101
|
Bhusal A, Rahman MH, Lee WH, Bae YC, Lee IK, Suk K. Paradoxical role of lipocalin-2 in metabolic disorders and neurological complications. Biochem Pharmacol 2019; 169:113626. [PMID: 31476294 DOI: 10.1016/j.bcp.2019.113626] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2019] [Accepted: 08/28/2019] [Indexed: 02/07/2023]
Abstract
Lipocalin-2 (LCN2), also known as 24p3 and neutrophil gelatinase-associated lipocalin (NGAL), is a 25-kDa secreted protein implicated in various metabolic and inflammatory diseases. Early studies suggest the protective function of LCN2 in which it acts as a bacteriostatic agent that competes with bacteria for iron-bound siderophores. However, both detrimental and beneficial roles of LCN2 have recently been documented in metabolic and neuroinflammatory diseases. Metabolic inflammation, as observed in diabetes and obesity, has been closely associated with the upregulation of LCN2 in blood plasma and several tissues in both humans and rodents, suggesting its pro-diabetic and pro-obesogenic role. On the contrary, other studies imply an anti-diabetic and anti-obesogenic role of LCN2 whereby a deficiency in the Lcn2 gene results in the impairment of insulin sensitivity and enhances the high-fat-diet-induced expansion of fat. A similar dual role of LCN2 has also been reported in various animal models for neurological disorders. In the midst of these mixed findings, there is no experimental evidence to explain why LCN2 shows such a contrasting role in the various studies. This debate needs to be resolved (or reconciled) and an integrated view on the topic is desirable. Herein, we attempt to address this issue by reviewing the recent findings on LCN2 in metabolic disorders and assess the potential cellular or molecular mechanisms underlying the dual role of LCN2. We further discuss the possibilities and challenges of targeting LCN2 as a potential therapeutic strategy for metabolic disorders and neurological complications.
Collapse
Affiliation(s)
- Anup Bhusal
- Department of Pharmacology, Brain Science & Engineering Institute, School of Medicine, Kyungpook National University, Daegu, Republic of Korea; Department of Biomedical Science, BK21 PLUS KNU Biomedical Convergence Program, School of Medicine, Kyungpook National University, Daegu, Republic of Korea
| | - Md Habibur Rahman
- Department of Pharmacology, Brain Science & Engineering Institute, School of Medicine, Kyungpook National University, Daegu, Republic of Korea; Department of Biomedical Science, BK21 PLUS KNU Biomedical Convergence Program, School of Medicine, Kyungpook National University, Daegu, Republic of Korea
| | - Won-Ha Lee
- School of Life Sciences, BK21 Plus KNU Creative BioResearch Group, Kyungpook National University, Daegu, Republic of Korea
| | - Yong Chul Bae
- Department of Anatomy and Neurobiology, School of Dentistry, Kyungpook National University, Daegu, Republic of Korea
| | - In-Kyu Lee
- Department of Internal Medicine, Division of Endocrinology and Metabolism, School of Medicine, Kyungpook National University, Daegu, Republic of Korea
| | - Kyoungho Suk
- Department of Pharmacology, Brain Science & Engineering Institute, School of Medicine, Kyungpook National University, Daegu, Republic of Korea; Department of Biomedical Science, BK21 PLUS KNU Biomedical Convergence Program, School of Medicine, Kyungpook National University, Daegu, Republic of Korea.
| |
Collapse
|
102
|
Gu Y, Su X, Li Y, Tang Y, Bao Y, Ying H. Do free thyroxine levels influence the relationship between maternal serum ferritin and gestational diabetes mellitus in early pregnancy? Diabetes Res Clin Pract 2019; 151:114-119. [PMID: 30935929 DOI: 10.1016/j.diabres.2019.03.033] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/31/2018] [Revised: 03/08/2019] [Accepted: 03/27/2019] [Indexed: 12/30/2022]
Abstract
PURPOSE The objective of this study was to estimate the combined effect of serum ferritin (SF) concentration and free thyroxine (fT4) levels on the risk of gestational diabetes mellitus (GDM). METHODS Women presented for antenatal care at a tertiary hospital in Shanghai, China were included in this study from December 2012 to March 2014. Women were divided into six groups according to the SF and fT4 level. Multiple logistical regression model was used to estimate odds ratio (OR) among different groups. Relative excess risk of interaction (RERI), the attributable proportion (AP) of the interaction and the synergy index (SI) were applied to evaluate the additive interaction of SF concentration and fT4 level. RESULTS A total of 6542 qualifying pregnant women were included in this study. We observed that a high SF concentration in early pregnancy was related to an increased risk of GDM (OR = 1.21, 95%CI: 1.02-1.43); while a low fT4 level was not (OR = 1.18, 95%CI: 0.89-1.58). There is no addictive interaction between SF and fT4 level on the presence of GDM. CONCLUSIONS The study suggests that only high serum ferritin concentration is associated with an increased risk of GDM in early pregnancy. The level of fT4 in early pregnancy might have no effect on the association between high SF and risk of GDM.
Collapse
Affiliation(s)
- Yanqiong Gu
- Department of Obstetrics, Shanghai First Maternity and Infant Hospital, Tongji University School of Medicine, Shanghai 201204, PR China.
| | - Xiujuan Su
- Department of Women & Children's Health Care, Shanghai First Maternity and Infant Hospital, Tongji University School of Medicine, Shanghai 201204, PR China.
| | - Yuhong Li
- Department of Obstetrics, Shanghai First Maternity and Infant Hospital, Tongji University School of Medicine, Shanghai 201204, PR China.
| | - Yuping Tang
- Department of Obstetrics, Shanghai First Maternity and Infant Hospital, Tongji University School of Medicine, Shanghai 201204, PR China
| | - Yirong Bao
- Department of Obstetrics, Shanghai First Maternity and Infant Hospital, Tongji University School of Medicine, Shanghai 201204, PR China.
| | - Hao Ying
- Department of Obstetrics, Shanghai First Maternity and Infant Hospital, Tongji University School of Medicine, Shanghai 201204, PR China.
| |
Collapse
|
103
|
Suárez-Ortegón MF, Echeverri I, Prats-Puig A, Bassols J, Carreras-Badosa G, López-Bermejo A, Fernández-Real JM. Iron Status and Metabolically Unhealthy Obesity in Prepubertal Children. Obesity (Silver Spring) 2019; 27:636-644. [PMID: 30821086 DOI: 10.1002/oby.22425] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/05/2018] [Accepted: 12/10/2018] [Indexed: 01/19/2023]
Abstract
OBJECTIVE This study aimed to evaluate the association of metabolically healthy obesity (MHO) and metabolically unhealthy obesity (MUO) with iron status markers in prepubertal children. METHODS Three hundred twelve prepubertal children with overweight and obesity from a pediatric general Spanish population were evaluated. MHO and MUO were defined as obesity with the absence or presence of metabolic syndrome components. Phenotypes of metabolically healthy overweight including obesity (MHOV) and metabolically unhealthy overweight including obesity (MUOV) were also studied and defined using the same criteria. Serum ferritin, transferrin, and blood hemoglobin levels were evaluated. RESULTS Prevalence rates of MHOV and MHO were 35% (n = 111/312) and 27.1% (n = 42/155), respectively. Ferritin and hemoglobin levels were higher in children with MUOV versus MHOV (P < 0.05). MUO was positively associated with ferritin (beta [95% CI] = 0.43 [0.05 to 0.81]) and hemoglobin levels (0.43 [0.05 to 0.81]). These associations remained significant independently of age, sex, C-reactive protein, physical activity, and BMI/waist z scores in bivariate linear regression models. In multivariable models, transaminase levels attenuated the association of MUO with ferritin and hemoglobin levels (P > 0.05). CONCLUSIONS MUOV and MUO are associated with higher ferritin and hemoglobin levels in prepubertal children affected by overweight and obesity. Increased circulating ferritin in MUO might be influenced by liver injury.
Collapse
Affiliation(s)
- Milton Fabian Suárez-Ortegón
- Basic and Clinic Sciences Group-Department of Basic Sciences of Health, Pontificia Universidad Javeriana, Cali, Colombia
- Nutrition Group, University of Valle, Cali, Colombia
| | | | - Anna Prats-Puig
- Pediatrics Research Group, Girona Institute for Biomedical Research, Girona, Spain
- Department of Pediatrics, Dr Josep Trueta Hospital, Girona, Spain
- Department of Physical Therapy, University School of Health and Sports, University of Girona, Girona, Spain
| | - Judit Bassols
- Pediatrics Research Group, Girona Institute for Biomedical Research, Girona, Spain
- Department of Pediatrics, Dr Josep Trueta Hospital, Girona, Spain
| | - Gemma Carreras-Badosa
- Pediatrics Research Group, Girona Institute for Biomedical Research, Girona, Spain
- Department of Pediatrics, Dr Josep Trueta Hospital, Girona, Spain
| | - Abel López-Bermejo
- Pediatrics Research Group, Girona Institute for Biomedical Research, Girona, Spain
- Department of Pediatrics, Dr Josep Trueta Hospital, Girona, Spain
- TransLab Research Group, Department of Medical Sciences, Faculty of Medicine, University of Girona, Girona, Spain
| | - Jose Manuel Fernández-Real
- Department of Diabetes, Endocrinology and Nutrition, Department of Medical Sciences, Faculty of Medicine, Girona Institute for Biomedical Research, Spanish Biomedical Research Center in Physiopathology of Obesity and Nutrition and National Institute of Health Carlos III, Girona, Spain
| |
Collapse
|
104
|
Grammer TB, Scharnagl H, Dressel A, Kleber ME, Silbernagel G, Pilz S, Tomaschitz A, Koenig W, Mueller-Myhsok B, März W, Strnad P. Iron Metabolism, Hepcidin, and Mortality (the Ludwigshafen Risk and Cardiovascular Health Study). Clin Chem 2019; 65:849-861. [PMID: 30917972 DOI: 10.1373/clinchem.2018.297242] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2018] [Accepted: 02/21/2019] [Indexed: 12/16/2022]
Abstract
BACKGROUND Anemia has been shown to be a risk factor for coronary artery disease (CAD) and mortality, whereas the role of iron metabolism remains controversial. METHODS We analyzed iron metabolism and its associations with cardiovascular death and total mortality in patients undergoing coronary angiography with a median follow-up of 9.9 years. Hemoglobin and iron status were determined in 1480 patients with stable CAD and in 682 individuals in whom significant CAD had been excluded by angiography. RESULTS Multivariate-adjusted hazard ratios (HRs) for total mortality in the lowest quartiles of iron, transferrin saturation, ferritin, soluble transferrin receptor (sTfR), and hemoglobin were 1.22 (95% CI, 0.96-1.60), 1.23 (95% CI, 0.97-1.56), 1.27 (95% CI, 1.02-1.58), 1.26 (95% CI, 0.97-1.65), and 0.99 (95% CI, 0.79-1.24), respectively, compared to the second or third quartile, which served as reference (1.00) because of a J-shaped association. The corresponding HRs for total mortality in the highest quartiles were 1.44 (95% CI, 1.10-1.87), 1.37 (95% CI, 1.05-1.77), 1.17 (95% CI, 0.92-1.50), 1.76 (95% CI, 1.39-2.22), and 0.83 (95% CI, 0.63-1.09). HRs for cardiovascular death were similar. For hepcidin, the adjusted HRs for total mortality and cardiovascular deaths were 0.62 (95% CI, 0.49-0.78) and 0.70 (95% CI, 0.52-0.90) in the highest quartile compared to the lowest one. CONCLUSIONS In stable patients undergoing angiography, serum iron, transferrin saturation, sTfR, and ferritin had J-shaped associations and hemoglobin only a marginal association with cardiovascular and total mortality. Hepcidin was continuously and inversely related to mortality.
Collapse
Affiliation(s)
- Tanja B Grammer
- Mannheim Institute of Public Health, Social and Preventive Medicine, Mannheim Medical Faculty, University of Heidelberg, Mannheim, Germany; .,Department of Internal Medicine V (Nephrology, Hypertensiology, Endocrinology, Diabetolgy, and Rheumatology), Mannheim Medical Faculty, University of Heidelberg, Mannheim, Germany
| | - Hubert Scharnagl
- Clinical Institute of Medical and Chemical Laboratory Diagnostics, Medical University of Graz, Graz, Austria
| | - Alexander Dressel
- DACH Society for the Prevention of Cardiovascular Diseases, Hamburg, Germany
| | - Marcus E Kleber
- Department of Internal Medicine V (Nephrology, Hypertensiology, Endocrinology, Diabetolgy, and Rheumatology), Mannheim Medical Faculty, University of Heidelberg, Mannheim, Germany
| | - Günther Silbernagel
- Division of Angiology, Department of Internal Medicine, Medical University of Graz, Graz, Austria.,Department of Cardiology, Charité Berlin, Berlin Institute of Health and German Research Centre for Cardiovascular Research, Berlin, Germany
| | - Stefan Pilz
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Medical University of Graz, Graz, Austria
| | | | - Wolfgang Koenig
- Deutsches Herzzentrum München, Technische Universität München, Munich, Germany.,DZHK (German Centre for Cardiovascular Research), partner site Munich Heart Alliance, Munich, Germany
| | - Bertram Mueller-Myhsok
- Max Planck Institute of Psychiatry, Munich, Germany.,Munich Cluster for Systems Neurology (SyNergy), Munich, Germany.,Institute of Translational Medicine, University of Liverpool, Liverpool, UK
| | - Winfried März
- Department of Internal Medicine V (Nephrology, Hypertensiology, Endocrinology, Diabetolgy, and Rheumatology), Mannheim Medical Faculty, University of Heidelberg, Mannheim, Germany.,Clinical Institute of Medical and Chemical Laboratory Diagnostics, Medical University of Graz, Graz, Austria.,Synlab Academy, Synlab Holding Deutschland GmbH, Augsburg and Mannheim, Germany
| | - Pavel Strnad
- Department of Internal Medicine III and IZKF, University Hospital Aaachen, Aachen, Germany
| |
Collapse
|
105
|
Shu T, Lv Z, Xie Y, Tang J, Mao X. Hepcidin as a key iron regulator mediates glucotoxicity-induced pancreatic β-cell dysfunction. Endocr Connect 2019; 8:150-161. [PMID: 30776286 PMCID: PMC6391907 DOI: 10.1530/ec-18-0516] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/01/2019] [Accepted: 01/21/2019] [Indexed: 02/06/2023]
Abstract
It has been well established that glucotoxicity induces pancreatic β-cells dysfunction; however, the precise mechanism remains unclear. Our previous studies demonstrated that high glucose concentrations are associated with decreased hepcidin expression, which inhibits insulin synthesis. In this study, we focused on the role of low hepcidin level-induced increased iron deposition in β-cells and the relationship between abnormal iron metabolism and β-cell dysfunction. Decreased hepcidin expression increased iron absorption by upregulating transferrin receptor 1 (TfR1) and divalent metal transporter 1 (DMT1) expression, resulting in iron accumulation within cells. Prussia blue stain and calcein-AM assays revealed greater iron accumulation in the cytoplasm of pancreatic tissue isolated from db/db mice, cultured islets and Min6 cells in response to high glucose stimulation. Increased cytosolic iron deposition was associated with greater Fe2+ influx into the mitochondria, which depolarized the mitochondria membrane potential, inhibited ATP synthesis, generated excessive ROS and induced oxidative stress. The toxic effect of excessive iron on mitochondrial function eventually resulted in impaired insulin secretion. The restricted iron content in db/db mice via reduced iron intake or accelerated iron clearance improved blood glucose levels with decreased fasting blood glucose (FBG), fasting blood insulin (FIns), HbA1c level, as well as improved intraperitoneal glucose tolerance test (IPGTT) results. Thus, our study may reveal the mechanism involved in the role of hepcidin in the glucotoxcity impaired pancreatic β cell function pathway.
Collapse
Affiliation(s)
- Tingting Shu
- Department of Central Laboratory, Jiangsu Province Official Hospital, Nanjing, Jiangsu, China
| | - Zhigang Lv
- Department of Central Laboratory, Jiangsu Province Official Hospital, Nanjing, Jiangsu, China
| | - Yuchun Xie
- Department of Central Laboratory, Jiangsu Province Official Hospital, Nanjing, Jiangsu, China
| | - Junming Tang
- Department of Clinical Laboratory, Yixing People Hospital, Affiliated Jiangsu University, Yixing, Wuxi, Jiangsu, China
| | - Xuhua Mao
- Department of Clinical Laboratory, Yixing People Hospital, Affiliated Jiangsu University, Yixing, Wuxi, Jiangsu, China
- Correspondence should be addressed to X Mao:
| |
Collapse
|
106
|
Suárez-Ortegón MF, Blanco E, McLachlan S, Fernandez-Real JM, Burrows R, Wild SH, Lozoff B, Gahagan S. Ferritin levels throughout childhood and metabolic syndrome in adolescent stage. Nutr Metab Cardiovasc Dis 2019; 29:268-278. [PMID: 30648600 PMCID: PMC6758555 DOI: 10.1016/j.numecd.2018.11.008] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/27/2018] [Revised: 10/10/2018] [Accepted: 11/26/2018] [Indexed: 12/12/2022]
Abstract
BACKGROUND AND AIM Increased ferritin levels have been widely associated with cardiovascular risk in adults. Whether ferritin levels and their changes during childhood are related to metabolic syndrome (MetS) at adolescence is unknown. We aimed to evaluate these associations using levels of ferritin at 5, 10 and 16 years and their linear increases and patterns of sustained increased levels across childhood. METHODS AND RESULTS There were four samples evaluated according to non-missing values for study variables at each stage (5 years: 562; 10 years: 381; and 16 years: 567 children; non-missing values at any stage: 379). MetS risk was evaluated as a continuous Z score. Patterns of sustained increased ferritin (highest tertile) and slope of the change of ferritin per year across the follow-up were calculated. Ferritin levels in the highest versus lowest tertile at five and 16 years were significantly positively associated with MetS risk Z score at adolescence in boys and these associations were unaffected by adjustment for covariates. Having high, compared to low/moderate ferritin level at 2 or more time periods between 5 and 16 years was related to higher Mets Z-score in boys only [e.g. 5-10 years adjusted-beta (95 %CI):0.26 (0.05-0.48),P < 0.05]. In girls, ferritin Z score at 10 and 16 years was positively and independently associated with HOMA-IR Z score. In girls, the slope of ferritin per year in the highest tertile was positively associated with MetS risk Z-score [adjusted-beta (95 %CI):0.21 (0.05-0.38),P < 0.05]. CONCLUSIONS Ferritin levels throughout childhood are positively related to cardiometabolic risk in adolescence, with associations varying by sex.
Collapse
Affiliation(s)
- M F Suárez-Ortegón
- Basic and Clinic Sciences Group-Department of Basic Sciences of Health, Pontificia Universidad Javeriana, Cali, Colombia; Nutrition Group, Universidad del Valle, Cali, Colombia.
| | - E Blanco
- Division of Child Development and Community Health, University of California, San Diego, La Jolla, CA, USA
| | - S McLachlan
- Basic and Clinic Sciences Group-Department of Basic Sciences of Health, Pontificia Universidad Javeriana, Cali, Colombia
| | - J M Fernandez-Real
- Department of Diabetes, Endocrinology and Nutrition, Institut d'Investigació Biomèdica de Girona (IdIBGi), CIBEROBN (CB06/03/010) and Instituto de Salud Carlos III (ISCIII), Girona, Spain
| | - R Burrows
- University of Chile, Santiago, Chile
| | - S H Wild
- Basic and Clinic Sciences Group-Department of Basic Sciences of Health, Pontificia Universidad Javeriana, Cali, Colombia
| | - B Lozoff
- Center for Human Growth and Development, University of Michigan, Ann Arbor, MI, USA
| | - S Gahagan
- Division of Child Development and Community Health, University of California, San Diego, La Jolla, CA, USA
| |
Collapse
|
107
|
Zhao L, Bartnikas T, Chu X, Klein J, Yun C, Srinivasan S, He P. Hyperglycemia promotes microvillus membrane expression of DMT1 in intestinal epithelial cells in a PKCα-dependent manner. FASEB J 2019; 33:3549-3561. [PMID: 30423260 PMCID: PMC6404579 DOI: 10.1096/fj.201801855r] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2018] [Accepted: 10/15/2018] [Indexed: 01/24/2023]
Abstract
Excessive iron increases the incidence of diabetes and worsens diabetic complications. Reciprocally, diabetes induces iron loading, partially attributable to elevated intestinal iron export according to a recent report. Herein, we show that iron uptake and the mRNA expression of iron importer divalent metal transporter 1 (DMT1) were significantly increased in the duodenum of streptozotocin-induced diabetic mice. Immunofluorescence staining of human intestinal biopsies revealed increased brush border membrane (BBM) and decreased cytoplasmic DMT1 expression in patients with diabetes, suggesting translocation of DMT1. This pattern of DMT1 regulation was corroborated by immunoblotting results in diabetic mice showing that BBM DMT1 expression was increased by 210%, in contrast to a 60% increase in total DMT1. PKC mediates many diabetic complications, and PKCα activity was increased in diabetic mouse intestine. Intriguingly, diabetic mice with PKCα deficiency did not show increases in iron uptake and BBM DMT1 expression. High-glucose treatment increased plasma membrane DMT1 expression via the activation of PKCα in cultured IECs. Inhibition of PKCα potentiated the ubiquitination and degradation of DMT1 protein. We further showed that high glucose suppressed membrane DMT1 internalization. These findings demonstrate that PKCα promotes microvillus membrane DMT1 expression and intestinal iron uptake, contributing to diabetic iron loading.-Zhao, L., Bartnikas, T., Chu, X., Klein, J., Yun, C., Srinivasan, S., He, P. Hyperglycemia promotes microvillus membrane expression of DMT1 in intestinal epithelial cells in a PKCα-dependent manner.
Collapse
Affiliation(s)
- Luqing Zhao
- Department of Gastroenterology, Beijing Hospital of Traditional Chinese Medicine Affiliated With Capital Medical University, Beijing, China
- Division of Digestive Diseases, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Thomas Bartnikas
- Department of Pathology and Laboratory Medicine, Brown University, Providence, Rhode Island, USA
| | - Xiangpeng Chu
- Department of Thoracic Surgery, People’s Hospital of Rizhao, Shandong, China
| | - Janet Klein
- Division of Renal Medicine, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia, USA; and
| | - Chris Yun
- Division of Digestive Diseases, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia, USA
- Atlanta Veterans Administration Medical Center, Decatur, Georgia, USA
| | - Shanthi Srinivasan
- Division of Digestive Diseases, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia, USA
- Atlanta Veterans Administration Medical Center, Decatur, Georgia, USA
| | - Peijian He
- Division of Digestive Diseases, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia, USA
| |
Collapse
|
108
|
Looman M, Geelen A, Samlal RAK, Heijligenberg R, Klein Gunnewiek JMT, Balvers MGJ, Wijnberger LDE, Brouwer-Brolsma EM, Feskens EJM. Changes in Micronutrient Intake and Status, Diet Quality and Glucose Tolerance from Preconception to the Second Trimester of Pregnancy. Nutrients 2019; 11:nu11020460. [PMID: 30813281 PMCID: PMC6412670 DOI: 10.3390/nu11020460] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2018] [Revised: 02/10/2019] [Accepted: 02/19/2019] [Indexed: 12/14/2022] Open
Abstract
Data on changes in dietary intake and related blood parameters throughout pregnancy are scarce; moreover, few studies have examined their association with glucose homeostasis. Therefore, we monitored intake of folate, vitamin B6, vitamin B12, vitamin D and iron, their status markers, and diet quality from preconception to the second trimester of pregnancy, and we examined whether these dietary factors were associated with glucose homeostasis during pregnancy. We included 105 women aged 18⁻40 years with a desire to get pregnancy or who were already <24 weeks pregnant. Women at increased gestational diabetes (GDM) risk were oversampled. Measurements were scheduled at preconception (n = 67), and 12 (n =53) and 24 weeks of pregnancy (n =66), including a fasting venipuncture, 75-grams oral glucose tolerance test, and completion of a validated food frequency questionnaire. Changes in micronutrient intake and status, and associations between dietary factors and glucose homeostasis, were examined using adjusted repeated measures mixed models. Micronutrient intake of folate, vitamin B6 and vitamin D and related status markers significantly changed throughout pregnancy, which was predominantly due to changes in the intake of supplements. Micronutrient intake or status levels were not associated with glucose homeostasis, except for iron intake (FE µg/day) with fasting glucose (β = -0.069 mmol/L, p = 0.013) and HbA1c (β = -0.4843 mmol, p = 0.002). Diet quality was inversely associated with fasting glucose (β = -0.006 mmol/L for each DHD15-index point, p = 0.017). It was shown that micronutrient intakes and their status markers significantly changed during pregnancy. Only iron intake and diet quality were inversely associated with glucose homeostasis.
Collapse
Affiliation(s)
- Moniek Looman
- Division of Human Nutrition, Wageningen University, P.O. Box 17, 6700 AA Wageningen, The Netherlands.
| | - Anouk Geelen
- Division of Human Nutrition, Wageningen University, P.O. Box 17, 6700 AA Wageningen, The Netherlands.
| | - Rahul A K Samlal
- Department of Gynaecology and Obstetrics, Hospital Gelderse Vallei Ede, P.O. Box 9025, 6710 HN Ede, The Netherlands.
| | - Rik Heijligenberg
- Department of Internal Medicine, Hospital Gelderse Vallei Ede, P.O. Box 9025, 6710 HN Ede, The Netherlands.
| | - Jacqueline M T Klein Gunnewiek
- Clinical Chemistry and Haematology Laboratory, Hospital Gelderse Vallei Ede, P.O. Box 9025, 6710 HN Ede, The Netherlands.
| | - Michiel G J Balvers
- Clinical Chemistry and Haematology Laboratory, Hospital Gelderse Vallei Ede, P.O. Box 9025, 6710 HN Ede, The Netherlands.
| | - Lia D E Wijnberger
- Department of Obstetrics and Gynaecology, Rijnstate Hospital, P.O. Box 9555, 6800 TA Arnhem, The Netherlands.
| | - Elske M Brouwer-Brolsma
- Division of Human Nutrition, Wageningen University, P.O. Box 17, 6700 AA Wageningen, The Netherlands.
| | - Edith J M Feskens
- Division of Human Nutrition, Wageningen University, P.O. Box 17, 6700 AA Wageningen, The Netherlands.
| |
Collapse
|
109
|
Gao Y, Liu J, Bai Z, Sink S, Zhao C, Lorenzo FR, McClain DA. Iron down-regulates leptin by suppressing protein O-GlcNAc modification in adipocytes, resulting in decreased levels of O-glycosylated CREB. J Biol Chem 2019; 294:5487-5495. [PMID: 30709903 DOI: 10.1074/jbc.ra118.005183] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2018] [Revised: 01/28/2019] [Indexed: 11/06/2022] Open
Abstract
We previously reported that iron down-regulates transcription of the leptin gene by increasing occupancy of phosphorylated cAMP response element-binding protein (pCREB) at two sites in the leptin gene promoter. Several nutrient-sensing pathways including O-GlcNAcylation also regulate leptin. We therefore investigated whether O-glycosylation plays a role in iron- and CREB-mediated regulation of leptin. We found that high iron decreases protein O-GlcNAcylation both in cultured 3T3-L1 adipocytes and in mice fed high-iron diets and down-regulates leptin mRNA and protein levels. Glucosamine treatment, which bypasses the rate-limiting step in the synthesis of substrate for glycosylation, increased both O-GlcNAc and leptin, whereas inhibition of O-glycosyltransferase (OGT) decreased O-GlcNAc and leptin. The increased leptin levels induced by glucosamine were susceptible to the inhibition by iron, but in the case of OGT inhibition, iron did not further decrease leptin. Mice with deletion of the O-GlcNAcase gene, either via whole-body heterozygous deletion or through adipocyte-targeted homozygous deletion, exhibited increased O-GlcNAc levels in adipose tissue and increased leptin levels that were inhibited by iron. Of note, iron increased the occupancy of pCREB and decreased the occupancy of O-GlcNAcylated CREB on the leptin promoter. These patterns observed in our experimental models suggest that iron exerts its effects on leptin by decreasing O-glycosylation and not by increasing protein deglycosylation and that neither O-GlcNAcase nor OGT mRNA and protein levels are affected by iron. We conclude that iron down-regulates leptin by decreasing CREB glycosylation, resulting in increased CREB phosphorylation and leptin promoter occupancy by pCREB.
Collapse
Affiliation(s)
- Yan Gao
- From the Department of Internal Medicine, Wake Forest School of Medicine, Winston Salem, North Carolina 27157 and
| | - Jingfang Liu
- From the Department of Internal Medicine, Wake Forest School of Medicine, Winston Salem, North Carolina 27157 and
| | - Zhenzhong Bai
- From the Department of Internal Medicine, Wake Forest School of Medicine, Winston Salem, North Carolina 27157 and
| | - Sandy Sink
- From the Department of Internal Medicine, Wake Forest School of Medicine, Winston Salem, North Carolina 27157 and
| | - Chengyu Zhao
- From the Department of Internal Medicine, Wake Forest School of Medicine, Winston Salem, North Carolina 27157 and
| | - Felipe Ramos Lorenzo
- From the Department of Internal Medicine, Wake Forest School of Medicine, Winston Salem, North Carolina 27157 and
| | - Donald A McClain
- From the Department of Internal Medicine, Wake Forest School of Medicine, Winston Salem, North Carolina 27157 and .,the W. G. Hefner Veterans Affairs Medical Center, Salisbury, North Carolina 28144
| |
Collapse
|
110
|
Iron regulatory protein 2 deficiency may correlate with insulin resistance. Biochem Biophys Res Commun 2019; 510:191-197. [PMID: 30685084 DOI: 10.1016/j.bbrc.2019.01.022] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2018] [Accepted: 01/05/2019] [Indexed: 12/12/2022]
Abstract
Iron is known to be a crucial regulator of glucose, and several studies have demonstrated that iron overload is one of the risk factors for insulin resistance and diabetes; however, the mechanism has not yet been clarified. To investigate the effect of iron overload on glucose metabolism and the underlying mechanism, Irp2 knockout (Irp2-/-) mice (endogenous iron overload model) were used. We found that Irp2-/- mice exhibited hyperglycemia and iron overload in the liver and skeletal muscle. Increased MDA, decreased SOD levels, and increased cell apoptosis were also found in the liver and muscle of Irp2-/- mice. Glucose concentrations were significantly higher in Irp2-/- mice in insulin tolerance tests. However, early-phase insulin secretion was not altered in Irp2-/- mice. The expression of hepatic IRS2 and muscle GLUT4 was declined in Irp2-/- mice at both mRNA and protein levels when compared with those of wild-type control. In conclusions, Irp2-/- mice showed hyperglycemia, which might due to insulin resistance rather than due to impaired insulin secretion.
Collapse
|
111
|
Kumar TU, Pawar S, Nag A, Bhattacharya A. Selective Sensing of Iron by Pyrrolo[2,3-c]Quinolines. J Fluoresc 2019; 29:271-277. [PMID: 30623284 DOI: 10.1007/s10895-018-02337-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2018] [Accepted: 12/26/2018] [Indexed: 10/27/2022]
Abstract
This paper reports development of an iron sensor, 2-(3H-pyrrolo[2,3-c]quinolin-4-yl)aniline (APQ). The fluorophore facilitates micromolar detection of Fe3+/Fe2+ in the presence of various cations, including well-known interfering cations Co2+and Cu2+ by the process of fluorescence quenching. Graphical Abstract.
Collapse
Affiliation(s)
- Togiti Uday Kumar
- Department of Chemistry, Birla Institute of Technology and Science-Pilani, Hyderabad Campus, Hyderabad, 500078, India
| | - Shweta Pawar
- Department of Chemistry, Birla Institute of Technology and Science-Pilani, Hyderabad Campus, Hyderabad, 500078, India
| | - Amit Nag
- Department of Chemistry, Birla Institute of Technology and Science-Pilani, Hyderabad Campus, Hyderabad, 500078, India
| | - Anupam Bhattacharya
- Department of Chemistry, Birla Institute of Technology and Science-Pilani, Hyderabad Campus, Hyderabad, 500078, India.
| |
Collapse
|
112
|
Misra S, Wai Yew Y, Seok Shin T. Maternal dietary patterns, diet quality and micronutrient status in gestational diabetes mellitus across different economies: A review. AIMS MEDICAL SCIENCE 2019. [DOI: 10.3934/medsci.2019.1.76] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
|
113
|
Gamede M, Mabuza L, Ngubane P, Khathi A. Plant-derived oleanolic acid ameliorates markers associated with non-alcoholic fatty liver disease in a diet-induced pre-diabetes rat model. Diabetes Metab Syndr Obes 2019; 12:1953-1962. [PMID: 31632109 PMCID: PMC6778448 DOI: 10.2147/dmso.s218626] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/07/2019] [Accepted: 07/24/2019] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND The increased prevalence of non-alcoholic fatty liver disease (NAFLD) in type 2 diabetes mellitus (T2DM) patients is becoming a worldwide health burden. Studies have indicated, however, that the onset of NAFLD occurs during pre-diabetes, a condition that often precedes the onset of T2DM. Oleanolic acid has been reported to improve glucose homeostasis in diet-induced pre-diabetes; however, the effects of this triterpene on liver function have not been evaluated. PURPOSE This study was aimed at evaluating the therapeutic effects of oleanolic acid (OA) on selected markers of NAFLD in a pre-diabetes rat model. METHODS AND MATERIALS Pre-diabetes was induced by exposing Sprague Dawley rats to a high-fat high-carbohydrate diet for 20 weeks. The pre-diabetic rats were then treated with OA (80 mg/kg) or metformin (500 mg/kg) in the presence and absence of dietary interventions for a period of 12 weeks. The effects of OA were evaluated on parameters including plasma triglycerides (TGs), very low-density lipoprotein (VLDL) particles, bilirubin, AST, ALT, SREBP and antioxidant profile while the livers were collected for histological analysis. RESULTS The findings of this study showed that the administration of OA to pre-diabetic rats ameliorated body/liver weights ratio and significantly decreased plasma triglycerides (TGs) and VLDL. Furthermore, OA also ameliorated hepatic oxidative stress, lowered the SREBP expression and intrahepatic TGs. In addition, OA administration decreased plasma concentrations of bilirubin and liver damage enzyme biomarkers. CONCLUSION The findings of the study suggest that OA ameliorates the risk of developing pre-diabetes-related NAFLD through the prevention of intrahepatic fat accumulation while also lowering hepatic inflammation.
Collapse
Affiliation(s)
- Mlindeli Gamede
- School of Laboratory Medicine and Medical Sciences, College of Health Sciences, University of KwaZulu-Natal, Durban, South Africa
| | - Lindokuhle Mabuza
- School of Laboratory Medicine and Medical Sciences, College of Health Sciences, University of KwaZulu-Natal, Durban, South Africa
| | - Phikelelani Ngubane
- School of Laboratory Medicine and Medical Sciences, College of Health Sciences, University of KwaZulu-Natal, Durban, South Africa
| | - Andile Khathi
- School of Laboratory Medicine and Medical Sciences, College of Health Sciences, University of KwaZulu-Natal, Durban, South Africa
- Correspondence: Andile KhathiDepartment of Human Physiology, School of Laboratory Medicine & Medical Sciences, University of KwaZulu-Natal, Room E3-408, Durban, South AfricaTel +27 31 260 7585Fax +27 31 260 7132Email
| |
Collapse
|
114
|
McClain DA, Sharma NK, Jain S, Harrison A, Salaye LN, Comeau ME, Langefeld CD, Lorenzo FR, Das SK. Adipose Tissue Transferrin and Insulin Resistance. J Clin Endocrinol Metab 2018; 103:4197-4208. [PMID: 30099506 PMCID: PMC6194856 DOI: 10.1210/jc.2018-00770] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/10/2018] [Accepted: 08/01/2018] [Indexed: 12/27/2022]
Abstract
Context Excessive body iron stores are a risk factor for decreased insulin sensitivity (SI) and diabetes. We hypothesized that transcriptional dysregulation of genes involved in iron metabolism in adipocytes causes insulin resistance. Objective and Design To define the genetic regulation of iron metabolism and its role in SI, we used gene expression, genotype, and SI data from an African American cohort (N = 256). Replication studies were performed in independent European ancestry cohorts. In vitro studies in human adipocytes were performed to define the role of a selected gene in causing insulin resistance. Results Among 62 transcripts representing iron homeostasis genes, expression of 30 in adipose tissue were correlated with SI. Transferrin (TF) and ferritin heavy polypeptide were most positively and negatively associated with SI, respectively. These observations were replicated in two independent European ancestry adipose data sets. The strongest cis-regulatory variant for TF expression (rs6785596; P = 7.84 × 10-18) was identified in adipose but not muscle or liver tissue. Variants significantly affected the normal relationship of serum ferritin to insulin resistance. Knockdown of TF in differentiated Simpson-Golabi-Behmel syndrome adipocytes by short hairpin RNA decreased intracellular iron, reduced maximal insulin-stimulated glucose uptake, and reduced Akt phosphorylation. Knockdown of TF caused differential expression of 465 genes, including genes involved in glucose transport, mitochondrial function, Wnt-pathway/ SI, chemokine activity, and obesity. Iron chelation recapitulated key changes in the expression profile induced by TF knockdown. Conclusion Genetic regulation of TF expression in adipose tissue plays a novel role in regulating SI.
Collapse
Affiliation(s)
- Donald A McClain
- Department of Internal Medicine, Wake Forest School of Medicine, Winston-Salem, North Carolina
- W. G. (Bill) Hefner VA Medical Center - Salisbury, Salisbury, North Carolina
| | - Neeraj K Sharma
- Department of Internal Medicine, Wake Forest School of Medicine, Winston-Salem, North Carolina
| | - Shalini Jain
- Department of Internal Medicine, Wake Forest School of Medicine, Winston-Salem, North Carolina
| | - Alexandria Harrison
- Department of Internal Medicine, Wake Forest School of Medicine, Winston-Salem, North Carolina
| | - Lipika N Salaye
- Department of Internal Medicine, Wake Forest School of Medicine, Winston-Salem, North Carolina
| | - Mary E Comeau
- Department of Biostatistical Sciences, Division of Public Health Sciences, Wake Forest School of Medicine, Winston-Salem, North Carolina
| | - Carl D Langefeld
- Department of Biostatistical Sciences, Division of Public Health Sciences, Wake Forest School of Medicine, Winston-Salem, North Carolina
| | - Felipe R Lorenzo
- Department of Internal Medicine, Wake Forest School of Medicine, Winston-Salem, North Carolina
- W. G. (Bill) Hefner VA Medical Center - Salisbury, Salisbury, North Carolina
| | - Swapan K Das
- Department of Internal Medicine, Wake Forest School of Medicine, Winston-Salem, North Carolina
| |
Collapse
|
115
|
Carlos AR, Weis S, Soares MP. Cross-Talk Between Iron and Glucose Metabolism in the Establishment of Disease Tolerance. Front Immunol 2018; 9:2498. [PMID: 30425714 PMCID: PMC6218924 DOI: 10.3389/fimmu.2018.02498] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2018] [Accepted: 10/10/2018] [Indexed: 12/13/2022] Open
Abstract
Infectious diseases are associated with disruption of host homeostasis. This can be triggered directly by pathogens or indirectly by host immune-driven resistance mechanisms. Disease tolerance is a defense strategy against infection that sustains host homeostasis, without exerting a direct negative impact on pathogens. The mechanisms governing disease tolerance encompass host metabolic responses that maintain vital homeostatic parameters within a range compatible with survival. Central to this defense strategy is the host's ability to sense and adapt to variations in nutrients, such as iron and glucose. Here we address how host responses regulating iron and glucose metabolism interact to establish disease tolerance and possibly modulate resistance to infection.
Collapse
Affiliation(s)
| | - Sebastian Weis
- Department of Anesthesiology and Intensive Care Medicine, Jena University Hospital, Jena, Germany.,Institute for Infectious Disease and Infection Control, Jena University Hospital, Jena, Germany.,Center for Sepsis Control and Care, Jena University Hospital, Jena, Germany
| | | |
Collapse
|
116
|
Ryan BJ, Van Pelt DW, Guth LM, Ludzki AC, Gioscia-Ryan RA, Ahn C, Foug KL, Horowitz JF. Plasma ferritin concentration is positively associated with in vivo fatty acid mobilization and insulin resistance in obese women. Exp Physiol 2018; 103:1443-1447. [PMID: 30178895 DOI: 10.1113/ep087283] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2018] [Accepted: 08/31/2018] [Indexed: 01/27/2023]
Abstract
NEW FINDINGS What is the central question of this study? Do obese women with relatively high whole-body iron stores exhibit elevated in vivo rates of fatty acid (FA) release from adipose tissue compared with a well-matched cohort of obese women with relatively low iron stores? What is the main finding and its importance? Obese women with high plasma [ferritin] (a marker of whole-body iron stores) had greater FA mobilization, lipolytic activation in adipose tissue and insulin resistance (IR) compared with obese women with lower plasma [ferritin]. Given that elevated FA mobilization is intimately linked with the development of IR, these findings suggest that elevated iron stores might contribute to IR in obesity by increasing systemic FA availability. ABSTRACT High rates of fatty acid (FA) mobilization from adipose tissue are associated with insulin resistance (IR) in obesity. In vitro evidence suggests that iron stimulates lipolysis in adipocytes, but whether iron is related to in vivo FA mobilization is unknown. We hypothesized that plasma ferritin concentration ([ferritin]), a marker of body iron stores, would be positively associated with FA mobilization. We measured [ferritin], the rate of appearance of FA in the systemic circulation (FA Ra; stable isotope dilution), key adipose tissue lipolytic proteins and IR (hyperinsulinaemic-euglycaemic clamp) in 20 obese, premenopausal women. [Ferritin] was correlated with FA Ra (r = 0.65; P = 0.002) and IR (r = 0.57; P = 0.008); these relationships remained significant after controlling for body mass index and plasma [C-reactive protein] (a marker of systemic inflammation) in multiple regression analyses. We then stratified subjects into tertiles based on [ferritin] to compare subjects with 'High-ferritin' versus 'Low-ferritin'. Plasma [hepcidin] was more than fivefold greater (P < 0.05) in the High-ferritin versus Low-ferritin group, but there was no difference in plasma [C-reactive protein] between groups, indicating that the large difference in plasma [ferritin] reflects a difference in iron stores, not systemic inflammation. We found that FA Ra, adipose protein abundance of hormone-sensitive lipase and adipose triglyceride lipase, and IR were significantly greater in subjects with High-ferritin versus Low-ferritin (all P < 0.05). These data provide the first evidence linking iron and in vivo FA mobilization and suggest that elevated iron stores might contribute to IR in obesity by increasing systemic FA availability.
Collapse
Affiliation(s)
- Benjamin J Ryan
- Substrate Metabolism Laboratory, School of Kinesiology, University of Michigan, Ann Arbor, MI, USA
| | - Douglas W Van Pelt
- Substrate Metabolism Laboratory, School of Kinesiology, University of Michigan, Ann Arbor, MI, USA
| | - Lisa M Guth
- Substrate Metabolism Laboratory, School of Kinesiology, University of Michigan, Ann Arbor, MI, USA
| | - Alison C Ludzki
- Substrate Metabolism Laboratory, School of Kinesiology, University of Michigan, Ann Arbor, MI, USA
| | - Rachel A Gioscia-Ryan
- Substrate Metabolism Laboratory, School of Kinesiology, University of Michigan, Ann Arbor, MI, USA
| | - Chiwoon Ahn
- Substrate Metabolism Laboratory, School of Kinesiology, University of Michigan, Ann Arbor, MI, USA
| | - Katherine L Foug
- Substrate Metabolism Laboratory, School of Kinesiology, University of Michigan, Ann Arbor, MI, USA
| | - Jeffrey F Horowitz
- Substrate Metabolism Laboratory, School of Kinesiology, University of Michigan, Ann Arbor, MI, USA
| |
Collapse
|
117
|
Kell DB, Pretorius E. No effects without causes: the Iron Dysregulation and Dormant Microbes hypothesis for chronic, inflammatory diseases. Biol Rev Camb Philos Soc 2018; 93:1518-1557. [PMID: 29575574 PMCID: PMC6055827 DOI: 10.1111/brv.12407] [Citation(s) in RCA: 70] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2017] [Revised: 02/12/2018] [Accepted: 02/15/2018] [Indexed: 12/11/2022]
Abstract
Since the successful conquest of many acute, communicable (infectious) diseases through the use of vaccines and antibiotics, the currently most prevalent diseases are chronic and progressive in nature, and are all accompanied by inflammation. These diseases include neurodegenerative (e.g. Alzheimer's, Parkinson's), vascular (e.g. atherosclerosis, pre-eclampsia, type 2 diabetes) and autoimmune (e.g. rheumatoid arthritis and multiple sclerosis) diseases that may appear to have little in common. In fact they all share significant features, in particular chronic inflammation and its attendant inflammatory cytokines. Such effects do not happen without underlying and initially 'external' causes, and it is of interest to seek these causes. Taking a systems approach, we argue that these causes include (i) stress-induced iron dysregulation, and (ii) its ability to awaken dormant, non-replicating microbes with which the host has become infected. Other external causes may be dietary. Such microbes are capable of shedding small, but functionally significant amounts of highly inflammagenic molecules such as lipopolysaccharide and lipoteichoic acid. Sequelae include significant coagulopathies, not least the recently discovered amyloidogenic clotting of blood, leading to cell death and the release of further inflammagens. The extensive evidence discussed here implies, as was found with ulcers, that almost all chronic, infectious diseases do in fact harbour a microbial component. What differs is simply the microbes and the anatomical location from and at which they exert damage. This analysis offers novel avenues for diagnosis and treatment.
Collapse
Affiliation(s)
- Douglas B. Kell
- School of ChemistryThe University of Manchester, 131 Princess StreetManchesterLancsM1 7DNU.K.
- The Manchester Institute of BiotechnologyThe University of Manchester, 131 Princess StreetManchesterLancsM1 7DNU.K.
- Department of Physiological SciencesStellenbosch University, Stellenbosch Private Bag X1Matieland7602South Africa
| | - Etheresia Pretorius
- Department of Physiological SciencesStellenbosch University, Stellenbosch Private Bag X1Matieland7602South Africa
| |
Collapse
|
118
|
Aregbesola A, de Mello VDF, Lindström J, Voutilainen S, Virtanen JK, Keinänen-Kiukaanniemi S, Tuomainen TP, Tuomilehto J, Uusitupa M. Serum adiponectin/Ferritin ratio in relation to the risk of type 2 diabetes and insulin sensitivity. Diabetes Res Clin Pract 2018; 141:264-274. [PMID: 29777745 DOI: 10.1016/j.diabres.2018.05.012] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/23/2017] [Revised: 04/21/2018] [Accepted: 05/08/2018] [Indexed: 12/15/2022]
Abstract
AIMS Body iron inhibits the metabolism of adiponectin, an insulin sensitizing adipokine. We investigated the relationships of baseline and average of 4-year change in values of serum adiponectin (sA), serum ferritin (sF) and sA/sF ratio on type 2 diabetes (T2D) risk and insulin sensitivity (Matsuda ISI) and secretion (disposition index; DI30). METHODS Prospective analyses were conducted in participants with impaired glucose tolerance of the Finnish Diabetes Prevention Study (n = 516) recruited in 1993-1998. Cox and linear regression analyses were used to investigate the associations of sA, sF and sA/sF ratio, as continuous variables, with incident T2D, Matsuda ISI, and DI30. RESULTS During the mean follow-up of 8.2 years, 157 incident T2D cases occurred (intervention group, n = 65 and control group, n = 92). In adjusted models, baseline sA and sA/sF ratio were inversely associated with T2D risk (HR = 0.49, 95% CI 0.31-0.76, P = 0.002 and HR = 0.83, 95% CI 0.70-0.99, P = 0.044, respectively). Furthermore, a direct association was observed with Matsuda ISI (β=0.13, 95% CI 0.03-0.22, P = 0.009, for sA and β=0.04, 95% CI 0.01-0.07, P = 0.035, for sA/sF ratio) during the average 4-year follow-up. The changes in sA and sA/sF ratio were also inversely associated with T2D risk (HR = 0.36, 95% CI 0.20-0.63, P < 0.001 and HR = 0.76, 95% CI 0.62-0.92, P = 0.006, respectively), and directly with Matsuda ISI (β=0.27, 95% CI 0.17-0.38, P < 0.001, for sA and β=0.07, 95% CI 0.03-0.11, P < 0.001, for sA/sF ratio). No consistent associations were found with DI30. CONCLUSIONS: Baseline levels and changes during the follow-up in sA and sA/sF ratio are related to T2D risk and insulin sensitivity.
Collapse
Affiliation(s)
- Alex Aregbesola
- Institute of Public Health and Clinical Nutrition, University of Eastern Finland, Yliopistonranta 1C, P.O. Box 1627, FI70211 Kuopio, Finland.
| | - Vanessa D F de Mello
- Institute of Public Health and Clinical Nutrition, University of Eastern Finland, Yliopistonranta 1C, P.O. Box 1627, FI70211 Kuopio, Finland
| | - Jaana Lindström
- Diabetes Prevention Unit, National Institute for Health and Welfare, Helsinki, Finland
| | - Sari Voutilainen
- Institute of Public Health and Clinical Nutrition, University of Eastern Finland, Yliopistonranta 1C, P.O. Box 1627, FI70211 Kuopio, Finland
| | - Jyrki K Virtanen
- Institute of Public Health and Clinical Nutrition, University of Eastern Finland, Yliopistonranta 1C, P.O. Box 1627, FI70211 Kuopio, Finland
| | - Sirkka Keinänen-Kiukaanniemi
- Center for Life Course Health Research, University of Oulu, Finland; Medical Research Center and Unit of General Practice, Oulu University Hospital and Oulu Health Center, Oulu, Finland
| | - Tomi-Pekka Tuomainen
- Institute of Public Health and Clinical Nutrition, University of Eastern Finland, Yliopistonranta 1C, P.O. Box 1627, FI70211 Kuopio, Finland
| | - Jaakko Tuomilehto
- Centre for Vascular Prevention, Danube-University Krems, Krems, Austria; Dasman Diabetes Institute, Dasman, Kuwait; Diabetes Research Group, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Matti Uusitupa
- Institute of Public Health and Clinical Nutrition, University of Eastern Finland, Yliopistonranta 1C, P.O. Box 1627, FI70211 Kuopio, Finland
| |
Collapse
|
119
|
Suárez-Ortegón MF, Ensaldo-Carrasco E, Shi T, McLachlan S, Fernández-Real JM, Wild SH. Ferritin, metabolic syndrome and its components: A systematic review and meta-analysis. Atherosclerosis 2018; 275:97-106. [PMID: 29886355 DOI: 10.1016/j.atherosclerosis.2018.05.043] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/07/2018] [Revised: 05/03/2018] [Accepted: 05/22/2018] [Indexed: 01/28/2023]
Abstract
BACKGROUND AND AIMS Mechanisms for the association between iron stores and risk factors for diabetes and cardiovascular disease, such as metabolic syndrome (MetS) and its components, are still not clear. We evaluated the associations between ferritin levels, MetS and its individual components, and potential role of confounding, in a meta-analysis. METHODS We searched articles in MEDLINE and EMBASE until February 14th, 2018. There were two approaches: meta-analysis of 1) cross-sectional and longitudinal studies and 2) only cross-sectional studies. Meta-regressions were conducted to identify sources of heterogeneity in the associations of ferritin with MetS and its individual components. RESULTS Information from 26 studies (5 prospective) was systematically reviewed and 21 studies were meta-analysed. The pooled OR for MetS by increased ferritin was 1.78 (95%CI: 1.60-1.97) in the meta-analysis 1, and 1.70 (95%CI: 1.49-1.95) in the meta-analysis 2. The pooled association was weaker in studies adjusted for hepatic injury markers (meta-regression coefficient (95% CI): -0.34 (-0.60,-0.09) p = 0.008) and body mass index (BMI) (meta-regression coefficient (95% CI): -0.27 (-0.53,-0.01) p = 0.039). Among MetS components, the pooled association with increased ferritin was strongest with high triglycerides [OR (95%CI): 1.96 (1.65-2.32)] and high glucose levels [OR 95%CI: 1.60 (1.40-1.82)]. Higher cut-off points used to define high ferritin concentrations were more strongly associated with high triglycerides [meta-regression coefficient (95% CI): 0.22 (0.03, 0.041), p = 0.023]. CONCLUSIONS High triglycerides and glucose are the components more strongly associated with ferritin. Hepatic injury and BMI appear to influence the ferritin-MetS association, and a threshold effect of high ferritin concentration on the ferritin-high triglycerides association was observed.
Collapse
Affiliation(s)
- Milton Fabian Suárez-Ortegón
- Centre for Population Health Sciences, University of Edinburgh, Edinburgh, Scotland, United Kingdom; Nutrition Group, Universidad del Valle, Cali, Colombia.
| | - Eduardo Ensaldo-Carrasco
- Centre for Population Health Sciences, University of Edinburgh, Edinburgh, Scotland, United Kingdom
| | - Ting Shi
- Centre for Population Health Sciences, University of Edinburgh, Edinburgh, Scotland, United Kingdom
| | - Stela McLachlan
- Centre for Population Health Sciences, University of Edinburgh, Edinburgh, Scotland, United Kingdom
| | - José Manuel Fernández-Real
- Department of Diabetes, Endocrinology and Nutrition, Institut d'Investigació Biomèdica de Girona, CIBEROBN (CB06/03/010) and Instituto de Salud Carlos III (ISCIII), Girona, Spain.
| | - Sarah H Wild
- Centre for Population Health Sciences, University of Edinburgh, Edinburgh, Scotland, United Kingdom
| |
Collapse
|
120
|
Chand SK, Singh RG, Pendharkar SA, Petrov MS. Iron: a Strong Element in the Pathogenesis of Chronic Hyperglycaemia After Acute Pancreatitis. Biol Trace Elem Res 2018; 183:71-79. [PMID: 28836100 DOI: 10.1007/s12011-017-1131-y] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/06/2017] [Accepted: 08/11/2017] [Indexed: 12/12/2022]
Abstract
Evidence shows an association between markers of iron metabolism and glucose metabolism in type 2 diabetes mellitus. Acute pancreatitis is the largest contributor to diabetes of the exocrine pancreas. However, the pathogenesis of new-onset pre-diabetes or diabetes after pancreatitis remains unclear. This study aimed to investigate associations between markers of iron metabolism and glucose metabolism following acute pancreatitis. Fasting blood samples were collected to analyse markers of glucose metabolism (haemoglobin A1c) and iron metabolism (hepcidin, ferritin, and soluble transferrin receptor). Participants were categorised into two groups: normoglycaemia after acute pancreatitis and chronic hyperglycaemia after acute pancreatitis. Binary logistic and linear regression analyses were conducted, and potential confounders were adjusted for in multivariable analyses. A total of 83 individuals following an episode of acute pancreatitis were included, of whom 19 developed chronic hyperglycaemia. Hepcidin was significantly increased in individuals with chronic hyperglycaemia after acute pancreatitis in two adjusted models (p = 0.045 and p = 0.048). Ferritin was significantly decreased in individuals with chronic hyperglycaemia after acute pancreatitis in three adjusted models (p = 0.016, p = 0.009, and p = 0.011). Soluble transferrin receptor was not significantly associated with chronic hyperglycaemia after acute pancreatitis. These findings suggest that iron metabolism is significantly altered in individuals with chronic hyperglycaemia after acute pancreatitis and may provide better insights into the pathogenesis of new-onset diabetes after pancreatitis.
Collapse
Affiliation(s)
- Shayal K Chand
- Department of Surgery, University of Auckland, Room 12.085A, Level 12, Auckland City Hospital, Private Bag 92019, Victoria Street West, Auckland, 1142, New Zealand
| | - Ruma G Singh
- Department of Surgery, University of Auckland, Room 12.085A, Level 12, Auckland City Hospital, Private Bag 92019, Victoria Street West, Auckland, 1142, New Zealand
| | - Sayali A Pendharkar
- Department of Surgery, University of Auckland, Room 12.085A, Level 12, Auckland City Hospital, Private Bag 92019, Victoria Street West, Auckland, 1142, New Zealand
| | - Maxim S Petrov
- Department of Surgery, University of Auckland, Room 12.085A, Level 12, Auckland City Hospital, Private Bag 92019, Victoria Street West, Auckland, 1142, New Zealand.
| |
Collapse
|
121
|
Vela D, Sopi RB, Mladenov M. Low Hepcidin in Type 2 Diabetes Mellitus: Examining the Molecular Links and Their Clinical Implications. Can J Diabetes 2018; 42:179-187. [DOI: 10.1016/j.jcjd.2017.04.007] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/07/2016] [Revised: 04/18/2017] [Accepted: 04/21/2017] [Indexed: 01/14/2023]
|
122
|
Yan HF, Liu ZY, Guan ZA, Guo C. Deferoxamine ameliorates adipocyte dysfunction by modulating iron metabolism in ob/ob mice. Endocr Connect 2018; 7:604-616. [PMID: 29678877 PMCID: PMC5911700 DOI: 10.1530/ec-18-0054] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/13/2018] [Accepted: 03/27/2018] [Indexed: 12/18/2022]
Abstract
OBJECTIVE The mechanisms underlying obesity and anti-obesity processes have garnered remarkable attention as potential therapeutic targets for obesity-associated metabolic syndromes. Our prior work has shown the healing efficacy of iron reduction therapies for hepatic steatosis in a rodent model of diabetes and obesity. In this study, we investigated how iron depletion by deferoxamine (DFO) affected adipocyte dysfunction in the epididymal adipose tissues of ob/ob mice. METHODS Male ob/ob mice were assigned to either a vehicle-treated or DFO-treated group. DFO (100 mg/kg body weight) was injected intraperitoneally for 15 days. RESULTS We confirmed that iron deposits were statistically increased in the epididymal fat pad of 26-week-old ob/ob mice compared with wild-type (WT) mice. DFO significantly improved vital parameters of adipose tissue biology by reducing reactive oxygen species and inflammatory marker (TNFα, IL-2, IL-6, and Hepcidin) secretion, by increasing the levels of antioxidant enzymes, hypoxia-inducible factor-1α (HIF-1α) and HIF-1α-targeted proteins, and by altering adipocytic iron-, glucose- and lipid-associated metabolism proteins. Meanwhile, hypertrophic adipocytes were decreased in size, and insulin signaling pathway-related proteins were also activated after 15 days of DFO treatment. CONCLUSIONS These findings suggest that dysfunctional iron homeostasis contributes to the pathophysiology of obesity and insulin resistance in adipose tissues of ob/ob mice. Further investigation is required to develop safe iron chelators as effective treatment strategies against obesity, with potential for rapid clinical application.
Collapse
Affiliation(s)
- Hong-Fa Yan
- College of Life and Health SciencesNortheastern University, Shenyang, China
| | - Zhao-Yu Liu
- College of Life and Health SciencesNortheastern University, Shenyang, China
| | - Zhi-Ang Guan
- College of Life and Health SciencesNortheastern University, Shenyang, China
| | - Chuang Guo
- College of Life and Health SciencesNortheastern University, Shenyang, China
| |
Collapse
|
123
|
Abstract
Abstract
Gestational diabetes mellitus (GDM), one of the most common pregnancy complications, is defined as glucose intolerance with onset or first recognition during pregnancy. Its prevalence varies worldwide in dependence on characteristics of the underlying population and applied diagnostic criteria. The etiology is multifactorial and not sufficiently elucidated. Available evidence suggests that the base of pathogenesis is relatively diminished insulin secretion coupled with pregnancy-induced insulin resistance. Modifiable and non-modifiable risk factors for development have been identified. Trace elements and vitamin D could be contributed to modifiable factors for prediction the risk in a large population. Essential trace elements in pregnancy are necessary to overcome systemic oxidative, metabolic and inflammatory stress. Evidence, still inconclusive, has been accumulated about the relation between higher incidence of vitamin D failure/deficiency during pregnancy and GDM. The lower level of 25-OH vitamin D could be associated with increased risk for anemia development, also including pregnant women. This review intends to provide an overview of the possible link between both vitamin D and trace elements as risk factors for GDM development.
Collapse
|
124
|
Dos Santos Vieira DA, Hermes Sales C, Galvão Cesar CL, Marchioni DM, Fisberg RM. Influence of Haem, Non-Haem, and Total Iron Intake on Metabolic Syndrome and Its Components: A Population-Based Study. Nutrients 2018. [PMID: 29518910 PMCID: PMC5872732 DOI: 10.3390/nu10030314] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Studies suggest that haem, non-haem iron and total iron intake may be related to non-communicable diseases, especially metabolic syndrome. This study was undertaken to investigate the association of haem, non-haem iron and total iron intake with metabolic syndrome and its components. A cross-sectional population-based survey was performed in 2008, enrolling 591 adults and elderly adults living in São Paulo, Brazil. Dietary intake was measured by two 24 h dietary recalls. Metabolic syndrome was defined as the presence of at least three of the following: hypertension, hyperglycaemia, dyslipidaemia and central obesity. The association between different types of dietary iron and metabolic syndrome was evaluated using multiple logistic regression. After adjustment for potential confounders, a higher haem iron intake was positively associated with metabolic syndrome and with elevated triglyceride levels. A higher total iron intake was positively associated with hyperglycaemia. Non-haem iron intake was positively associated with hyperglycaemia in the fourth quintile. In conclusion, this study suggests that the different types of dietary iron are associated with metabolic syndrome, elevated triglyceride levels and hyperglycaemia. In addition, it emphasises the importance of investigating the roles of dietary iron in health outcomes, since its consumption may have different impacts on health.
Collapse
Affiliation(s)
- Diva Aliete Dos Santos Vieira
- Department of Nutrition, School of Public Health, University of Sao Paulo, Av. Dr. Arnaldo, 715, Cerqueira César, São Paulo, SP 01246-904, Brazil.
| | - Cristiane Hermes Sales
- Department of Nutrition, School of Public Health, University of Sao Paulo, Av. Dr. Arnaldo, 715, Cerqueira César, São Paulo, SP 01246-904, Brazil.
| | - Chester Luiz Galvão Cesar
- Department of Epidemiology, School of Public Health, University of Sao Paulo, Av. Dr. Arnaldo, 715, Cerqueira César, São Paulo, SP 01246-904, Brazil.
| | - Dirce Maria Marchioni
- Department of Nutrition, School of Public Health, University of Sao Paulo, Av. Dr. Arnaldo, 715, Cerqueira César, São Paulo, SP 01246-904, Brazil.
| | - Regina Mara Fisberg
- Department of Nutrition, School of Public Health, University of Sao Paulo, Av. Dr. Arnaldo, 715, Cerqueira César, São Paulo, SP 01246-904, Brazil.
| |
Collapse
|
125
|
Schindler C, Birkenfeld AL, Hanefeld M, Schatz U, Köhler C, Grüneberg M, Tschöpe D, Blüher M, Hasslacher C, Bornstein SR. Intravenous Ferric Carboxymaltose in Patients with Type 2 Diabetes Mellitus and Iron Deficiency: CLEVER Trial Study Design and Protocol. Diabetes Ther 2018; 9:37-47. [PMID: 29134606 PMCID: PMC5801218 DOI: 10.1007/s13300-017-0330-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/11/2017] [Indexed: 01/31/2023] Open
Abstract
INTRODUCTION HbA1c is the gold standard for glycemic control in pre-diabetes and diabetes. However, its validity has been questioned, especially in the presence of imbalanced iron homeostasis. The CLEVER trial aims to evaluate the relationship between iron deficiency and HbA1c (a biomarker for the diagnosis and therapeutic monitoring of type 2 diabetes) in a randomized, placebo-controlled, multicenter clinical trial. METHODS The CLEVER (intravenous ferric CarboxymaLtosE for improVement of mEtabolic parameters in type 2 diabetes patients with iRon deficiency) trial is a randomized, single-blind, proof-of-concept study with two treatment arms. 140 men and women diagnosed with type 2 diabetes and iron deficiency will receive either placebo or ferric carboxymaltose (500 or 1000 mg) as intravenous infusions. The primary outcome measure is the change in HbA1c level between baseline and after 12 weeks of treatment. Secondary endpoints include change of iron status and metabolic markers as well as treatment safety and tolerability. Furthermore, the potential clinical improvement in quality of life and the reliability of HbA1c measurement in patients with type 2 diabetes and iron deficiency will be investigated. RESULTS Both excessive iron and iron deficiency are associated with metabolic disorders; excessive iron is a risk factor for the development of diabetes, whereas iron deficiency is associated with obesity and insulin resistance. It has been suggested that iron increases insulin secretion in pancreatic beta-cells. CLEVER is the first study to investigate the hypothesis that intravenous substitution with ferric carboxymaltose reduces HbA1c levels in patients with type 2 diabetes and iron deficiency, thereby improving metabolic status and quality of life. CLINICAL TRIAL REGISTRATION ClinicalTrials.gov (NCT01513369). FUNDING GWT-TUD GmbH acts as sponsor of the clinical trial. Financial support is provided by Vifor Pharma.
Collapse
Affiliation(s)
- Christoph Schindler
- Clinical Research Center Hannover and Center for Pharmacology and Toxicology, Hannover Medical School, Hannover, Germany.
| | - Andreas L Birkenfeld
- Medical Clinic and Policlinic III at the University Hospital Dresden, Dresden, Germany
- Centre for Metabolic Vascular Medicine, GWT-TUD, Dresden, Germany
| | - Markolf Hanefeld
- Centre for Metabolic Vascular Medicine, GWT-TUD, Dresden, Germany
| | - Ulrike Schatz
- Medical Clinic and Policlinic III at the University Hospital Dresden, Dresden, Germany
| | | | | | - Diethelm Tschöpe
- Herz- und Diabeteszentrum Nordrhein-Westfalen, Ruhr-Universität Bochum, Bad Oeynhausen, Germany
| | - Matthias Blüher
- Department of Medicine at the University Hospital Leipzig, Leipzig, Germany
| | - Christoph Hasslacher
- Diabetesinstitut Heidelberg and Department of Clinical Studies, St. Josefskrankenhaus Heidelberg, Heidelberg, Germany
| | - Stefan R Bornstein
- Medical Clinic and Policlinic III at the University Hospital Dresden, Dresden, Germany
| |
Collapse
|
126
|
Abstract
Pregnant women are particularly vulnerable to iron deficiency and related adverse pregnancy outcomes and, as such, are routinely recommended for iron supplementation. Emerging evidence from both animal and population-based studies, however, has raised potential concerns because significant associations have been observed between greater iron stores and disturbances in glucose metabolism, including increased risk of type 2 diabetes among nonpregnant individuals. Yet, the evidence is uncertain regarding the role of iron in the development of gestational diabetes mellitus (GDM), a common pregnancy complication which has short-term and long-term adverse health ramifications for both women and their children. In this review, we critically and systematically evaluate available data examining the risk of GDM associated with dietary iron, iron supplementation, and iron status as measured by blood concentrations of several indicators. We also discuss major methodologic concerns regarding the available epidemiologic studies on iron and GDM.
Collapse
Affiliation(s)
- Cuilin Zhang
- Epidemiology Branch, Division of Intramural Population Health Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, Bethesda, MD
| | | |
Collapse
|
127
|
Fernández Real JM, Moreno-Navarrete JM, Manco M. Iron influences on the Gut-Brain axis and development of type 2 diabetes. Crit Rev Food Sci Nutr 2017; 59:443-449. [DOI: 10.1080/10408398.2017.1376616] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Affiliation(s)
- José Manuel Fernández Real
- University Hospital of Girona ‘Dr JosepTrueta’, Department of Diabetes, Endocrinology and Nutrition, Institut d'Investigació Biomèdica de Girona IdibGi
- CIBER Fisiopatología de la Obesidad y Nutrición, Girona, Spain
| | - José Maria Moreno-Navarrete
- University Hospital of Girona ‘Dr JosepTrueta’, Department of Diabetes, Endocrinology and Nutrition, Institut d'Investigació Biomèdica de Girona IdibGi
- CIBER Fisiopatología de la Obesidad y Nutrición, Girona, Spain
| | - Melania Manco
- Research Area for multifactorial diseases, Bambino Gesù Children's Hospital and Research Institute, Research Unit for Multifactorial Disease, Rome, Italy
| |
Collapse
|
128
|
Raffield LM, Louie T, Sofer T, Jain D, Ipp E, Taylor KD, Papanicolaou GJ, Avilés-Santa L, Lange LA, Laurie CC, Conomos MP, Thornton TA, Chen YDI, Qi Q, Cotler S, Thyagarajan B, Schneiderman N, Rotter JI, Reiner AP, Lin HJ. Genome-wide association study of iron traits and relation to diabetes in the Hispanic Community Health Study/Study of Latinos (HCHS/SOL): potential genomic intersection of iron and glucose regulation? Hum Mol Genet 2017; 26:1966-1978. [PMID: 28334935 DOI: 10.1093/hmg/ddx082] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2016] [Accepted: 03/01/2017] [Indexed: 12/14/2022] Open
Abstract
Genetic variants contribute to normal variation of iron-related traits and may also cause clinical syndromes of iron deficiency or excess. Iron overload and deficiency can adversely affect human health. For example, elevated iron storage is associated with increased diabetes risk, although mechanisms are still being investigated. We conducted the first genome-wide association study of serum iron, total iron binding capacity (TIBC), transferrin saturation, and ferritin in a Hispanic/Latino cohort, the Hispanic Community Health Study/Study of Latinos (>12 000 participants) and also assessed the generalization of previously known loci to this population. We then evaluated whether iron-associated variants were associated with diabetes and glycemic traits. We found evidence for a novel association between TIBC and a variant near the gene for protein phosphatase 1, regulatory subunit 3B (PPP1R3B; rs4841132, β = -0.116, P = 7.44 × 10-8). The effect strengthened when iron deficient individuals were excluded (β = -0.121, P = 4.78 × 10-9). Ten of sixteen variants previously associated with iron traits generalized to HCHS/SOL, including variants at the transferrin (TF), hemochromatosis (HFE), fatty acid desaturase 2 (FADS2)/myelin regulatory factor (MYRF), transmembrane protease, serine 6 (TMPRSS6), transferrin receptor (TFR2), N-acetyltransferase 2 (arylamine N-acetyltransferase) (NAT2), ABO blood group (ABO), and GRB2 associated binding protein 3 (GAB3) loci. In examining iron variant associations with glucose homeostasis, an iron-raising variant of TMPRSS6 was associated with lower HbA1c levels (P = 8.66 × 10-10). This association was attenuated upon adjustment for iron measures. In contrast, the iron-raising allele of PPP1R3B was associated with higher levels of fasting glucose (P = 7.70 × 10-7) and fasting insulin (P = 4.79 × 10-6), but these associations were not attenuated upon adjustment for TIBC-so iron is not likely a mediator. These results provide new genetic information on iron traits and their connection with glucose homeostasis.
Collapse
Affiliation(s)
- Laura M Raffield
- Department of Genetics, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Tin Louie
- Department of Biostatistics, University of Washington, Seattle, WA 98195, USA
| | - Tamar Sofer
- Department of Biostatistics, University of Washington, Seattle, WA 98195, USA
| | - Deepti Jain
- Department of Biostatistics, University of Washington, Seattle, WA 98195, USA
| | - Eli Ipp
- Department of Medicine and Division of Endocrinology, Harbor-UCLA Medical Center, Torrance, CA 90502, USA and the David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Kent D Taylor
- Institute for Translational Genomics and Population Sciences, Los Angeles Biomedical Research Institute, and Department of Pediatrics, Harbor-UCLA Medical Center, Torrance, CA 90502, and the David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | | | | | - Leslie A Lange
- Department of Genetics, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Cathy C Laurie
- Department of Biostatistics, University of Washington, Seattle, WA 98195, USA
| | - Matthew P Conomos
- Department of Biostatistics, University of Washington, Seattle, WA 98195, USA
| | - Timothy A Thornton
- Department of Biostatistics, University of Washington, Seattle, WA 98195, USA
| | - Yii-Der Ida Chen
- Institute for Translational Genomics and Population Sciences, Los Angeles Biomedical Research Institute, and Department of Pediatrics, Harbor-UCLA Medical Center, Torrance, CA 90502, and the David Geffen School of Medicine at UCLA, Los Angeles, CA, USA.,Department of Medicine, Harbor-UCLA Medical Center, Torrance, CA 90502, and the David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Qibin Qi
- Department of Epidemiology and Population Health, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Scott Cotler
- Department of Medicine, Division of Hepatology, Loyola University Medical Center, Maywood, IL 60153, USA
| | - Bharat Thyagarajan
- Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, MN 55455, USA
| | - Neil Schneiderman
- Department of Psychology and Behavioral Medicine, University of Miami, FL 33124, USA
| | - Jerome I Rotter
- Institute for Translational Genomics and Population Sciences, Los Angeles Biomedical Research Institute, and Department of Pediatrics, Harbor-UCLA Medical Center, Torrance, CA 90502, and the David Geffen School of Medicine at UCLA, Los Angeles, CA, USA.,Department of Medicine, Harbor-UCLA Medical Center, Torrance, CA 90502, and the David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Alex P Reiner
- Department of Epidemiology, University of Washington, Seattle, WA 98195, USA
| | - Henry J Lin
- Institute for Translational Genomics and Population Sciences, Los Angeles Biomedical Research Institute, and Department of Pediatrics, Harbor-UCLA Medical Center, Torrance, CA 90502, and the David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| |
Collapse
|
129
|
Chand SK, Singh RG, Pendharkar SA, Bharmal SH, Petrov MS. Interplay between innate immunity and iron metabolism after acute pancreatitis. Cytokine 2017; 103:90-98. [PMID: 28982582 DOI: 10.1016/j.cyto.2017.09.014] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2017] [Revised: 08/21/2017] [Accepted: 09/12/2017] [Indexed: 12/12/2022]
Abstract
Emerging evidence shows that chronic low-grade inflammation and changes in markers of innate immunity are implicated in a range of metabolic abnormalities following an episode of acute pancreatitis. Also, deranged iron metabolism has been linked to type 2 diabetes mellitus, gestational diabetes, and new-onset diabetes after pancreatitis - the conditions characterized by high haemoglobin glycation index (HGI). This study aimed to investigate the associations between markers of innate immunity and iron metabolism in individuals after acute pancreatitis. Fasting blood samples were collected to analyse lipopolysaccharide binding protein (LBP), interleukin (IL)-6, tumor necrosis factor-α, hepcidin, ferritin, soluble transferrin receptor, HbA1c, and glucose. Participants were categorized into two groups: low HGI and high HGI. Linear regression analyses were conducted, and potential confounders (age, sex, ethnicity, body mass index, diabetes mellitus status, smoking status, aetiology of pancreatitis, duration, recurrence, and severity of pancreatitis) were adjusted for in 5 statistical models. A total of 93 patients following an episode of acute pancreatitis were included, of who 40 (43%) had high HGI. In the overall cohort, LBP was significantly associated with hepcidin and ferritin, and IL-6 was significantly associated with hepcidin, consistently in all the models. Further, LBP contributed to 7.7% and 9.5% of variance in hepcidin and ferritin levels, respectively, whereas IL-6 contributed to 5.3% of hepcidin variance. Upon subgroup analysis, the observed LBP associations were maintained in the high HGI subgroup only and the IL-6 association in the low HGI subgroup only. No consistently significant associations were found between any of the other markers. The interplay between LBP, IL-6, hepcidin, and ferritin characterizes metabolic derangements after acute pancreatitis and may play a role in the pathogenesis of new-onset diabetes after pancreatitis.
Collapse
Affiliation(s)
- Shayal K Chand
- Department of Surgery, University of Auckland, Auckland, New Zealand
| | - Ruma G Singh
- Department of Surgery, University of Auckland, Auckland, New Zealand
| | | | - Sakina H Bharmal
- Department of Surgery, University of Auckland, Auckland, New Zealand
| | - Maxim S Petrov
- Department of Surgery, University of Auckland, Auckland, New Zealand.
| |
Collapse
|
130
|
Koppe T, Patchen B, Cheng A, Bhasin M, Vulpe C, Schwartz RE, Moreno‐Navarrete JM, Fernandez‐Real JM, Pissios P, Fraenkel PG. Nicotinamide N-methyltransferase expression decreases in iron overload, exacerbating toxicity in mouse hepatocytes. Hepatol Commun 2017; 1:803-815. [PMID: 29404495 PMCID: PMC5678920 DOI: 10.1002/hep4.1083] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/17/2017] [Revised: 07/27/2017] [Accepted: 08/01/2017] [Indexed: 12/19/2022] Open
Abstract
Iron overload causes the generation of reactive oxygen species that can lead to lasting damage to the liver and other organs. The goal of this study was to identify genes that modify the toxicity of iron overload. We studied the effect of iron overload on the hepatic transcriptional and metabolomic profile in mouse models using a dietary model of iron overload and a genetic model, the hemojuvelin knockout mouse. We then evaluated the correlation of nicotinamide N-methyltransferase (NNMT) expression with body iron stores in human patients and the effect of NNMT knockdown on gene expression and viability in primary mouse hepatocytes. We found that iron overload induced significant changes in the expression of genes and metabolites involved in glucose and nicotinamide metabolism and that NNMT, an enzyme that methylates nicotinamide and regulates hepatic glucose and cholesterol metabolism, is one of the most strongly down-regulated genes in the liver in both genetic and dietary iron overload. We found that hepatic NNMT expression is inversely correlated with serum ferritin levels and serum transferrin saturation in patients who are obese, suggesting that body iron stores regulate human liver NNMT expression. Furthermore, we demonstrated that adenoviral knockdown of NNMT in primary mouse hepatocytes exacerbates iron-induced hepatocyte toxicity and increases expression of transcriptional markers of oxidative and endoplasmic reticulum stress, while overexpression of NNMT partially reversed these effects. Conclusion: Iron overload alters glucose and nicotinamide transcriptional and metabolic pathways in mouse hepatocytes and decreases NNMT expression, while NNMT deficiency worsens the toxic effect of iron overload. For these reasons, NNMT may be a drug target for the prevention of iron-induced hepatotoxicity. (Hepatology Communications 2017;1:803-815).
Collapse
Affiliation(s)
- Tiago Koppe
- Division of Hematology/Oncology
- Cancer Research Institute, Beth Israel Deaconess Medical CenterBostonMA
- Department of MedicineHarvard Medical SchoolBostonMA
| | - Bonnie Patchen
- Division of Hematology/Oncology
- Cancer Research Institute, Beth Israel Deaconess Medical CenterBostonMA
- Department of MedicineHarvard Medical SchoolBostonMA
| | - Aaron Cheng
- Division of Hematology/Oncology
- Cancer Research Institute, Beth Israel Deaconess Medical CenterBostonMA
- Department of MedicineHarvard Medical SchoolBostonMA
| | - Manoj Bhasin
- Department of MedicineHarvard Medical SchoolBostonMA
- Division of Interdisciplinary Medicine and Biotechnology, Beth Israel Deaconess Medical CenterBostonMA
| | - Chris Vulpe
- Department of Physiological SciencesUniversity of FloridaGainesvilleFL
| | - Robert E. Schwartz
- Division of Gastroenterology and Hepatology, Weill Cornell Medical SchoolNew YorkNY
| | - Jose Maria Moreno‐Navarrete
- Department of Diabetes, Endocrinology and Nutrition, Institut d'Investigació Biomèdica de GironaHospital de Girona Dr. Josep Trueta and Universitat de GironaGironaSpain
- CIBER Fisopatologia de la Obesidad y Nutricion, Instituto de Salud Carlos IIIMadridSpain
| | - Jose Manuel Fernandez‐Real
- Department of Diabetes, Endocrinology and Nutrition, Institut d'Investigació Biomèdica de GironaHospital de Girona Dr. Josep Trueta and Universitat de GironaGironaSpain
- CIBER Fisopatologia de la Obesidad y Nutricion, Instituto de Salud Carlos IIIMadridSpain
| | - Pavlos Pissios
- Division of Endocrinology, Beth Israel Deaconess Medical CenterBostonMA
| | - Paula G. Fraenkel
- Division of Hematology/Oncology
- Cancer Research Institute, Beth Israel Deaconess Medical CenterBostonMA
- Department of MedicineHarvard Medical SchoolBostonMA
| |
Collapse
|
131
|
Barros RK, Cotrim HP, Daltro CH, Oliveira YA. Hyperferritinemia in patients with nonalcoholic fatty liver disease. Rev Assoc Med Bras (1992) 2017; 63:284-289. [PMID: 28489136 DOI: 10.1590/1806-9282.63.03.284] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2016] [Accepted: 08/06/2016] [Indexed: 12/17/2022] Open
Abstract
Objective: In liver diseases, hyperferritinemia (HYF) is related to injured cells in acquired and genetic conditions with or without iron overload. It is frequent in patients with nonalcoholic fatty liver disease (NAFLD), in which it is necessary to define the mean of HYF to establish the better approach for them. The present study evaluated the significance of elevated ferritin in patients with NAFLD and steatohepatitis (NASH). Method: The review was performed using search instruments of indexed scientific material, including MEDLINE (by PubMed), Web of Science, IBECS and LILACS, to identify articles published in Portuguese, English and Spanish, from 2005 to May, 2016. Studies eligible included place and year of publication, diagnose criteria to NAFLD, specifications of serum ferritin measurements and/or liver histopathologic study. Exclusion criteria included studies with patients with alcohol consumption ≥ 20 g/day and other liver diseases. Results: A total of 11 from 30 articles were selected. It included 3,564 patients and they were cross-sectional, retrospective, case series and case-control. The result's analyses showed in 10 of these studies a relationship between ferritin elevated serum levels and NAFLD/NASH with and without fibrosis and insulin resistance. Conclusion: Hyperferritinemia in patients with NAFLD/NASH is associated more frequently with hepatocellular injury than hemochromatosis. These data suggest the relevance to evaluate carefully HYF in patients with NAFLD/NASH to establish appropriate clinical approach.
Collapse
Affiliation(s)
- Raffaelle K Barros
- Medicine and Health Graduate Program (PPgMS), Study Group on Nonalcoholic Steatohepatitis, Faculdade de Medicina da Bahia, Universidade Federal da Bahia, Salvador, BA, Brazill
| | - Helma Pinchemel Cotrim
- Medicine and Health Graduate Program (PPgMS), Study Group on Nonalcoholic Steatohepatitis, Faculdade de Medicina da Bahia, Universidade Federal da Bahia, Salvador, BA, Brazill
| | - Carla H Daltro
- Medicine and Health Graduate Program (PPgMS), Study Group on Nonalcoholic Steatohepatitis, Faculdade de Medicina da Bahia, Universidade Federal da Bahia, Salvador, BA, Brazill
| | - Yanaihara A Oliveira
- Medicine and Health Graduate Program (PPgMS), Study Group on Nonalcoholic Steatohepatitis, Faculdade de Medicina da Bahia, Universidade Federal da Bahia, Salvador, BA, Brazill
| |
Collapse
|
132
|
Vela D, Leshoski J, Vela Z, Jakupaj M, Mladenov M, Sopi RB. Insulin treatment corrects hepcidin but not YKL-40 levels in persons with type 2 diabetes mellitus matched by body mass index, waist-to-height ratio, C-reactive protein and Creatinine. BMC Endocr Disord 2017; 17:53. [PMID: 28841871 PMCID: PMC5574085 DOI: 10.1186/s12902-017-0204-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/08/2017] [Accepted: 08/21/2017] [Indexed: 01/08/2023] Open
Abstract
BACKGROUND It has been shown that hepcidin and YKL-40 levels change in persons with insulin resistance in different circumstances. However, variations of the levels of these parameters through the stages of prediabetes and type 2 diabetes mellitus are unclear. We hypothesized that hepcidin levels will decrease in persons with prediabetes, while these levels will tend to correct when persons with diabetes are treated with insulin. Finally we sought to determine the levels of YKL-40 in all groups of participants included in the study. METHODS Serum hepcidin levels and YKL-40 levels were measured in control group (n = 20), persons with prediabetes (n = 30) and persons with diabetes on insulin therapy (n = 30) using ELISA method. Patients in all three groups were matched by Body Mass Index, Waist-to-Height Ratio, C-Reactive Protein and creatinine levels. RESULTS Hepcidin levels were lower in persons with prediabetes compared to control, while persons with diabetes on insulin therapy had higher values than those with prediabetes (p = 0,00001). YKL-40 levels showed no significant changes. CONCLUSIONS Serum hepcidin levels in matched persons with prediabetes are a stronger marker of early changes in glucose metabolism compared to YKL-40 levels. Also, treatment with insulin corrects hepcidin levels, but not YKL-40 levels. Correcting levels of hepcidin is important for reducing iron-overload, which is a risk factor for diabetes.
Collapse
Affiliation(s)
- Driton Vela
- Faculty of Medicine, University of Prishtina, Martyr’s Boulevard n.n, 10000 Prishtina, Kosovo
| | - Jovica Leshoski
- Institute of Biology, Faculty of Natural Sciences, “Sts. Cyril and Methodius” University, Skopje, 1000 Macedonia
| | - Zana Vela
- Faculty of Medicine, University of Prishtina, Martyr’s Boulevard n.n, 10000 Prishtina, Kosovo
| | - Muharrem Jakupaj
- Faculty of Medicine, University of Prishtina, Martyr’s Boulevard n.n, 10000 Prishtina, Kosovo
| | - Mitko Mladenov
- Institute of Biology, Faculty of Natural Sciences, “Sts. Cyril and Methodius” University, Skopje, 1000 Macedonia
| | - Ramadan B. Sopi
- Faculty of Medicine, University of Prishtina, Martyr’s Boulevard n.n, 10000 Prishtina, Kosovo
| |
Collapse
|
133
|
Increased adipose tissue heme levels and exportation are associated with altered systemic glucose metabolism. Sci Rep 2017; 7:5305. [PMID: 28706239 PMCID: PMC5509649 DOI: 10.1038/s41598-017-05597-2] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2017] [Accepted: 05/31/2017] [Indexed: 02/06/2023] Open
Abstract
Iron status is known to be associated with the physiology of adipose tissue (AT). We aimed to investigate AT heme and expression of heme exporter (FLVCR1) in association with obesity and type 2 diabetes (T2D). Substantial amounts of FLVCR1 mRNA and protein levels were detected in AT, being significantly increased in subjects with T2D, and positively correlated with fasting glucose, fasting triglycerides and with circulating markers of iron stores (serum ferritin, blood hemoglobin and hematocrit). In both visceral (VAT) and subcutaneous AT (SAT), increased heme levels were found in subjects with T2D. Reinforcing these associations, FLVCR1 mRNA levels were positively linked to fasting glucose in an independent cohort. Longitudianlly, the percent change of FLVCR1 positively correlated with the percent change in fasting glucose (r = 0.52, p = 0.03) after bariatric surgery-induced weight loss. High-fat diet-induced weight gain in rats did not result in significant changes in AT Flvcr1 mRNA but, remarkably, the expression of this gene positively correlated with fasting glucose and negatively with insulin sensitivity (QUICKI). Altogether, these findings showed a direct association between FLVCR1 mRNA levels and hyperglycemia, suggesting that increased adipose tissue heme exportation might disrupt, or is the consequence of, impaired systemic glucose metabolism during the progression to T2D.
Collapse
|
134
|
Effects of Protein-Iron Complex Concentrate Supplementation on Iron Metabolism, Oxidative and Immune Status in Preweaning Calves. Int J Mol Sci 2017; 18:ijms18071501. [PMID: 28704940 PMCID: PMC5535991 DOI: 10.3390/ijms18071501] [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: 05/31/2017] [Revised: 07/05/2017] [Accepted: 07/07/2017] [Indexed: 01/20/2023] Open
Abstract
The objective of this study was to determine the effects of feeding protein-iron complex (PIC) on productive performance and indicators of iron metabolism, hematology parameters, antioxidant and immune status during first 35 days of a calf’s life. Preparation of the complex involved enzymatic hydrolysis of milk casein (serine protease from Yarrowia lipolytica yeast). Iron chloride was then added to the hydrolyzate and lyophilizate. Calves were divided into treated groups: LFe (low iron dose) 10 g/day calf of protein-iron complex, HFe (height iron dose) 20 g/day calf, and control group. Dietary supplements containing the lower dose of concentrate had a significant positive effect on iron metabolism, while the higher dose of concentrate resulted in increase of total iron binding capacity (TIBC), saturation of transferrin and decrease of and unsaturated iron binding capacity (UIBC), which suggest iron overload. Additionally, treatment with the lower dose of iron remarkably increased the antioxidant parameters, mainly total antioxidant (TAS) and glutathione peroxidase activity (GPx). Higher doses of PIC were related to lower total antioxidant status. IgG, IgM, insulin, glucose, TNFα and IGF-1 concentration did not change significantly in either group after supplementation. In practice, the use of protein-iron complex concentrate requires taking into account the iron content in milk replacers and other feedstuffs.
Collapse
|
135
|
Crielaard BJ, Lammers T, Rivella S. Targeting iron metabolism in drug discovery and delivery. Nat Rev Drug Discov 2017; 16:400-423. [PMID: 28154410 PMCID: PMC5455971 DOI: 10.1038/nrd.2016.248] [Citation(s) in RCA: 244] [Impact Index Per Article: 30.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Iron fulfils a central role in many essential biochemical processes in human physiology; thus, proper processing of iron is crucial. Although iron metabolism is subject to relatively strict physiological control, numerous disorders, such as cancer and neurodegenerative diseases, have recently been linked to deregulated iron homeostasis. Consequently, iron metabolism constitutes a promising and largely unexploited therapeutic target for the development of new pharmacological treatments for these diseases. Several iron metabolism-targeted therapies are already under clinical evaluation for haematological disorders, and these and newly developed therapeutic agents are likely to have substantial benefit in the clinical management of iron metabolism-associated diseases, for which few efficacious treatments are currently available.
Collapse
Affiliation(s)
- Bart J. Crielaard
- Department of Polymer Chemistry and Bioengineering, Zernike Institute for Advanced Materials, Faculty of Mathematics and Natural Sciences, University of Groningen, Groningen, The Netherlands
- W.J. Kolff Institute for Biomedical Engineering and Materials Science, University Medical Center Groningen, Groningen, The Netherlands
| | - Twan Lammers
- Department of Nanomedicine and Theranostics, Institute for Experimental Molecular Imaging, University Clinic and Helmholtz Institute for Biomedical Engineering, RWTH Aachen University, Aachen, Germany
- Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, The Netherlands
- Department of Targeted Therapeutics, MIRA Institute for Biomedical Technology and Technical Medicine, University of Twente, Enschede, The Netherlands
| | - Stefano Rivella
- Children’s Hospital of Philadelphia, Abramson Research Center, Philadelphia, PA, United States of America
| |
Collapse
|
136
|
Reichert CO, da Cunha J, Levy D, Maselli LMF, Bydlowski SP, Spada C. Hepcidin: Homeostasis and Diseases Related to Iron Metabolism. Acta Haematol 2017; 137:220-236. [PMID: 28514781 DOI: 10.1159/000471838] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2017] [Accepted: 03/20/2017] [Indexed: 12/14/2022]
Abstract
Iron is an essential metal for cell survival that is regulated by the peptide hormone hepcidin. However, its influence on certain diseases is directly related to iron metabolism or secondary to underlying diseases. Genetic alterations influence the serum hepcidin concentration, which can lead to an iron overload in tissues, as observed in haemochromatosis, in which serum hepcidin or defective hepcidin synthesis is observed. Another genetic imbalance of iron is iron-refractory anaemia, in which serum concentrations of hepcidin are increased, precluding the flow and efflux of extra- and intracellular iron. During the pathogenesis of certain diseases, the resulting oxidative stress, as well as the increase in inflammatory cytokines, influences the transcription of the HAMP gene to generate a secondary anaemia due to the increase in the serum concentration of hepcidin. To date, there is no available drug to inhibit or enhance hepcidin transcription, mostly due to the cytotoxicity described in the in vitro models. The proposed therapeutic targets are still in the early stages of clinical trials. Some candidates are promising, such as heparin derivatives and minihepcidins. This review describes the main pathways of systemic and genetic regulation of hepcidin, as well as its influence on the disorders related to iron metabolism.
Collapse
Affiliation(s)
- Cadiele Oliana Reichert
- Clinical Analysis Department, Health Sciences Center, Federal University of Santa Catarina (UFSC), Florianópolis, Brazil
| | | | | | | | | | | |
Collapse
|
137
|
Uwaezuoke SN. Vitamin D deficiency and anemia risk in children: a review of emerging evidence. PEDIATRIC HEALTH MEDICINE AND THERAPEUTICS 2017; 8:47-55. [PMID: 29388633 PMCID: PMC5774601 DOI: 10.2147/phmt.s129362] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
There has been renewed scientific interest in the sequelae of vitamin D deficiency, given the emerging evidence on the diverse biologic functions of vitamin D, besides its fundamental role in bone and mineral metabolism. For the past decade, the evidence in the medical literature pointing to a relationship between anemia risk and vitamin D deficiency has been accumulating. This paper critically reviews the current evidence linking vitamin D deficiency to anemia risk in children. The synthesized evidence indicates that the studies, which were preponderantly conducted among the adult population, not only reported a bidirectional relationship between vitamin D deficiency and anemia but also showed a racial effect. In studies conducted among children, similar results were reported. Although the causal association of vitamin D deficiency with anemia risk (especially iron-deficiency anemia) remains debatable, the noncalcemic actions of the vitamin and its analogs hold prospects for several novel clinical applications. There is, however, unanimity in many reports suggesting that vitamin D deficiency is directly associated with anemia of chronic disease or inflammation. Despite the advances in unraveling the role of vitamin D in iron homeostasis, further research is still required to validate causality in the relationship between vitamin D deficiency and anemia, as well as to determine its optimal dosing, the ideal recipients for therapeutic intervention, and the preferred analogs to administer.
Collapse
Affiliation(s)
- Samuel N Uwaezuoke
- Department of Paediatrics, College of Medicine, University of Nigeria, Nsukka.,Department of Paediatrics, University of Nigeria Teaching Hospital, Ituku-Ozalla, Nigeria
| |
Collapse
|
138
|
Mao X, Chen H, Tang J, Wang L, Shu T. Hepcidin links gluco-toxicity to pancreatic beta cell dysfunction by inhibiting Pdx-1 expression. Endocr Connect 2017; 6:121-128. [PMID: 28179377 PMCID: PMC5424768 DOI: 10.1530/ec-16-0115] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/28/2016] [Accepted: 02/08/2017] [Indexed: 12/16/2022]
Abstract
OBJECTIVE Gluco-toxicity is a term used to convey the detrimental effect of hyperglycemia on β-cell function through impaired insulin synthesis. Although it is known that the expression and activity of several key insulin transcription regulators is inhibited, other molecular mechanisms that mediate gluco-toxicity are poorly defined. Our objective was to explore the role of hepcidin in β-cell gluco-toxicity. DESIGN We first confirmed that high glucose levels inhibited hepcidin expression in the mouse insulinoma cell line, MIN6. The downregulation of hepcidin decreased Pdx-1 expression, which reduced insulin synthesis. METHODS MIN6 cells were exposed to high glucose concentrations (33.3 mmol/L). Glucose-stimulated insulin secretion (GSIS) and serum hepcidin levels were measured by ELISA. The mRNA levels of insulin1, insulin2, Pdx-1 and hepcidin were measured by real-time polymerase chain reaction. Western blot analysis was used to detect the changes in PDX-1 expression. Transient overexpression with hepcidin was used to reverse the downregulation of Pdx-1 and insulin synthesis induced by gluco-toxicity. RESULTS Exposure of MIN6 cells to high glucose significantly decreased GSIS and inhibited insulin synthesis as well as Pdx-1 transcriptional activity and expression at both the mRNA and protein levels. High glucose also decreased hepcidin expression and secretion. Hepcidin overexpression in MIN6 cells partially reversed the gluco-toxicity-induced downregulation of Pdx-1 and insulin expression and improved GSIS. The restoration of insulin synthesis by transfection of a hepcidin overexpression plasmid confirmed the role of hepcidin in mediating the gluco-toxic inhibition of insulin synthesis. CONCLUSIONS Our observations suggest that hepcidin is associated with gluco-toxicity-reduced pancreatic β-cell insulin synthesis in type 2 diabetes by inhibiting Pdx-1 expression.
Collapse
Affiliation(s)
- Xuhua Mao
- Department of Clinical LaboratoryYixing People's Hospital, Yixing, Wuxi, Jiangsu, China
| | - Hucheng Chen
- Department of Nuclear MedicineNanjing Hospital Affiliated to Nanjing Medical University, Nanjing, Jiangsu, China
| | - Junmin Tang
- Department of Clinical LaboratoryYixing People's Hospital, Yixing, Wuxi, Jiangsu, China
| | - Liangliang Wang
- Department of NeurologyYixing People's Hospital Affiliated to Jiangsu University, Yixing, Wuxi, Jiangsu, China
| | - Tingting Shu
- Department of Central LaboratoryJiangsu Province Official Hospital, Nanjing, Jiangsu, China
| |
Collapse
|
139
|
Spahis S, Borys JM, Levy E. Metabolic Syndrome as a Multifaceted Risk Factor for Oxidative Stress. Antioxid Redox Signal 2017; 26:445-461. [PMID: 27302002 DOI: 10.1089/ars.2016.6756] [Citation(s) in RCA: 89] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
SIGNIFICANCE Metabolic syndrome (MetS) is associated with a greater risk of diabetes and cardiovascular diseases. It is estimated that this multifactorial condition affects 20%-30% of the world's population. A detailed understanding of MetS mechanisms is crucial for the development of effective prevention strategies and adequate intervention tools that could curb its increasing prevalence and limit its comorbidities, particularly in younger age groups. With advances in basic redox biology, oxidative stress (OxS) involvement in the complex pathophysiology of MetS has become widely accepted. Nevertheless, its clear association with and causative effects on MetS require further elucidation. Recent Advances: Although a better understanding of the causes, risks, and effects of MetS is essential, studies suggest that oxidant/antioxidant imbalance is a key contributor to this condition. OxS is now understood to be a major underlying mechanism for mitochondrial dysfunction, ectopic lipid accumulation, and gut microbiota impairment. CRITICAL ISSUES Further studies, particularly in the field of translational research, are clearly required to understand and control the production of reactive oxygen species (ROS) levels, especially in the mitochondria, since the various therapeutic trials conducted to date have not targeted this major ROS-generating system, aimed to delay MetS onset, or prevent its progression. FUTURE DIRECTIONS Multiple relevant markers need to be identified to clarify the role of ROS in the etiology of MetS. Future clinical trials should provide important proof of concept for the effectiveness of antioxidants as useful therapeutic approaches to simultaneously counteract mitochondrial OxS, alleviate MetS symptoms, and prevent complications. Antioxid. Redox Signal. 26, 445-461.
Collapse
Affiliation(s)
- Schohraya Spahis
- 1 Research Center , Ste-Justine MUHC, Montreal, Canada .,2 Department of Nutrition, Université de Montréal , Montreal, Canada
| | | | - Emile Levy
- 1 Research Center , Ste-Justine MUHC, Montreal, Canada .,2 Department of Nutrition, Université de Montréal , Montreal, Canada .,3 EPODE International Network , Paris, France
| |
Collapse
|
140
|
Vela D, Leshoski J, Gjorgievska ES, Hadzi-Petrushev N, Jakupaj M, Sopi RB, Mladenov M. The Role of Insulin Therapy in Correcting Hepcidin Levels in Patients with Type 2 Diabetes Mellitus. Oman Med J 2017; 32:195-200. [PMID: 28584599 DOI: 10.5001/omj.2017.37] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
OBJECTIVES Iron overload can cause or contribute to the pathogenesis of type 2 diabetes mellitus (T2DM), but how the major parameters of iron metabolism change in different settings of diabetes are still unclear. The aim of this study was to determine the relationship between iron, ferritin, and hepcidin levels in diabetic patients and the effect of insulin treatment. METHODS The study included 80 subjects, 60 with T2DM and 20 without (control group). Serum hepcidin, insulin, ferritin, and iron levels were determined as well as other clinical parameters. The associations between these parameters were analyzed between both groups. RESULTS Hepcidin levels expressed as mean± standard deviation between groups showed no significant changes (14.4±6.7 ng/mL for the control group, and 18.4±7.9 ng/mL for patients with diabetes, p = 0.069). Parameters of iron metabolism showed modest correlation with the parameters of glucose metabolism. However, the correlation between ferritin and insulin in both groups was statistically significant (p = 0.032; ρ = 0.480 vs. p = 0.011; ρ = 0.328). CONCLUSIONS Our study showed that hepcidin levels in patients with T2DM on insulin therapy do not change, which might be a result of treatment with insulin. In this context, insulin treatment can be used as a novel method for correction of hepcidin levels. By correcting hepcidin levels, we can prevent cellular iron overload and reduce the risk of diabetes.
Collapse
Affiliation(s)
- Driton Vela
- Faculty of Medicine, University of Prishtina, Prishtina, Kosovo
| | - Jovica Leshoski
- Institute of Biology, Faculty of Natural Sciences, Saints Cyril and Methodius University of Skopje, Skopje, Macedonia
| | - Elizabeta S Gjorgievska
- Faculty of Dental Medicine, Saints Cyril and Methodius University of Skopje, Skopje, Macedonia
| | - Nikola Hadzi-Petrushev
- Institute of Biology, Faculty of Natural Sciences, Saints Cyril and Methodius University of Skopje, Skopje, Macedonia
| | | | - Ramadan B Sopi
- Faculty of Medicine, University of Prishtina, Prishtina, Kosovo
| | - Mitko Mladenov
- Institute of Biology, Faculty of Natural Sciences, Saints Cyril and Methodius University of Skopje, Skopje, Macedonia
| |
Collapse
|
141
|
González N, Prieto I, del Puerto-Nevado L, Portal-Nuñez S, Ardura JA, Corton M, Fernández-Fernández B, Aguilera O, Gomez-Guerrero C, Mas S, Moreno JA, Ruiz-Ortega M, Sanz AB, Sanchez-Niño MD, Rojo F, Vivanco F, Esbrit P, Ayuso C, Alvarez-Llamas G, Egido J, García-Foncillas J, Ortiz A. 2017 update on the relationship between diabetes and colorectal cancer: epidemiology, potential molecular mechanisms and therapeutic implications. Oncotarget 2017; 8:18456-18485. [PMID: 28060743 PMCID: PMC5392343 DOI: 10.18632/oncotarget.14472] [Citation(s) in RCA: 124] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2016] [Accepted: 12/26/2016] [Indexed: 02/06/2023] Open
Abstract
Worldwide deaths from diabetes mellitus (DM) and colorectal cancer increased by 90% and 57%, respectively, over the past 20 years. The risk of colorectal cancer was estimated to be 27% higher in patients with type 2 DM than in non-diabetic controls. However, there are potential confounders, information from lower income countries is scarce, across the globe there is no correlation between DM prevalence and colorectal cancer incidence and the association has evolved over time, suggesting the impact of additional environmental factors. The clinical relevance of these associations depends on understanding the mechanism involved. Although evidence is limited, insulin use has been associated with increased and metformin with decreased incidence of colorectal cancer. In addition, colorectal cancer shares some cellular and molecular pathways with diabetes target organ damage, exemplified by diabetic kidney disease. These include epithelial cell injury, activation of inflammation and Wnt/β-catenin pathways and iron homeostasis defects, among others. Indeed, some drugs have undergone clinical trials for both cancer and diabetic kidney disease. Genome-wide association studies have identified diabetes-associated genes (e.g. TCF7L2) that may also contribute to colorectal cancer. We review the epidemiological evidence, potential pathophysiological mechanisms and therapeutic implications of the association between DM and colorectal cancer. Further studies should clarify the worldwide association between DM and colorectal cancer, strengthen the biological plausibility of a cause-and-effect relationship through characterization of the molecular pathways involved, search for specific molecular signatures of colorectal cancer under diabetic conditions, and eventually explore DM-specific strategies to prevent or treat colorectal cancer.
Collapse
Affiliation(s)
- Nieves González
- Renal, Vascular and Diabetes Research Laboratory, IIS-Fundacion Jimenez Diaz-UAM, Spanish Biomedical Research Network in Diabetes and Associated Metabolic Disorders (CIBERDEM), Madrid, Spain
| | - Isabel Prieto
- Radiation Oncology, Oncohealth Institute, IIS-Fundacion Jimenez Diaz-UAM, Madrid, Spain
| | - Laura del Puerto-Nevado
- Translational Oncology Division, Oncohealth Institute, IIS-Fundacion Jimenez Diaz-UAM, Madrid, Spain
| | - Sergio Portal-Nuñez
- Bone and Mineral Metabolism laboratory, IIS-Fundacion Jimenez Diaz-UAM, Madrid, Spain
| | - Juan Antonio Ardura
- Bone and Mineral Metabolism laboratory, IIS-Fundacion Jimenez Diaz-UAM, Madrid, Spain
| | - Marta Corton
- Genetics, IIS-Fundacion Jimenez Diaz-UAM, Madrid, Spain
| | | | - Oscar Aguilera
- Translational Oncology Division, Oncohealth Institute, IIS-Fundacion Jimenez Diaz-UAM, Madrid, Spain
| | | | - Sebastián Mas
- Nephrology, IIS-Fundacion Jimenez Diaz-UAM, Madrid, Spain
| | | | | | - Ana Belen Sanz
- Nephrology, IIS-Fundacion Jimenez Diaz-UAM, Madrid, Spain
- REDINREN, Madrid, Spain
| | | | - Federico Rojo
- Pathology, IIS-Fundacion Jimenez Diaz-UAM, Madrid, Spain
| | | | - Pedro Esbrit
- Bone and Mineral Metabolism laboratory, IIS-Fundacion Jimenez Diaz-UAM, Madrid, Spain
| | - Carmen Ayuso
- Genetics, IIS-Fundacion Jimenez Diaz-UAM, Madrid, Spain
| | | | - Jesús Egido
- Renal, Vascular and Diabetes Research Laboratory, IIS-Fundacion Jimenez Diaz-UAM, Spanish Biomedical Research Network in Diabetes and Associated Metabolic Disorders (CIBERDEM), Madrid, Spain
- Nephrology, IIS-Fundacion Jimenez Diaz-UAM, Madrid, Spain
| | - Jesús García-Foncillas
- Translational Oncology Division, Oncohealth Institute, IIS-Fundacion Jimenez Diaz-UAM, Madrid, Spain
| | - Alberto Ortiz
- Nephrology, IIS-Fundacion Jimenez Diaz-UAM, Madrid, Spain
- REDINREN, Madrid, Spain
| | | |
Collapse
|
142
|
Vassalle C, Meloni A, Pistoia L, Pepe A. LIVER PANCREAS HEART TRIANGLE AND HCV IN THALASSEMIA: EXPANDING THE HORIZON THROUGH BIOMARKER NETWORKS. ACTA ACUST UNITED AC 2017. [DOI: 10.15436/2381-1404.17.017] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/09/2022]
|
143
|
Barton JC, Acton RT. Diabetes in HFE Hemochromatosis. J Diabetes Res 2017; 2017:9826930. [PMID: 28331855 PMCID: PMC5346371 DOI: 10.1155/2017/9826930] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/17/2016] [Revised: 12/12/2016] [Accepted: 01/04/2017] [Indexed: 02/08/2023] Open
Abstract
Diabetes in whites of European descent with hemochromatosis was first attributed to pancreatic siderosis. Later observations revealed that the pathogenesis of diabetes in HFE hemochromatosis is multifactorial and its clinical manifestations are heterogeneous. Increased type 2 diabetes risk in HFE hemochromatosis is associated with one or more factors, including abnormal iron homeostasis and iron overload, decreased insulin secretion, cirrhosis, diabetes in first-degree relatives, increased body mass index, insulin resistance, and metabolic syndrome. In p.C282Y homozygotes, serum ferritin, usually elevated at hemochromatosis diagnosis, largely reflects body iron stores but not diabetes risk. In persons with diabetes type 2 without hemochromatosis diagnoses, serum ferritin levels are higher than those of persons without diabetes, but most values are within the reference range. Phlebotomy therapy to achieve iron depletion does not improve diabetes control in all persons with HFE hemochromatosis. The prevalence of type 2 diabetes diagnosed today in whites of European descent with and without HFE hemochromatosis is similar. Routine iron phenotyping or HFE genotyping of patients with type 2 diabetes is not recommended. Herein, we review diabetes in HFE hemochromatosis and the role of iron in diabetes pathogenesis in whites of European descent with and without HFE hemochromatosis.
Collapse
Affiliation(s)
- James C. Barton
- Southern Iron Disorders Center, Birmingham, AL 35209, USA
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Ronald T. Acton
- Southern Iron Disorders Center, Birmingham, AL 35209, USA
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| |
Collapse
|
144
|
Rawal S, Hinkle SN, Bao W, Zhu Y, Grewal J, Albert PS, Weir NL, Tsai MY, Zhang C. A longitudinal study of iron status during pregnancy and the risk of gestational diabetes: findings from a prospective, multiracial cohort. Diabetologia 2017; 60:249-257. [PMID: 27830277 PMCID: PMC6331052 DOI: 10.1007/s00125-016-4149-3] [Citation(s) in RCA: 60] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/21/2016] [Accepted: 09/29/2016] [Indexed: 12/12/2022]
Abstract
AIMS/HYPOTHESIS The aim of this study was to prospectively and longitudinally investigate maternal iron status during early to mid-pregnancy, and subsequent risk of gestational diabetes mellitus (GDM), using a comprehensive panel of conventional and novel iron biomarkers. METHODS A case-control study of 107 women with GDM and 214 controls (matched on age, race/ethnicity and gestational week during blood collection) was conducted within the the Eunice Kennedy Shriver National Institute of Child Health and Human Development Fetal Growth Studies-Singleton Cohort (2009-2013), a prospective and multiracial pregnancy cohort. Plasma hepcidin, ferritin and soluble transferrin receptor (sTfR) were measured and sTfR:ferritin ratio was derived, twice before GDM diagnosis (gestational weeks 10-14 and 15-26) and at weeks 23-31 and 33-39. GDM diagnosis was ascertained from medical records. Adjusted ORs (aORs) for GDM were estimated using conditional logistic regression analysis, adjusting for demographics, prepregnancy BMI and other major risk factors. RESULTS Hepcidin concentrations during weeks 15-26 were 16% higher among women with GDM vs controls (median 6.4 vs 5.5 ng/ml; p = 0.02 ), and were positively associated with GDM risk; the aOR (95% CI) for highest vs lowest quartile was 2.61 (1.07, 6.36). Ferritin levels were also positively associated with GDM risk; the aOR (95% CI) for highest vs lowest quartile was 2.43 (1.12, 5.28) at weeks 10-14 and 3.95 (1.38, 11.30) at weeks 15-26. The sTfR:ferritin ratio was inversely related to GDM risk; the aOR (95% CI) for highest vs lowest quartile was 0.33 (0.14, 0.80) at weeks 10-14 and 0.15 (0.05, 0.48) at weeks 15-26. CONCLUSIONS/INTERPRETATION Our findings suggest that elevated iron stores may be involved in the development of GDM from as early as the first trimester. This raises potential concerns for the recommendation of routine iron supplementation among iron-replete pregnant women.
Collapse
Affiliation(s)
- Shristi Rawal
- Epidemiology Branch, Division of Intramural Population Health Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, 6710B Rockledge Drive, MSC 7004, Bethesda, MD, 20817, USA
| | - Stefanie N Hinkle
- Epidemiology Branch, Division of Intramural Population Health Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, 6710B Rockledge Drive, MSC 7004, Bethesda, MD, 20817, USA
| | - Wei Bao
- Department of Epidemiology, University of Iowa College of Public Health, Iowa City, IA, USA
| | - Yeyi Zhu
- Epidemiology Branch, Division of Intramural Population Health Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, 6710B Rockledge Drive, MSC 7004, Bethesda, MD, 20817, USA
| | - Jagteshwar Grewal
- Office of the Director, Division of Intramural Population Health Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, USA
| | - Paul S Albert
- Biostatistics and Bioinformatics Branch, Division of Intramural Population Health Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, USA
| | - Natalie L Weir
- Department of Laboratory Medicine and Pathology, University of Minnesota Medical School, Minneapolis, MN, USA
| | - Michael Y Tsai
- Department of Laboratory Medicine and Pathology, University of Minnesota Medical School, Minneapolis, MN, USA
| | - Cuilin Zhang
- Epidemiology Branch, Division of Intramural Population Health Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, 6710B Rockledge Drive, MSC 7004, Bethesda, MD, 20817, USA.
| |
Collapse
|
145
|
Lainé F, Ruivard M, Loustaud-Ratti V, Bonnet F, Calès P, Bardou-Jacquet E, Sacher-Huvelin S, Causse X, Beusnel C, Renault A, Bellissant E, Deugnier Y. Metabolic and hepatic effects of bloodletting in dysmetabolic iron overload syndrome: A randomized controlled study in 274 patients. Hepatology 2017; 65:465-474. [PMID: 27685251 DOI: 10.1002/hep.28856] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/26/2016] [Revised: 08/18/2016] [Accepted: 09/28/2016] [Indexed: 01/06/2023]
Abstract
UNLABELLED Dysmetabolic iron overload syndrome (DIOS) is a common cause of hyperferritinemia, accounting for a mild increase of iron stores in insulin-resistant subjects. Iron removal could improve insulin sensitivity. We performed a prospective, randomized, controlled trial (NCT01015525) in nondiabetic DIOS patients with hepatic iron >50 μmol/g at magnetic resonance imaging to compare the metabolic and hepatic outcomes of 1-year maintenance of serum ferritin levels <50 μg/L by bloodletting associated with lifestyle and diet advice (LFDA) to those of LFDA only. Patients were randomly assigned (1:1) with stratification by center (n = 8) and hyperglycemia (>5.6 mmol/L). Sample size was calculated to provide 90% power and a difference in fasting glycemia of 0.25 mmol/L. Analysis was done in an intention-to-treat population. In 2010-2014, 146 patients were randomly assigned to receive venesections with LFDA and 128 to LFDA only. At the end of the study, comparison of iron-depleted patients and controls showed ferritin levels 71 ± 48 μg/L after removal of 4.9 ± 1.6 L of blood versus 733 ± 277 μg/L (P < 0.0001), glycemia 5.44 ± 0.7 versus 5.49 ± 0.7 mmol/L (P = 0.57), body weight +0.5 ± 4.3% versus -0.6 ± 3.3% (P = 0.03), homeostasis model of assessment of insulin resistance 3.39 versus 2.40 (P = 0.002), alanine aminotransaminase 33 ± 22 versus 37 ± 21 IU/L (P = 0.10), aspartate aminotransaminase 27 ± 13 versus 27 ± 10 IU/L (P = 0.81), gamma-glutamyl transferase 54 ± 138 versus 49 ± 35 IU/L (P = 0.72), Fatty Liver Index 58.9 ± 24.6 versus 61.2 ± 22.9 (P = 0.37), and Fibrosis-4 score 1.5 ± 0.6 versus 1.30 ± 0.6 (P = 0.51). Fatigue occurred in 25.3% of venesected patients versus 2.3% of controls (P < 0.0001). In the subgroup of patients who lost weight, glycemia, homeostasis model of assessment of insulin resistance, serum ferritin, lipid profile, and liver function tests improved irrespective of bloodletting. CONCLUSION In DIOS patients, iron depletion by bloodletting does not improve metabolic and hepatic features, is associated with weight gain, and is not as well tolerated as expected; sustained modification of diet and lifestyle habits remains the first therapeutic intervention in DIOS. (Hepatology 2017;65:465-474).
Collapse
Affiliation(s)
- Fabrice Lainé
- INSERM CIC 1414, CHU Rennes, Rennes, France.,INSERM U 991, CHU Rennes, Rennes, France.,Liver Unit, CHU Rennes, Rennes, France
| | - Marc Ruivard
- Department of Internal Medicine, CHU Estaing, and Clermont University, Auvergne University, EA 4681, PEPRADE, Clermont-Ferrand, France
| | | | - Fabrice Bonnet
- INSERM CIC 1414, CHU Rennes, Rennes, France.,Department of Endocrinology-Diabetology, CHU Rennes, France.,University of Rennes 1, Rennes, France
| | - Paul Calès
- Liver-Gastroenterology Department, University Hospital, and UPRES 3859, LUNAM University, Angers, France
| | - Edouard Bardou-Jacquet
- INSERM CIC 1414, CHU Rennes, Rennes, France.,INSERM U 991, CHU Rennes, Rennes, France.,Liver Unit, CHU Rennes, Rennes, France.,University of Rennes 1, Rennes, France
| | | | - Xavier Causse
- Liver-Gastroenterology and Oncology Department, CHR Orléans, Orléans, France
| | | | - Alain Renault
- INSERM CIC 1414, CHU Rennes, Rennes, France.,University of Rennes 1, Rennes, France
| | - Eric Bellissant
- INSERM CIC 1414, CHU Rennes, Rennes, France.,University of Rennes 1, Rennes, France
| | - Yves Deugnier
- INSERM CIC 1414, CHU Rennes, Rennes, France.,University of Rennes 1, Rennes, France
| | | |
Collapse
|
146
|
Shi Q, Xu J, Huo N, Cai C, Liu H. Does a higher glycemic level lead to a higher rate of dental implant failure? J Am Dent Assoc 2016; 147:875-881. [DOI: 10.1016/j.adaj.2016.06.011] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2016] [Revised: 06/02/2016] [Accepted: 06/03/2016] [Indexed: 01/15/2023]
|
147
|
Stroh MA, Winter MK, Swerdlow RH, McCarson KE, Zhu H. Loss of NCB5OR in the cerebellum disturbs iron pathways, potentiates behavioral abnormalities, and exacerbates harmaline-induced tremor in mice. Metab Brain Dis 2016; 31:951-64. [PMID: 27188291 PMCID: PMC5929129 DOI: 10.1007/s11011-016-9834-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/25/2016] [Accepted: 05/01/2016] [Indexed: 12/13/2022]
Abstract
Iron dyshomeostasis has been implicated in many diseases, including a number of neurological conditions. Cytosolic NADH cytochrome b5 oxidoreductase (NCB5OR) is ubiquitously expressed in animal tissues and is capable of reducing ferric iron in vitro. We previously reported that global gene ablation of NCB5OR resulted in early-onset diabetes and altered iron homeostasis in mice. To further investigate the specific effects of NCB5OR deficiency on neural tissue without contributions from known phenotypes, we generated a conditional knockout (CKO) mouse that lacks NCB5OR only in the cerebellum and midbrain. Assessment of molecular markers in the cerebellum of CKO mice revealed changes in pathways associated with cellular and mitochondrial iron homeostasis. (59)Fe pulse-feeding experiments revealed cerebellum-specific increased or decreased uptake of iron by 7 and 16 weeks of age, respectively. Additionally, we characterized behavioral changes associated with loss of NCB5OR in the cerebellum and midbrain in the context of dietary iron deprivation-evoked generalized iron deficiency. Locomotor activity was reduced and complex motor task execution was altered in CKO mice treated with an iron deficient diet. A sucrose preference test revealed that the reward response was intact in CKO mice, but that iron deficient diet consumption altered sucrose preference in all mice. Detailed gait analysis revealed locomotor changes in CKO mice associated with dysfunctional proprioception and locomotor activation independent of dietary iron deficiency. Finally, we demonstrate that loss of NCB5OR in the cerebellum and midbrain exacerbated harmaline-induced tremor activity. Our findings suggest an essential role for NCB5OR in maintaining both iron homeostasis and the proper functioning of various locomotor pathways in the mouse cerebellum and midbrain.
Collapse
Affiliation(s)
- Matthew A Stroh
- Landon Center on Aging, University of Kansas Medical Center, 3901 Rainbow Blvd., MSN 1005, Kansas City, KS, 66160, USA
- Department of Biochemistry and Molecular Biology, University of Kansas Medical Center, 3901 Rainbow Blvd., MSN 3030, Kansas City, KS, 66160, USA
- Neuroscience Graduate Program, University of Kansas Medical Center, 3901 Rainbow Blvd., MSN 3038, Kansas City, KS, 66160, USA
| | - Michelle K Winter
- Kansas Intellectual and Developmental Disabilities Research Center, University of Kansas Medical Center, 3901 Rainbow Blvd., MSN 3051, Kansas City, KS, 66160, USA
| | - Russell H Swerdlow
- Department of Biochemistry and Molecular Biology, University of Kansas Medical Center, 3901 Rainbow Blvd., MSN 3030, Kansas City, KS, 66160, USA
- Neuroscience Graduate Program, University of Kansas Medical Center, 3901 Rainbow Blvd., MSN 3038, Kansas City, KS, 66160, USA
- Department of Neurology, University of Kansas Medical Center, 3599 Rainbow Blvd., MSN 2012, Kansas City, KS, 66160, USA
| | - Kenneth E McCarson
- Kansas Intellectual and Developmental Disabilities Research Center, University of Kansas Medical Center, 3901 Rainbow Blvd., MSN 3051, Kansas City, KS, 66160, USA
- Department of Pharmacology, Toxicology and Therapeutics, University of Kansas Medical Center, 3901 Rainbow Blvd., MSN 1018, Kansas City, KS, 66160, USA
| | - Hao Zhu
- Department of Biochemistry and Molecular Biology, University of Kansas Medical Center, 3901 Rainbow Blvd., MSN 3030, Kansas City, KS, 66160, USA.
- Neuroscience Graduate Program, University of Kansas Medical Center, 3901 Rainbow Blvd., MSN 3038, Kansas City, KS, 66160, USA.
- Department of Clinical Laboratory Sciences, University of Kansas Medical Center, 3901 Rainbow Blvd., MSN 4048G-Eaton, Kansas City, KS, 66160, USA.
| |
Collapse
|