR), ferritin and serum iron (SF) were evaluated. Data regarding demographic characteristics, diet, and physical activity were collected by self-reported questionnaires. Results The prevalence of dyslipidaemia and iron deficiency in children and adolescents increased based on BMI categories (both P < 0.05) and were 58.3 and 8.9% in subjects with obesity, respectively. The lowest SF and the highest ferritin levels were observed in subjects who were obese (both P < 0.001). Subjects with dyslipidaemia had lower SF, transferrin and sTfR levels by different BMI categories, and those who were obese had higher ferritin levels (all P < 0.05). Most importantly, higher concentrations of transferrin and sTfR were related to lower dyslipidaemia risk (OR for transferrin: 0.49, 95% CI: 0.33–0.71; OR for sTfR: 0.68, 95% CI: 0.46–0.99). Conclusions A downward trend in SF level by BMI categories and the highest ferritin level in subjects with obesity suggested that iron storage was associated with BMI in children and adolescents. Moreover, an inverse relationship was observed between transferrin and sTfR concentrations and dyslipidaemia risk in children with different BMI.
Collapse
Affiliation(s)
- Yanna Zhu
- Department of Maternal and Child Health, School of Public Health, and Global Health Institute (SGHI), Sun Yat-sen University, No.74 Zhongshan Road II, Guangzhou, 510080, Guangdong Province, China
| | - Baoting He
- Department of Maternal and Child Health, School of Public Health, and Global Health Institute (SGHI), Sun Yat-sen University, No.74 Zhongshan Road II, Guangzhou, 510080, Guangdong Province, China
| | - Yunjun Xiao
- Department of Nutrition and Food Hygiene, Shenzhen Center for Disease Control and Prevention, Shenzhen, China
| | - Yajun Chen
- Department of Maternal and Child Health, School of Public Health, and Global Health Institute (SGHI), Sun Yat-sen University, No.74 Zhongshan Road II, Guangzhou, 510080, Guangdong Province, China.
| |
Collapse
|
30
|
Wang X, Sheng L, Ye P, Cao R, Yang X, Xiao W, Zhang Y, Bai Y, Wu H. The association between Hepcidin and arterial stiffness in a community-dwelling population. Lipids Health Dis 2018; 17:244. [PMID: 30373612 PMCID: PMC6206657 DOI: 10.1186/s12944-018-0866-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2018] [Accepted: 09/11/2018] [Indexed: 11/10/2022] Open
Abstract
Background An association of hepcidin with cardiovascular (CV) disease and atherosclerosis has been reported in different patient groups. However, it has not been well described clinically the association between hepcidin and arterial stiffness. In this study,We analysed the possible mechanism of Hepcidin and arterial stiffness. Methods This article related measurements of plasma hepcidin and arterial stiffness (carotid–femoral pulse wave velocity [PWV]) in a community-based sample. Results After a median follow-up interval of 4.8 years, multiple linear regression analysis revealed that hepcidin was independently associated with carotid–femoral PWV (β = 1.498, P < 0.001). In a multivariable linear regression analysis, HDL3-C levels were negatively and independently associated with hepcidin at baseline (β = − 0.857, P = 0.024). HDL2-C was not associated with hepcidin at baseline (β = − 1.121, P = 0.133). Conclusions We found an association between baseline hepcidin and follow-up arterial stiffness that was independent of age, gender and other vascular risk factors. We also identified an association between hepcidin and HDL3-C at baseline, which indicates that the HDL3-C level may reflect the change in cholesterol efflux from peripheral arteries and partly explain the relationship between hepcidin and the change of arterial stiffness.
Collapse
Affiliation(s)
- Xiaona Wang
- Department of Geriatric Cardiology, Chinese PLA General Hospital, Fuxing Road #28, Beijing, 100853, China
| | - Li Sheng
- Department of Geriatric Cardiology, Chinese PLA General Hospital, Fuxing Road #28, Beijing, 100853, China
| | - Ping Ye
- Department of Geriatric Cardiology, Chinese PLA General Hospital, Fuxing Road #28, Beijing, 100853, China.
| | - Ruihua Cao
- Department of Geriatric Cardiology, Chinese PLA General Hospital, Fuxing Road #28, Beijing, 100853, China
| | - Xu Yang
- Department of Geriatric Cardiology, Chinese PLA General Hospital, Fuxing Road #28, Beijing, 100853, China
| | - Wenkai Xiao
- Department of Geriatric Cardiology, Chinese PLA General Hospital, Fuxing Road #28, Beijing, 100853, China
| | - Yun Zhang
- Department of Geriatric Cardiology, Chinese PLA General Hospital, Fuxing Road #28, Beijing, 100853, China
| | - Yongyi Bai
- Department of Geriatric Cardiology, Chinese PLA General Hospital, Fuxing Road #28, Beijing, 100853, China
| | - Hongmei Wu
- Department of Geriatric Cardiology, Chinese PLA General Hospital, Fuxing Road #28, Beijing, 100853, China
| |
Collapse
|
31
|
Kong WN, Cui Y, Fu YJ, Lei Y, Ci Y, Bao Y, Zhao S, Xie L, Chang YZ, Zhao SE. The α1-adrenergic receptor is involved in hepcidin upregulation induced by adrenaline and norepinephrine via the STAT3 pathway. J Cell Biochem 2018; 119:5517-5527. [PMID: 29377263 DOI: 10.1002/jcb.26715] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2017] [Accepted: 01/24/2018] [Indexed: 02/06/2023]
Abstract
Elevated body iron stores are associated with hypertension progression, while hypertension is associated with elevated plasma catecholamine levels in patients. However, there is a gap in our understanding of the connection between catecholamines and iron regulation. Hepcidin is a key iron-regulatory hormone, which maintains body iron balance. In the present study, we investigated the effects of adrenaline (AD) and norepinephrine (NE) on hepatic hepcidin regulation. Mice were treated with AD, NE, phenylephrine (PE, α1-adrenergic receptor agonist), prazosin (PZ, α1-adrenergic receptor antagonist), and/or propranolol (Pro, β-adrenergic receptor antagonist). The levels of hepcidin, as well as signal transducer and activator of transcription 3 (STAT3), ferroportin 1 (FPN1), and ferritin-light (Ft-L) protein in the liver or spleen, were assessed. Six hours after AD, NE, or PE treatment, hepatic hepcidin mRNA levels increased. Pretreatment with PZ, but not Pro, abolished the effects of AD or NE on STAT3 phosphorylation and hepatic hepcidin expression. When mice were treated with AD or NE continuously for 7 days, an increase in hepatic hepcidin mRNA levels and serum hepcidin concentration was also observed. Meanwhile, the expected downstream effects of elevated hepcidin, namely decreased FPN1 expression and increased Ft-L protein and non-heme iron concentrations in the spleen, were observed after the continuous AD or NE treatments. Taken together, we found that AD or NE increase hepatic hepcidin expression via the α1-adrenergic receptor and STAT3 pathways in mice. The elevated hepatic hepcidin decreased FPN1 levels in the spleen, likely causing the increased iron accumulation in the spleen.
Collapse
Affiliation(s)
- Wei-Na Kong
- Laboratory of Molecular Iron Metabolism, The Key Laboratory of Animal Physiology, Biochemistry and Molecular Biology of Hebei Province, College of Life Science, Hebei Normal University, Shijiazhuang, Hebei Province, P. R. China.,Bioreactor and Protein Drug Research and Development Center of Hebei Universities, Hebei Chemical and Pharmaceutical College, Shijiazhuang, Hebei Province, P. R. China
| | - Yanmei Cui
- Laboratory of Molecular Iron Metabolism, The Key Laboratory of Animal Physiology, Biochemistry and Molecular Biology of Hebei Province, College of Life Science, Hebei Normal University, Shijiazhuang, Hebei Province, P. R. China
| | - Yu-Jian Fu
- Laboratory of Molecular Iron Metabolism, The Key Laboratory of Animal Physiology, Biochemistry and Molecular Biology of Hebei Province, College of Life Science, Hebei Normal University, Shijiazhuang, Hebei Province, P. R. China
| | - Yuhua Lei
- Laboratory of Molecular Iron Metabolism, The Key Laboratory of Animal Physiology, Biochemistry and Molecular Biology of Hebei Province, College of Life Science, Hebei Normal University, Shijiazhuang, Hebei Province, P. R. China
| | - Yunzhe Ci
- Laboratory of Molecular Iron Metabolism, The Key Laboratory of Animal Physiology, Biochemistry and Molecular Biology of Hebei Province, College of Life Science, Hebei Normal University, Shijiazhuang, Hebei Province, P. R. China.,Chengde Medical University, Chengde, Hebei Province, P. R. China
| | - Yongping Bao
- Norwich Medical School, University of East Anglia, Norwich, UK
| | - Shuqiang Zhao
- Laboratory of Molecular Iron Metabolism, The Key Laboratory of Animal Physiology, Biochemistry and Molecular Biology of Hebei Province, College of Life Science, Hebei Normal University, Shijiazhuang, Hebei Province, P. R. China
| | - Lide Xie
- Chengde Medical University, Chengde, Hebei Province, P. R. China
| | - Yan-Zhong Chang
- Laboratory of Molecular Iron Metabolism, The Key Laboratory of Animal Physiology, Biochemistry and Molecular Biology of Hebei Province, College of Life Science, Hebei Normal University, Shijiazhuang, Hebei Province, P. R. China
| | - Shu-E Zhao
- Laboratory of Molecular Iron Metabolism, The Key Laboratory of Animal Physiology, Biochemistry and Molecular Biology of Hebei Province, College of Life Science, Hebei Normal University, Shijiazhuang, Hebei Province, P. R. China.,The 3rd Hospital of Hebei Medical University, Shijiazhuang, Hebei Province, P. R. China
| |
Collapse
|
32
|
Relation between high serum hepcidin-25 level and subclinical atherosclerosis and cardiovascular mortality in hemodialysis patients. Anatol J Cardiol 2018; 19:117-122. [PMID: 29339674 PMCID: PMC5864805 DOI: 10.14744/anatoljcardiol.2017.8019] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
OBJECTIVE In hemodialysis (HD) patients, cardiovascular disease (CVD) is the major cause of mortality and morbidity. In atherosclerotic diseases, iron gets accumulated in the arterial wall. Hepcidin is an important hormone in iron metabolism. Furthermore, hepcidin is associated with atherosclerotic disease. Therefore, this study aims to investigate the relation of serum hepcidin-25 (SH-25) and sub-clinic atherosclerosis measured by carotid intima-media thickness (CIMT) and mortality in HD patients. METHODS We enrolled 82 HD patients in a cross-control study. We measured SH-25 using ELISA kit and CIMT using high-resolution real-time ultrasonography. After 4 years of first assessment, we investigated the relation between all-cause and cardiovascular mortality and SH-25 and CIMT. RESULTS Two patients were excluded because of renal transplantation. The survivors were younger (53.7±15.1 vs. 65.2±15.5; p<0.05) and CIMT was lower (0.83±0.2 vs. 0.95±0.2; p<0.05); however, there was no significant difference in SH-25 levels between the groups (29.1±13 vs. 32.4±22.4; p=0.767). The patients who died of CVD were significantly older (63.7±16.1 vs. 53.7±15.1; p<0.05) and had significantly higher CIMT (0.94±0.2 vs. 83±0.2; p<0.05). The SH-25 levels were statistically significantly higher in patients who died of CVD (40.3±25 vs. 29.1±13; p<0.05). Linear regression analysis showed a positive correlation between CIMT and SH-25 in the study population and in those who died from CVD (r=0.41; p<0.05 and r=0.606; p<0.05, respectively). CONCLUSION This study suggests that hepcidin is effective in cardiovascular mortality and pathophysiology of subclinical atherosclerosis in HD patients.
Collapse
|
33
|
Mohammadifard N, Humphries KH, Gotay C, Mena-Sánchez G, Salas-Salvadó J, Esmaillzadeh A, Ignaszewski A, Sarrafzadegan N. Trace minerals intake: Risks and benefits for cardiovascular health. Crit Rev Food Sci Nutr 2017; 59:1334-1346. [PMID: 29236516 DOI: 10.1080/10408398.2017.1406332] [Citation(s) in RCA: 101] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Minerals play a major role in regulating cardiovascular function. Imbalances in electrolyte minerals are frequent and potentially hazardous occurrences that may lead to the development of cardiovascular diseases (CVDs). Transition metals, such as iron, zinc, copper and selenium, play a major role in cell metabolism. However, there is controversy over the effects of dietary and supplemental intake of these metals on cardiovascular risk factors and events. Since their pro-oxidant or antioxidant functions can have different effects on cardiovascular health. While deficiency of these trace elements can cause cardiovascular dysfunction, several studies have also shown a positive association between metal serum levels and cardiovascular risk factors and events. Thus, a J- or U-shaped relationship between the transition minerals and cardiovascular events has been proposed. Given the existing controversies, large, well-designed, long-term, randomized clinical trials are required to better examine the effects of trace mineral intake on cardiovascular events and all-cause mortality in the general population. In this review, we discuss the role of dietary and/or supplemental iron, copper, zinc, and selenium on cardiovascular health. We will also clarify their clinical applications, benefits, and harms in CVDs prevention.
Collapse
Affiliation(s)
- Noushin Mohammadifard
- a Hypertension Research Center , Cardiovascular Research Institute, Isfahan University of Medical Sciences , Isfahan , Iran
- b Interventional Cardiology Research Center , Cardiovascular Research Institute, Isfahan University of Medical Sciences , Isfahan , Iran
| | - Karin H Humphries
- c Women's Cardiovascular Health , Department of Medicine, The University of British Columbia , Vancouver , Canada
| | - Carolyn Gotay
- d Centre of Excellence in Cancer Prevention, Faculty of Medicine, School of Population and Public Health, The University of British Columbia , Vancouver , Canada
| | - Guillermo Mena-Sánchez
- e Human Nutrition Unit , Department of Biochemistry & Biotechnology , IISPV, School of Medicine, Rovira i Virgili University, and CIBER Obesity and Nutrition , Reus , Spain
| | - Jordi Salas-Salvadó
- e Human Nutrition Unit , Department of Biochemistry & Biotechnology , IISPV, School of Medicine, Rovira i Virgili University, and CIBER Obesity and Nutrition , Reus , Spain
| | - Ahmad Esmaillzadeh
- f Obesity and Eating Habits Research Center , Endocrinology and Metabolism Molecular, Cellular Sciences Institute, Tehran University of Medical Sciences , Tehran , Iran
- g Department of Community Nutrition , School of Nutritional Sciences and Dietetics, Tehran University of Medical Sciences , Tehran , Iran
- h Department of Community Nutrition , School of Nutrition and Food Science, Isfahan University of Medical Sciences , Isfahan , Iran
| | - Andrew Ignaszewski
- i Division of Cardiology, Faculty of Medicine, The University of British Columbia , Vancouver , Canada
| | - Nizal Sarrafzadegan
- j Isfahan Cardiovascular Research Center , Cardiovascular Research Institute, Isfahan University of Medical Sciences , Isfahan , Iran
| |
Collapse
|
34
|
Associations of plasma hepcidin with mortality risk in patients with coronary artery disease. Oncotarget 2017; 8:109497-109508. [PMID: 29312624 PMCID: PMC5752537 DOI: 10.18632/oncotarget.22722] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2017] [Accepted: 07/06/2017] [Indexed: 12/18/2022] Open
Abstract
Background Increased blood hepcidin may be associated with the presence and promotion of atherosclerosis, the association of hepcidin with mortality among coronary artery disease (CAD) patients remains unknown. We sought to assess the relationship of hepcidin and all-cause and cardiovascular disease (CVD) mortality among CAD patients with and without acute coronary syndrome (ACS). Methods and Results This study included 759 patients with ACS and 526 patients with stable CAD. After an average follow-up of 4.1 years, 154 deaths were recorded, 114 were due to CVD. After adjusting for CVD risk factors and inflammatory markers, the plasma hepcidin was positively associated with all-cause and CVD mortality in the ACS patients, the multivariable-adjusted hazard ratios (HRs) across tertiles of hepcidin were 1.00, 2.18 (95% CI 1.23-3.94), and 2.82 (95% CI 1.59-5.12) for all-cause mortality (Ptrend=0.006), and 1.00, 2.20 (95% CI 1.12-4.05), and 2.64 (95% CI 1.41-5.65) for CVD mortality (Ptrend=0.01). The C-index and net reclassification improvement when including hepcidin in traditional CVD models were 1.6% and 21.5% for all-cause mortality, 1.4% and 23.5% for CVD mortality, respectively, (P<0.001). Conclusions Plasma hepcidin was positively associated with mortality in ACS patients. Hepcidin may be a potential biomarker for risk prediction in ACS patients.
Collapse
|
35
|
Ambachew S, Biadgo B. Hepcidin in Iron Homeostasis: Diagnostic and Therapeutic Implications in Type 2 Diabetes Mellitus Patients. Acta Haematol 2017; 138:183-193. [PMID: 29136618 DOI: 10.1159/000481391] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2017] [Accepted: 09/08/2017] [Indexed: 12/21/2022]
Abstract
The prevalence of type 2 diabetes is increasing in epidemic proportions worldwide. Evidence suggests body iron overload is frequently linked and observed in patients with type 2 diabetes. Body iron metabolism is based on iron conservation and recycling by which only a part of the daily need is replaced by duodenal absorption. The principal liver-produced peptide called hepcidin plays a fundamental role in iron metabolism. It directly binds to ferroportin, the sole iron exporter, resulting in the internalization and degradation of ferroportin. However, inappropriate production of hepcidin has been shown to play a role in the pathogenesis of type 2 diabetes mellitus and its complications, based on the regulation and expression in iron-abundant cells. Underexpression of hepcidin results in body iron overload, which triggers the production of reactive oxygen species simultaneously thought to play a major role in diabetes pathogenesis mediated both by β-cell failure and insulin resistance. Increased hepcidin expression results in increased intracellular sequestration of iron, and is associated with the complications of type 2 diabetes. Besides, hepcidin concentrations have been linked to inflammatory cytokines, matriptase 2, and chronic hepatitis C infection, which have in turn been reported to be associated with diabetes by several approaches. Either hepcidin-targeted therapy alone or as adjunctive therapy with phlebotomy, iron chelators, or dietary iron restriction may be able to alter iron parameters in diabetic patients. Therefore, measuring hepcidin may improve differential diagnosis and the monitoring of disorders of iron metabolism.
Collapse
Affiliation(s)
- Sintayehu Ambachew
- Department of Clinical Chemistry, School of Biomedical and Laboratory Sciences, College of Medicine and Health Sciences, University of Gondar, Gondar, Ethiopia
| | | |
Collapse
|
36
|
Heme Oxygenase Induction Suppresses Hepatic Hepcidin and Rescues Ferroportin and Ferritin Expression in Obese Mice. J Nutr Metab 2017; 2017:4964571. [PMID: 29062571 PMCID: PMC5618758 DOI: 10.1155/2017/4964571] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2017] [Accepted: 08/08/2017] [Indexed: 12/17/2022] Open
Abstract
Hepcidin, a phase II reactant secreted by hepatocytes, regulates cellular iron levels by increasing internalization of ferroportin-a transmembrane protein facilitating egress of cellular iron. Chronic low-grade inflammatory states, such as obesity, have been shown to increase oxidative stress and enhance hepcidin secretion from hepatocytes and macrophages. Heme-heme oxygenase (HO) is a stress response system which reduces oxidative stress. We investigated the effects of HO-1 induction on hepatic hepcidin levels and on iron homeostasis in hepatic tissues from lean and obese mice. Obese mice exhibited hyperglycemia (p < 0.05); increased levels of proinflammatory cytokines (MCP-1, IL-6, p < 0.05); oxidative stress (p < 0.05); and increased hepatic hepcidin levels (p < 0.05). Enhancement of hepcidin was reflected in the reduced expression of ferroportin in obese mice (p < 0.05). However, this effect is accompanied by a significant decline in ferritin expression. Additionally, there are reduced insulin receptor phosphorylation and attenuation of metabolic regulators pAMPK, pAKT, and pLKB1. Cobalt protoporphyrin- (CoPP-) induced HO-1 upregulation in obese mice reversed these alterations (p < 0.05), while attenuating hepatic hepcidin levels. These effects of CoPP were prevented in obese mice concurrently exposed to an inhibitor of HO (SnMP) (p < 0.05). Our results highlight a modulatory effect of HO on iron homeostasis mediated through the suppression of hepatic hepcidin.
Collapse
|
37
|
Petrova J, Manolov V, Vasilev V, Tzatchev K, Marinov B. Ischemic stroke, inflammation, iron overload - Connection to a hepcidin. Int J Stroke 2016; 11:NP16-7. [PMID: 26763035 DOI: 10.1177/1747493015607509] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Affiliation(s)
- Julia Petrova
- Department of Neurology, Medical University, Sofia, Bulgaria
| | - Victor Manolov
- Department of Medical Genetics, Medical University, Sofia, Bulgaria
| | - Vasil Vasilev
- Department of Clinical Laboratory and Clinical Immunology, Medical University, Sofia, Bulgaria
| | - Kamen Tzatchev
- Department of Clinical Laboratory and Clinical Immunology, Medical University, Sofia, Bulgaria
| | | |
Collapse
|
38
|
Wang Q, Ji J, Hao S, Zhang M, Li K, Qiao T. Iron Together with Lipid Downregulates Protein Levels of Ceruloplasmin in Macrophages Associated with Rapid Foam Cell Formation. J Atheroscler Thromb 2016; 23:1201-1211. [PMID: 27040361 PMCID: PMC5098920 DOI: 10.5551/jat.32292] [Citation(s) in RCA: 16] [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: 07/25/2015] [Accepted: 01/26/2016] [Indexed: 01/31/2023] Open
Abstract
AIM Iron accumulation in foam cells was previously shown to be involved in atherogenesis. However, the mechanism for iron accumulation was not clarified. Ceruloplasmin (Cp) is an important factor in cellular iron efflux and was found to be downregulated in atherosclerotic plaques in our previous study. The current study is to investigate the role of Cp in atherosclerosis. METHODS We used RAW264.7 cells, a well-accepted cell model of atherosclerosis, which were treated with lipopolysaccharides (LPS), ferric ammonium citrate (FAC) or deferoxamine, and oxidized low density lipoprotein (ox-LDL) to detect the regulation of Cp and its influence in iron efflux and lipid accumulation using biochemical and histological assays. RESULTS Our results showed that the Cp protein level increased after 200-μM FAC treatment in LPS-activated RAW264.7 cells. Ox-LDL treatment (50 μg/ml) moderately reduced both mRNA and protein levels and ferroxidase activity of Cp (p<0.05). No significant difference was observed in the expression of ferritin and ferroportin, two important iron-related proteins for iron storage and efflux, respectively, after ox-LDL treatment. However, co-treatment with ox-LDL and FAC drastically reduced the expression of Cp. Accordingly, the ferroxidase activities simultaneously decreased, whereas the protein levels of Ft and Fpn1 significantly increased, indicating further iron accumulation. Moreover, co-treatment with FAC and ox-LDL enhanced the accumulation of cholesterol compared with ox-LDL-only treatment to trigger apoptosis. CONCLUSION Our findings suggest that physiological interaction of iron and lipid obstructs iron efflux and accelerates the lipid accumulation in macrophages during foam cell formation, which implicates the role of iron in the pathology of atherosclerosis.
Collapse
Affiliation(s)
- Qi Wang
- Department of Vascular Surgery, Drum Tower Clinical Medical College of Nanjing Medical University, Nanjing, P.R. China
| | - Jiajie Ji
- Department of Vascular Surgery, Drum Tower Clinical Medical College of Nanjing Medical University, Nanjing, P.R. China
| | - Shuangying Hao
- Jiangsu Key Laboratory for Molecular Medicine, Medical School of Nanjing University, Nanjing, China
| | - Meng Zhang
- Department of Vascular Surgery, Drum Tower Clinical Medical College of Nanjing Medical University, Nanjing, P.R. China
| | - Kuanyu Li
- Jiangsu Key Laboratory for Molecular Medicine, Medical School of Nanjing University, Nanjing, China
| | - Tong Qiao
- Department of Vascular Surgery, Drum Tower Clinical Medical College of Nanjing Medical University, Nanjing, P.R. China
- Department of Vascular Surgery, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, P.R. China
| |
Collapse
|
39
|
Mechanisms and Clinical Application of Tetramethylpyrazine (an Interesting Natural Compound Isolated from Ligusticum Wallichii): Current Status and Perspective. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2016; 2016:2124638. [PMID: 27668034 PMCID: PMC5030435 DOI: 10.1155/2016/2124638] [Citation(s) in RCA: 81] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/17/2016] [Accepted: 08/09/2016] [Indexed: 01/09/2023]
Abstract
Tetramethylpyrazine, a natural compound from Ligusticum wallichii (Chuan Xiong), has been extensively used in China for cardiovascular and cerebrovascular diseases for about 40 years. Because of its effectiveness in multisystems, especially in cardiovascular, its pharmacological action, clinical application, and the structural modification have attracted broad attention. In this paper its mechanisms of action, the clinical status, and synthetic derivatives will be reviewed briefly.
Collapse
|
40
|
Ha JY, Kim MK, Kang S, Nam JS, Ahn CW, Kim KR, Park JS. Serum ferritin levels are associated with arterial stiffness in healthy Korean adults. Vasc Med 2016; 21:325-30. [DOI: 10.1177/1358863x16629728] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Although an association between serum ferritin and atherosclerosis has been suggested, limited epidemiologic data are available regarding the association between ferritin and arterial stiffness in healthy adults. A total of 2932 healthy subjects were enrolled in this study. Anthropometric and biochemical profiles including ferritin were measured. The arterial stiffness was measured using brachial–ankle pulse wave velocity (baPWV). Serum ferritin levels were classified into quartiles and baPWV values gradually increased with each ferritin quartile. Multiple regression analysis showed that ferritin levels were independently correlated with baPWV. After adjusting for multiple risk factors, as compared with the lowest quartile, the odds ratios for high baPWV (>75th percentile) were 1.15 (0.84–1.56), 1.37 (0.97–1.73), and 1.46 (1.29–2.17) among men ( p for trend < 0.05) and 1.24 (0.87–1.79), 1.53 (1.09–2.16), and 1.80 (1.25–2.82) among women ( p for trend < 0.05), for the second, third, and fourth quartiles of ferritin, respectively. In conclusion, serum ferritin levels are independently associated with arterial stiffness in healthy Korean adults.
Collapse
Affiliation(s)
- Ji Yoon Ha
- Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Korea
| | - Min Kyung Kim
- Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Korea
| | - Shinae Kang
- Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Korea
- Severance Institute for Vascular and Metabolic Research, Yonsei University College of Medicine, Seoul, Korea
| | - Ji Sun Nam
- Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Korea
- Severance Institute for Vascular and Metabolic Research, Yonsei University College of Medicine, Seoul, Korea
| | - Chul Woo Ahn
- Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Korea
- Severance Institute for Vascular and Metabolic Research, Yonsei University College of Medicine, Seoul, Korea
| | - Kyung Rae Kim
- Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Korea
| | - Jong Suk Park
- Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Korea
- Severance Institute for Vascular and Metabolic Research, Yonsei University College of Medicine, Seoul, Korea
| |
Collapse
|
41
|
McLachlan S, Giambartolomei C, White J, Charoen P, Wong A, Finan C, Engmann J, Shah T, Hersch M, Podmore C, Cavadino A, Jefferis BJ, Dale CE, Hypponen E, Morris RW, Casas JP, Kumari M, Ben-Shlomo Y, Gaunt TR, Drenos F, Langenberg C, Kuh D, Kivimaki M, Rueedi R, Waeber G, Hingorani AD, Price JF, Walker AP, UCLEB Consortium. Replication and Characterization of Association between ABO SNPs and Red Blood Cell Traits by Meta-Analysis in Europeans. PLoS One 2016; 11:e0156914. [PMID: 27280446 PMCID: PMC4900668 DOI: 10.1371/journal.pone.0156914] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2016] [Accepted: 05/20/2016] [Indexed: 01/07/2023] Open
Abstract
Red blood cell (RBC) traits are routinely measured in clinical practice as important markers of health. Deviations from the physiological ranges are usually a sign of disease, although variation between healthy individuals also occurs, at least partly due to genetic factors. Recent large scale genetic studies identified loci associated with one or more of these traits; further characterization of known loci and identification of new loci is necessary to better understand their role in health and disease and to identify potential molecular mechanisms. We performed meta-analysis of Metabochip association results for six RBC traits—hemoglobin concentration (Hb), hematocrit (Hct), mean corpuscular hemoglobin (MCH), mean corpuscular hemoglobin concentration (MCHC), mean corpuscular volume (MCV) and red blood cell count (RCC)—in 11 093 Europeans from seven studies of the UCL-LSHTM-Edinburgh-Bristol (UCLEB) Consortium. We identified 394 non-overlapping SNPs in five loci at genome-wide significance: 6p22.1-6p21.33 (with HFE among others), 6q23.2 (with HBS1L among others), 6q23.3 (contains no genes), 9q34.3 (only ABO gene) and 22q13.1 (with TMPRSS6 among others), replicating previous findings of association with RBC traits at these loci and extending them by imputation to 1000 Genomes. We further characterized associations between ABO SNPs and three traits: hemoglobin, hematocrit and red blood cell count, replicating them in an independent cohort. Conditional analyses indicated the independent association of each of these traits with ABO SNPs and a role for blood group O in mediating the association. The 15 most significant RBC-associated ABO SNPs were also associated with five cardiometabolic traits, with discordance in the direction of effect between groups of traits, suggesting that ABO may act through more than one mechanism to influence cardiometabolic risk.
Collapse
Affiliation(s)
- Stela McLachlan
- Centre for Population Health Sciences, Usher Institute of Population Health Sciences and Informatics, University of Edinburgh, Edinburgh, United Kingdom
- * E-mail:
| | - Claudia Giambartolomei
- Department of Psychiatry, Icahn Institute for Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, The Leon and Norma Hess Center for Science and Medicine, New York, New York, United States of America
| | - Jon White
- University College London Genetics Institute, Department of Genetics, Environment and Evolution, London, United Kingdom
| | - Pimphen Charoen
- Department of Non-communicable Disease Epidemiology, Faculty of Epidemiology and Population Health, London School of Hygiene and Tropical Medicine, London, United Kingdom
- Department of Tropical Hygiene, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - Andrew Wong
- MRC Unit for Lifelong Health and Ageing at UCL, London, United Kingdom
| | - Chris Finan
- Institute of Cardiovascular Science, University College London, London, United Kingdom
| | - Jorgen Engmann
- Genetic Epidemiology Group, Institute of Cardiovascular Science, University College London, London, United Kingdom
- Farr Institute of Health Informatics Research, Department of Epidemiology & Public Health, University College London, London, United Kingdom
| | - Tina Shah
- Genetic Epidemiology Group, Institute of Cardiovascular Science, University College London, London, United Kingdom
| | - Micha Hersch
- Department of Medical Genetics, University of Lausanne, Lausanne, Switzerland
- Swiss Institute of Bioinformatics, Lausanne, Switzerland
| | - Clara Podmore
- MRC Epidemiology Unit, University of Cambridge School of Clinical Medicine, Institute of Metabolic Science, Cambridge Biomedical Campus, Cambridge, United Kingdom
| | - Alana Cavadino
- Centre for Environmental and Preventive Medicine, Wolfson Institute of Preventive Medicine, Queen Mary University of London, London, United Kingdom
- Population, Policy and Practice, UCL Institute of Child Health, University College London, London, United Kingdom
| | - Barbara J. Jefferis
- Department of Primary Care & Population Health, UCL Institute of Epidemiology & Health Care, University College London, London, United Kingdom
| | - Caroline E. Dale
- Farr Institute of Health Informatics Research, Department of Epidemiology & Public Health, University College London, London, United Kingdom
| | - Elina Hypponen
- Population, Policy and Practice, UCL Institute of Child Health, University College London, London, United Kingdom
- Centre for Population Health Research, School of Health Sciences and Sansom Institute of Health Research, University of South Australia, Adelaide, Australia
- South Australian Health and Medical Research Institute, Adelaide, Australia
| | - Richard W. Morris
- Department of Primary Care & Population Health, UCL Institute of Epidemiology & Health Care, University College London, London, United Kingdom
- School of Social and Community Medicine, University of Bristol, Bristol, United Kingdom
| | - Juan P. Casas
- Institute of Health Informatics, University College London, London, United Kingdom
| | - Meena Kumari
- Institute for Social and Economic Research, University of Essex, Colchester, United Kingdom
- Department of Epidemiology & Public Health, UCL Institute of Epidemiology & Health Care, University College London, London, United Kingdom
| | - Yoav Ben-Shlomo
- School of Social and Community Medicine, University of Bristol, Bristol, United Kingdom
| | - Tom R. Gaunt
- MRC Integrative Epidemiology Unit, School of Social and Community Medicine, University of Bristol, Bristol, United Kingdom
| | - Fotios Drenos
- MRC Integrative Epidemiology Unit, School of Social and Community Medicine, University of Bristol, Bristol, United Kingdom
- Centre for Cardiovascular Genetics, Institute of Cardiovascular Science, University College London, London, United Kingdom
| | - Claudia Langenberg
- MRC Epidemiology Unit, University of Cambridge School of Clinical Medicine, Institute of Metabolic Science, Cambridge Biomedical Campus, Cambridge, United Kingdom
- Institute of Health Informatics, University College London, London, United Kingdom
| | - Diana Kuh
- MRC Unit for Lifelong Health and Ageing at UCL, London, United Kingdom
| | - Mika Kivimaki
- Department of Epidemiology & Public Health, UCL Institute of Epidemiology & Health Care, University College London, London, United Kingdom
| | - Rico Rueedi
- Department of Medical Genetics, University of Lausanne, Lausanne, Switzerland
- Swiss Institute of Bioinformatics, Lausanne, Switzerland
| | - Gerard Waeber
- Department of Internal Medicine, Centre Hospitalier Universitaire Vaudois and University of Lausanne, Lausanne, Switzerland
| | - Aroon D. Hingorani
- Institute of Cardiovascular Science, University College London, London, United Kingdom
- Farr Institute of Health Informatics Research, Department of Epidemiology & Public Health, University College London, London, United Kingdom
| | - Jacqueline F. Price
- Centre for Population Health Sciences, Usher Institute of Population Health Sciences and Informatics, University of Edinburgh, Edinburgh, United Kingdom
| | - Ann P. Walker
- Centre for Cardiovascular Genetics, Institute of Cardiovascular Science, University College London, London, United Kingdom
| | | |
Collapse
|
42
|
Pisano G, Lombardi R, Fracanzani AL. Vascular Damage in Patients with Nonalcoholic Fatty Liver Disease: Possible Role of Iron and Ferritin. Int J Mol Sci 2016; 17:ijms17050675. [PMID: 27164079 PMCID: PMC4881501 DOI: 10.3390/ijms17050675] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2016] [Revised: 04/20/2016] [Accepted: 04/26/2016] [Indexed: 02/06/2023] Open
Abstract
Non Alcoholic Fatty Liver Disease (NAFLD) is the most common chronic liver disease in Western countries. Recent data indicated that NAFLD is a risk factor by itself contributing to the development of cardiovascular disease independently of classical known risk factors. Hyperferritinemia and mild increased iron stores are frequently observed in patients with NAFLD and several mechanisms have been proposed to explain the role of iron, through oxidative stress and interaction with insulin metabolism, in the development of vascular damage. Moreover, iron depletion has been shown to decrease atherogenesis in experimental models and in humans. This review presents the recent evidence on epidemiology, pathogenesis, and the possible explanation of the role of iron and ferritin in the development of cardiovascular damage in patients with NAFLD, and discusses the possible interplay between metabolic disorders associated with NAFLD and iron in the development of cardiovascular disease.
Collapse
Affiliation(s)
- Giuseppina Pisano
- Department of Pathophysiology and Transplantation, Ca' Granda IRCCS Foundation, Policlinico Hospital, University of Milan, Centre of the Study of Metabolic and Liver Diseases, Via Francesco Sforza 35, 20122 Milan, Italy.
| | - Rosa Lombardi
- Department of Pathophysiology and Transplantation, Ca' Granda IRCCS Foundation, Policlinico Hospital, University of Milan, Centre of the Study of Metabolic and Liver Diseases, Via Francesco Sforza 35, 20122 Milan, Italy.
| | - Anna Ludovica Fracanzani
- Department of Pathophysiology and Transplantation, Ca' Granda IRCCS Foundation, Policlinico Hospital, University of Milan, Centre of the Study of Metabolic and Liver Diseases, Via Francesco Sforza 35, 20122 Milan, Italy.
| |
Collapse
|
43
|
Marques L, Negre-Salvayre A, Costa L, Canonne-Hergaux F. Iron gene expression profile in atherogenic Mox macrophages. Biochim Biophys Acta Mol Basis Dis 2016; 1862:1137-46. [PMID: 26972048 DOI: 10.1016/j.bbadis.2016.03.004] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2015] [Revised: 03/08/2016] [Accepted: 03/09/2016] [Indexed: 02/06/2023]
Abstract
RATIONALE The role of macrophage iron in the physiopathology of atherosclerosis is an open question that needs to be clarified. In atherosclerotic lesions, recruited macrophages are submitted to cytokines and oxidized lipids which influence their phenotype. An important phenotypic population driven by oxidized phospholipids is the Mox macrophages which present unique biological properties but their iron phenotype is not well described. OBJECTIVE To investigate the effect of Mox polarization by oxidized LDL (oxLDL) on macrophage iron metabolism in the absence or presence of proinflammatory stimuli. METHODS Bone marrow-derived macrophages were treated with different sources of LDL and/or LPS/IFNγ (M1 activator). Expression of ferroportin (Slc40a1, alias Fpn), heme oxygenase-1 (Hmox1), H- and L-ferritin (Fth1 and Ftl1), hepcidin (Hamp), ceruloplasmin (Cp) and interleukine-6 (Il6) was followed by quantitative PCR. FPN and HMOX1 protein expression was analyzed by immunofluorescence and in-cell-Western blotting. RESULTS Mox macrophages expressed increased Hmox1 and Fth1 levels with basal FPN protein levels despite the significant increase of Fpn mRNA. Upregulation of Hmox1 and Fpn mRNA was specific to LDL oxidative modification and mediated by NRF2. The downregulation of both Cp isoforms and the upregulation of Hamp expression observed in Mox macrophages suggest that FPN mediated iron export could be compromised. Simultaneous exposure to oxLDL and LPS/IFNγ leads to a mixed Mox/M1 phenotype that is closer to M1. CONCLUSION A microenvironment rich in oxLDL and proinflammatory cytokines could promote macrophage iron retention and lipid accumulation profiles, a specific cell phenotype that likely contributes to lesion development and plaque instability in atherosclerosis.
Collapse
Affiliation(s)
- Liliana Marques
- INSERM UMR 1043, CNRS UMR 5282, Université de Toulouse, UPS, Centre de Physiopathologie de Toulouse Purpan (CPTP), Toulouse, France; Departamento da Promoção da Saúde e Prevenção de Doenças Não Transmissíveis, Instituto Nacional de Saúde Doutor Ricardo Jorge, Lisboa, Portugal; University of Lisboa, Faculty of Sciences, BioISI - Biosystems & Integrative Sciences Institute, Campo Grande, Lisboa, Portugal
| | | | - Luciana Costa
- Departamento da Promoção da Saúde e Prevenção de Doenças Não Transmissíveis, Instituto Nacional de Saúde Doutor Ricardo Jorge, Lisboa, Portugal; University of Lisboa, Faculty of Sciences, BioISI - Biosystems & Integrative Sciences Institute, Campo Grande, Lisboa, Portugal.
| | - François Canonne-Hergaux
- INSERM UMR 1043, CNRS UMR 5282, Université de Toulouse, UPS, Centre de Physiopathologie de Toulouse Purpan (CPTP), Toulouse, France; IRSD, Université de Toulouse, INSERM, INRA, INP-ENVT, Université de Toulouse 3 Paul Sabatier, Toulouse, France.
| |
Collapse
|
44
|
Prats-Puig A, Moreno M, Carreras-Badosa G, Bassols J, Ricart W, López-Bermejo A, Fernández-Real JM. Serum Ferritin Relates to Carotid Intima-Media Thickness in Offspring of Fathers With Higher Serum Ferritin Levels. Arterioscler Thromb Vasc Biol 2015; 36:174-80. [PMID: 26586658 DOI: 10.1161/atvbaha.115.306396] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2015] [Accepted: 11/09/2015] [Indexed: 01/09/2023]
Abstract
OBJECTIVE Body iron status has been linked to atherosclerosis in adults. The purposes of our study were to determine (1) the association between circulating ferritin levels and carotid intima-media thickness (cIMT) in a cohort of apparently healthy children and (2) the association between cIMT and parental ferritin levels. APPROACH AND RESULTS Circulating ferritin levels (microparticle enzyme immunoassay), metabolic parameters, and cIMT (ultrasonography) were analyzed cross-sectionally in a cohort of 692 healthy white children with a mean age of 8 ± 2 years (52% girls and 48% boys). In consecutive 123 children from the cross-sectional sample, the same serum assessments were also performed at baseline in their parents, and the cIMT was repeated after 3 years of follow-up in the children at a mean age of 11 ± 2 years (53% girls and 47% boys). Weak but significant positive associations were evident between children's circulating ferritin levels and cIMT (r=0.123; P=0.001) and with the change in cIMT 3 years later a tendency was also observed (r=0.185; P=0.048). In multiple regression analyses, circulating ferritin levels contributed independently to cIMT variance (β=0.090; P=0.026; R(2)=10%) and cIMT change variance (β=0.216; P=0.019; R(2)= 3.4%) after controlling for body mass index, high-sensitivity C-reactive protein, age, sex, and low-density lipoprotein-cholesterol levels. This association was, however, remarkably significant (β=0.509; P=0.001; R(2)= 20.4%) in children whose fathers had ferritin levels above the median value (122.5 ng/mL).The latter association remained significant after correction for multiple testing. Maternal's ferritin levels showed no interaction in this association. CONCLUSIONS These results suggest a paternal-specific effect on cIMT partially reflected by father's ferritin levels.
Collapse
Affiliation(s)
- Anna Prats-Puig
- From the Girona Institute for Biomedical Research, Girona, Spain (A.P.-P., M.M., G.C.-B., J.B., W.R., A.L.-B., J.M.F.-R.); Department of Physical Therapy, EUSES University School (A.P.-P.) and TransLab Research Group, Department of Medical Sciences, Faculty of Medicine (A.L.-B.), University of Girona, Girona, Spain; Pediatrics, Dr. JosepTrueta Hospital, Girona, Spain (A.P.-P., G.C.-B., J.B., A.L.-B.); and Department of Diabetes, Endocrinology, CIBEROBN (CB06/03/010) and Instituto de Salud Carlos III (ISCIII), Girona, Spain (M.M., W.R., J.M.F.-R.)
| | - María Moreno
- From the Girona Institute for Biomedical Research, Girona, Spain (A.P.-P., M.M., G.C.-B., J.B., W.R., A.L.-B., J.M.F.-R.); Department of Physical Therapy, EUSES University School (A.P.-P.) and TransLab Research Group, Department of Medical Sciences, Faculty of Medicine (A.L.-B.), University of Girona, Girona, Spain; Pediatrics, Dr. JosepTrueta Hospital, Girona, Spain (A.P.-P., G.C.-B., J.B., A.L.-B.); and Department of Diabetes, Endocrinology, CIBEROBN (CB06/03/010) and Instituto de Salud Carlos III (ISCIII), Girona, Spain (M.M., W.R., J.M.F.-R.)
| | - Gemma Carreras-Badosa
- From the Girona Institute for Biomedical Research, Girona, Spain (A.P.-P., M.M., G.C.-B., J.B., W.R., A.L.-B., J.M.F.-R.); Department of Physical Therapy, EUSES University School (A.P.-P.) and TransLab Research Group, Department of Medical Sciences, Faculty of Medicine (A.L.-B.), University of Girona, Girona, Spain; Pediatrics, Dr. JosepTrueta Hospital, Girona, Spain (A.P.-P., G.C.-B., J.B., A.L.-B.); and Department of Diabetes, Endocrinology, CIBEROBN (CB06/03/010) and Instituto de Salud Carlos III (ISCIII), Girona, Spain (M.M., W.R., J.M.F.-R.)
| | - Judit Bassols
- From the Girona Institute for Biomedical Research, Girona, Spain (A.P.-P., M.M., G.C.-B., J.B., W.R., A.L.-B., J.M.F.-R.); Department of Physical Therapy, EUSES University School (A.P.-P.) and TransLab Research Group, Department of Medical Sciences, Faculty of Medicine (A.L.-B.), University of Girona, Girona, Spain; Pediatrics, Dr. JosepTrueta Hospital, Girona, Spain (A.P.-P., G.C.-B., J.B., A.L.-B.); and Department of Diabetes, Endocrinology, CIBEROBN (CB06/03/010) and Instituto de Salud Carlos III (ISCIII), Girona, Spain (M.M., W.R., J.M.F.-R.)
| | - Wifredo Ricart
- From the Girona Institute for Biomedical Research, Girona, Spain (A.P.-P., M.M., G.C.-B., J.B., W.R., A.L.-B., J.M.F.-R.); Department of Physical Therapy, EUSES University School (A.P.-P.) and TransLab Research Group, Department of Medical Sciences, Faculty of Medicine (A.L.-B.), University of Girona, Girona, Spain; Pediatrics, Dr. JosepTrueta Hospital, Girona, Spain (A.P.-P., G.C.-B., J.B., A.L.-B.); and Department of Diabetes, Endocrinology, CIBEROBN (CB06/03/010) and Instituto de Salud Carlos III (ISCIII), Girona, Spain (M.M., W.R., J.M.F.-R.)
| | - Abel López-Bermejo
- From the Girona Institute for Biomedical Research, Girona, Spain (A.P.-P., M.M., G.C.-B., J.B., W.R., A.L.-B., J.M.F.-R.); Department of Physical Therapy, EUSES University School (A.P.-P.) and TransLab Research Group, Department of Medical Sciences, Faculty of Medicine (A.L.-B.), University of Girona, Girona, Spain; Pediatrics, Dr. JosepTrueta Hospital, Girona, Spain (A.P.-P., G.C.-B., J.B., A.L.-B.); and Department of Diabetes, Endocrinology, CIBEROBN (CB06/03/010) and Instituto de Salud Carlos III (ISCIII), Girona, Spain (M.M., W.R., J.M.F.-R.).
| | - José Manuel Fernández-Real
- From the Girona Institute for Biomedical Research, Girona, Spain (A.P.-P., M.M., G.C.-B., J.B., W.R., A.L.-B., J.M.F.-R.); Department of Physical Therapy, EUSES University School (A.P.-P.) and TransLab Research Group, Department of Medical Sciences, Faculty of Medicine (A.L.-B.), University of Girona, Girona, Spain; Pediatrics, Dr. JosepTrueta Hospital, Girona, Spain (A.P.-P., G.C.-B., J.B., A.L.-B.); and Department of Diabetes, Endocrinology, CIBEROBN (CB06/03/010) and Instituto de Salud Carlos III (ISCIII), Girona, Spain (M.M., W.R., J.M.F.-R.).
| |
Collapse
|
45
|
Kali A, Yayar O, Erdogan B, Eser B, Buyukbakkal M, Ercan Z, Merhametsiz O, Haspulat A, Gök Oğuz E, Canbakan B, Ayli MD. Is hepcidin-25 a predictor of atherosclerosis in hemodialysis patients? Hemodial Int 2015; 20:191-7. [PMID: 26374145 DOI: 10.1111/hdi.12355] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Atherosclerotic cardiovascular disease is an important cause of mortality and morbidity in hemodialysis patients. Iron accumulation in arterial wall macrophages is increased in atherosclerotic lesions. Hepcidin is a key hepatic hormone regulating iron balance. It inhibits iron release from macrophages and iron absorption from enterocytes by binding and inactivating the cellular iron exporter ferroportin. The aim of this study is to investigate the relation of hepcidin-25, iron parameters, and atherosclerosis measured by carotid intima media thickness (CIMT) in hemodialysis patients. Eighty-two hemodialysis patients were enrolled in this cross-sectional study. Predialysis blood samples were centrifuged at 1500 g and 4°C for 10 minutes and stored at -80°C for the measurement of hepcidin-25. DRG hepcidin enzyme-linked immunosorbent assay kit was used for the measurement of hepcidin-25. Ultrasonographical B-mode imaging of bilateral carotid arteries was performed with a high-resolution real-time ultrasonography (Mindray DC7). Mean age of the study population was 57.90 ± 16.08 years and 43.9% were men. Total study population was grouped into two according to median value of hepcidin-25. There was no difference between groups with respect to age, dialysis vintage, and C-reactive protein. CIMT was found to be statistically significantly higher in low hepcidin-25 group. In correlation analysis, CIMT was found to be correlated with age (P < 0.01, R = 0.33) and hepcidin-25 (P < 0.01, R = 0.46). In linear regression analysis, age (β = 0.31) and hepcidin-25 (β = 0.44) were found to be the determinants of CIMT in hemodialysis patients. Our results implicate that hepcidin may take part in pathophysiology of atherosclerosis and cardiovascular disease in hemodialysis patients.
Collapse
Affiliation(s)
- Alaaddin Kali
- Department of Nephrology, Diskapi Yildirim Beyazid Research and Training Hospital, Ankara, Turkey
| | - Ozlem Yayar
- Department of Nephrology, Diskapi Yildirim Beyazid Research and Training Hospital, Ankara, Turkey
| | - Bulent Erdogan
- Department of Nephrology, Diskapi Yildirim Beyazid Research and Training Hospital, Ankara, Turkey
| | - Baris Eser
- Department of Nephrology, Diskapi Yildirim Beyazid Research and Training Hospital, Ankara, Turkey
| | - Mehmet Buyukbakkal
- Department of Nephrology, Diskapi Yildirim Beyazid Research and Training Hospital, Ankara, Turkey
| | - Zafer Ercan
- Department of Nephrology, Diskapi Yildirim Beyazid Research and Training Hospital, Ankara, Turkey
| | - Ozgur Merhametsiz
- Department of Nephrology, Diskapi Yildirim Beyazid Research and Training Hospital, Ankara, Turkey
| | - Ayhan Haspulat
- Department of Nephrology, Diskapi Yildirim Beyazid Research and Training Hospital, Ankara, Turkey
| | - Ebru Gök Oğuz
- Department of Nephrology, Diskapi Yildirim Beyazid Research and Training Hospital, Ankara, Turkey
| | - Basol Canbakan
- Department of Nephrology, Diskapi Yildirim Beyazid Research and Training Hospital, Ankara, Turkey
| | - Mehmet D Ayli
- Department of Nephrology, Diskapi Yildirim Beyazid Research and Training Hospital, Ankara, Turkey
| |
Collapse
|
46
|
Iron Stores, Hepcidin, and Aortic Stiffness in Individuals with Hypertension. PLoS One 2015; 10:e0134635. [PMID: 26244503 PMCID: PMC4526526 DOI: 10.1371/journal.pone.0134635] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2015] [Accepted: 07/11/2015] [Indexed: 11/19/2022] Open
Abstract
Background & Aims Iron accumulation within the arterial wall has been hypothesized to promote atherosclerosis progression. Aim of this study was to evaluate whether the hormone hepcidin and iron stores are associated with arterial stiffness in subjects with essential hypertension. Methods Circulating hepcidin, ferritin, and mutations in the hemochromatosis gene were compared between subjects included in the first vs. third tertile (n=284 each) of carotid-femoral pulse wave velocity (PWV) in an unselected cohort of patients with arterial hypertension. Results At univariate logistic regression analysis, high PWV was associated with higher ferritin levels (p=0.010), but lower hepcidin (p=0.045), and hepcidin ferritin/ratio (p<0.001). Hemochromatosis mutations predisposing to iron overload were associated with high PWV (p=0.025). At multivariate logistic regression analysis, high aortic stiffness was associated with older age, male sex, lower BMI, higher systolic blood pressure and heart rate, hyperferritinemia (OR 2.05, 95% c.i. 1.11-3.17 per log ng/ml; p=0.022), and lower circulating hepcidin concentration (OR 0.29, 95% c.i. 0.16-0.51 per log ng/ml; p<0.001). In subgroup analyses, high PWV was associated with indices of target organ damage, including micro-albuminuria (n=125, p=0.038), lower ejection fraction (n=175, p=0.031), cardiac diastolic dysfunction (p=0.004), and lower S wave peak systolic velocity (p<0.001). Ferritin was associated with cardiac diastolic dysfunction, independently of confounders (p=0.006). Conclusions In conclusion, hyperferritinemia is associated with high aortic stiffness and cardiac diastolic dysfunction, while low circulating hepcidin with high aortic stiffness.
Collapse
|
47
|
Ji J, Zhou Y, Hao S, Wang Q, Li K, Qiao T. Low expression of ferroxidases is implicated in the iron retention in human atherosclerotic plaques. Biochem Biophys Res Commun 2015. [PMID: 26208458 DOI: 10.1016/j.bbrc.2015.07.091] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
The effect of iron on the progress of atherosclerosis is still controversial. To explore the relationship between atherosclerotic plaques and iron metabolism and how iron is accumulated in plaque macrophages, we performed Oil red O staining to detect the lipid of the atherosclerotic plaques, enzyme-linked immunosorbent assay to detect the intracellular lipids (total cholesterol, free cholesterol) and serum hepcidin, Western-blot to examine the iron-related proteins, immunohistochemical and immunofluorescence assays to localize ferroportin 1 in macrophages. The contents of serum iron and transferrin saturation were measured. The results confrimed that atherosclerotic plaques were all lipid-rich. Compared to normal vessel wall, atherosclerotic plaques had significantly higher levels of ferritin and ferroportin 1. Strikingly, we found the much lower levels of ferroxidases ceruloplasmin and hephaestin in plaque tissue than the normal vessel, while the content of serum hepcidin, iron and transferrin saturation were similar in these two groups. The novel finding suggests that the inability of ferrous iron to be oxidized into ferric iron might be a potential mechanism for iron retention in plaques.
Collapse
Affiliation(s)
- Jiajie Ji
- Department of Vascular Surgery, Drum Tower Clinical Medical College of Nanjing Medical University, Nanjing, PR China
| | - Yu Zhou
- Department of Vascular Surgery, Drum Tower Clinical Medical College of Nanjing Medical University, Nanjing, PR China
| | - Shuangying Hao
- Jiangsu Key Laboratory for Molecular Medicine, Medical School of Nanjing University, Nanjing, PR China
| | - Qi Wang
- Department of Vascular Surgery, Drum Tower Clinical Medical College of Nanjing Medical University, Nanjing, PR China
| | - Kuanyu Li
- Jiangsu Key Laboratory for Molecular Medicine, Medical School of Nanjing University, Nanjing, PR China.
| | - Tong Qiao
- Department of Vascular Surgery, Drum Tower Clinical Medical College of Nanjing Medical University, Nanjing, PR China; Department of Vascular Surgery, The Affiliated Drum Tower Hospital of Nanjing University, Nanjing, PR China.
| |
Collapse
|
48
|
Galesloot TE, Janss LL, Burgess S, Kiemeney LALM, den Heijer M, de Graaf J, Holewijn S, Benyamin B, Whitfield JB, Swinkels DW, Vermeulen SH. Iron and hepcidin as risk factors in atherosclerosis: what do the genes say? BMC Genet 2015; 16:79. [PMID: 26159428 PMCID: PMC4498499 DOI: 10.1186/s12863-015-0246-4] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2015] [Accepted: 06/30/2015] [Indexed: 01/05/2023] Open
Abstract
Background Previous reports suggested a role for iron and hepcidin in atherosclerosis. Here, we evaluated the causality of these associations from a genetic perspective via (i) a Mendelian randomization (MR) approach, (ii) study of association of atherosclerosis-related single nucleotide polymorphisms (SNPs) with iron and hepcidin, and (iii) estimation of genomic correlations between hepcidin, iron and atherosclerosis. Results Analyses were performed in a general population sample. Iron parameters (serum iron, serum ferritin, total iron-binding capacity and transferrin saturation), serum hepcidin and genome-wide SNP data were available for N = 1,819; non-invasive measurements of atherosclerosis (NIMA), i.e., presence of plaque, intima media thickness and ankle-brachial index (ABI), for N = 549. For the MR, we used 12 iron-related SNPs that were previously identified in a genome-wide association meta-analysis on iron status, and assessed associations of individual SNPs and quartiles of a multi-SNP score with NIMA. Quartile 4 versus quartile 1 of the multi-SNP score showed directionally consistent associations with the hypothesized direction of effect for all NIMA in women, indicating that increased body iron status is a risk factor for atherosclerosis in women. We observed no single SNP associations that fit the hypothesized directions of effect between iron and NIMA, except for rs651007, associated with decreased ferritin concentration and decreased atherosclerosis risk. Two of six NIMA-related SNPs showed association with the ratio hepcidin/ferritin, suggesting that an increased hepcidin/ferritin ratio increases atherosclerosis risk. Genomic correlations were close to zero, except for hepcidin and ferritin with ABI at rest [−0.27 (SE 0.34) and −0.22 (SE 0.35), respectively] and ABI after exercise [−0.29 (SE 0.34) and −0.30 (0.35), respectively]. The negative sign indicates an increased atherosclerosis risk with increased hepcidin and ferritin concentrations. Conclusions Our results suggest a potential causal role for hepcidin and ferritin in atherosclerosis, and may indicate that iron status is causally related to atherosclerosis in women. Electronic supplementary material The online version of this article (doi:10.1186/s12863-015-0246-4) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Tessel E Galesloot
- Radboud university medical center, Radboud Institute for Health Sciences, Nijmegen, The Netherlands.
| | - Luc L Janss
- Department of Molecular Biology and Genetics, Aarhus University, Aarhus, Denmark.
| | - Stephen Burgess
- Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK.
| | - Lambertus A L M Kiemeney
- Radboud university medical center, Radboud Institute for Health Sciences, Nijmegen, The Netherlands.
| | - Martin den Heijer
- Department of Internal Medicine, VU Medical Centre, Amsterdam, The Netherlands.
| | - Jacqueline de Graaf
- Department of General Internal Medicine, Division of Vascular Medicine, Radboud university medical center, Nijmegen, The Netherlands.
| | - Suzanne Holewijn
- Department of General Internal Medicine, Division of Vascular Medicine, Radboud university medical center, Nijmegen, The Netherlands. .,Research Vascular Center Rijnstate, Arnhem, The Netherlands.
| | - Beben Benyamin
- The University of Queensland, Queensland Brain Institute, St Lucia, Queensland, 4072, Australia. .,QIMR Berghofer Medical Research Institute, Brisbane, Queensland, 4029, Australia.
| | - John B Whitfield
- QIMR Berghofer Medical Research Institute, Brisbane, Queensland, 4029, Australia.
| | - Dorine W Swinkels
- Radboud university medical center, Radboud Institute for Health Sciences, Nijmegen, The Netherlands.
| | - Sita H Vermeulen
- Radboud university medical center, Radboud Institute for Health Sciences, Nijmegen, The Netherlands.
| |
Collapse
|
49
|
Connor JR, Zhang X, Nixon AM, Webb B, Perno JR. Comparative evaluation of nephrotoxicity and management by macrophages of intravenous pharmaceutical iron formulations. PLoS One 2015; 10:e0125272. [PMID: 25973894 PMCID: PMC4431721 DOI: 10.1371/journal.pone.0125272] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2014] [Accepted: 03/23/2015] [Indexed: 11/30/2022] Open
Abstract
BACKGROUND There is a significant clinical need for effective treatment of iron deficiency. A number of compounds that can be administered intravenously have been developed. This study examines how the compounds are handled by macrophages and their relative potential to provoke oxidative stress. METHODS Human kidney (HK-2) cells, rat peritoneal macrophages and renal cortical homogenates were exposed to pharmaceutical iron preparations. Analyses were performed for indices of oxidative stress and cell integrity. In addition, in macrophages, iron uptake and release and cytokine secretion was monitored. RESULTS HK-2 cell viability was decreased by iron isomaltoside and ferumoxytol and all compounds induced lipid peroxidation. In the renal cortical homogenates, lipid peroxidation occurred at lowest concentrations with ferric carboxymaltose, iron dextran, iron sucrose and sodium ferric gluconate. In the macrophages, iron sucrose caused loss of cell viability. Iron uptake was highest for ferumoxytol and iron isomaltoside and lowest for iron sucrose and sodium ferric gluconate. Iron was released as secretion of ferritin or as ferrous iron via ferroportin. The latter was blocked by hepcidin. Exposure to ferric carboxymaltose and iron dextran resulted in release of tumor necrosis factor α. CONCLUSIONS Exposure to iron compounds increased cell stress but was tissue and dose dependent. There was a clear difference in the handling of iron from the different compounds by macrophages that suggests in vivo responses may differ.
Collapse
Affiliation(s)
- James R. Connor
- Department of Neurosurgery, M.S. Hershey Penn State University College of Medicine, M.S. Hershey Medical Center, Hershey, Pennsylvania, United States of America
| | - Xuesheng Zhang
- Department of Neurosurgery, M.S. Hershey Penn State University College of Medicine, M.S. Hershey Medical Center, Hershey, Pennsylvania, United States of America
| | - Anne M. Nixon
- Department of Neurosurgery, M.S. Hershey Penn State University College of Medicine, M.S. Hershey Medical Center, Hershey, Pennsylvania, United States of America
| | - Becky Webb
- Department of Neurosurgery, M.S. Hershey Penn State University College of Medicine, M.S. Hershey Medical Center, Hershey, Pennsylvania, United States of America
| | - Joseph R. Perno
- Luitpold Pharmaceuticals, Inc., Norristown, Pennsylvania, United States of America
| |
Collapse
|
50
|
Peffer K, Verbeek ALM, Swinkels DW, Geurts-Moespot AJ, den Heijer M, Atsma F. Donation intensity and metabolic syndrome in active whole-blood donors. Vox Sang 2015; 109:25-34. [DOI: 10.1111/vox.12258] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2014] [Revised: 01/08/2015] [Accepted: 01/16/2015] [Indexed: 01/15/2023]
Affiliation(s)
- K. Peffer
- Department of Donor Studies; Sanquin Research; Nijmegen The Netherlands
- Department for Health Evidence; Radboud University Medical Center; Nijmegen The Netherlands
| | - A. L. M. Verbeek
- Department for Health Evidence; Radboud University Medical Center; Nijmegen The Netherlands
| | - D. W. Swinkels
- Laboratory of Genetic, Endocrine and Metabolic Diseases; Department of Laboratory Medicine; Radboud University Medical Center; Nijmegen The Netherlands
| | - A. J. Geurts-Moespot
- Laboratory of Genetic, Endocrine and Metabolic Diseases; Department of Laboratory Medicine; Radboud University Medical Center; Nijmegen The Netherlands
| | - M. den Heijer
- Department for Health Evidence; Radboud University Medical Center; Nijmegen The Netherlands
- Department of Internal Medicine; VU University Medical Center; Amsterdam The Netherlands
| | - F. Atsma
- Department of Donor Studies; Sanquin Research; Nijmegen The Netherlands
- Scientific Institute for Quality of Healthcare; Radboud University Medical Center; Nijmegen The Netherlands
| |
Collapse
|